Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Chin J Nat Med ; 22(5): 387-401, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38796213

RESUMO

Hernandezine (Her), a bisbenzylisoquinoline alkaloid extracted from Thalictrum flavum, is recognized for its range of biological activities inherent to this herbal medicine. Despite its notable properties, the anti-cancer effects of Her have remained largely unexplored. In this study, we elucidated that Her significantly induced cytotoxicity in cancer cells through the activation of apoptosis and necroptosis mechanisms. Furthermore, Her triggered autophagosome formation by activating the AMPK and ATG5 conjugation systems, leading to LC3 lipidation. Our findings revealed that Her caused damage to the mitochondrial membrane, with the damaged mitochondria undergoing mitophagy, as evidenced by the elevated expression of mitophagy markers. Conversely, Her disrupted autophagic flux, demonstrated by the upregulation of p62 and accumulation of autolysosomes, as observed in the RFP-GFP-LC3 reporter assay. Initially, we determined that Her did not prevent the fusion of autophagosomes and lysosomes. However, it inhibited the maturation of cathepsin D and increased lysosomal pH, indicating an impairment of lysosomal function. The use of the early-stage autophagy inhibitor, 3-methyladenine (3-MA), did not suppress LC3II, suggesting that Her also induces noncanonical autophagy in autophagosome formation. The application of Bafilomycin A1, an inhibitor of noncanonical autophagy, diminished the recruitment of ATG16L1 and the accumulation of LC3II by Her, thereby augmenting Her-induced cell death. These observations imply that while autophagy initially plays a protective role, the disruption of the autophagic process by Her promotes programmed cell death. This study provides the first evidence of Her's dual role in inducing apoptosis and necroptosis while also initiating and subsequently impairing autophagy to promote apoptotic cell death. These insights contribute to a deeper understanding of the mechanisms underlying programmed cell death, offering potential avenues for enhancing cancer prevention and therapeutic strategies.


Assuntos
Apoptose , Autofagia , Catepsina D , Lisossomos , Catepsina D/metabolismo , Catepsina D/genética , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Benzilisoquinolinas/farmacologia , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Concentração de Íons de Hidrogênio , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo
2.
Bioorg Med Chem Lett ; 108: 129803, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38777280

RESUMO

Targeted delivery of radionuclides to tumors is significant in theranostics applications for precision medicine. Pre-targeting, in which a tumor-targeting vehicle and a radionuclide-loaded effector small molecule are administered separately, holds promise since it can reduce unnecessary internal radiation exposure of healthy cells and can minimize radiation decay. The success of the pre-targeting delivery requires an in vivo-stable tumor-targeting vehicle selectively binding to tumor antigens and an in vivo-stable small molecule effector selectively binding to the vehicle accumulated on the tumor. We previously reported a drug delivery system composed of a low-immunogenic streptavidin with weakened affinity to endogenous biotin and a bis-iminobiotin with high affinity to the engineered streptavidin. It was, however, unknown whether the bis-iminobiotin is stable in vivo when administered alone for the pre-targeting applications. Here we report a new in vivo-stable bis-iminobiotin derivative. The keys to success were the identification of the degradation site of the original bis-iminobiotin treated with mouse plasma and the structural modification of the degradation site. We disclosed the successful pre-targeting delivery of astatine-211 (211At), α-particle emitter, to the CEACAM5-positive tumor in xenograft mouse models.


Assuntos
Biotina , Estreptavidina , Animais , Estreptavidina/química , Camundongos , Biotina/química , Humanos , Sistemas de Liberação de Medicamentos , Linhagem Celular Tumoral , Mutação , Estrutura Molecular
3.
Nucl Med Commun ; 42(9): 1052-1059, 2021 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-34001828

RESUMO

OBJECTIVE: This study aimed to evaluate the distribution of Astatine-211 (211At) solution dispersion in a small animal cage using autoradiography imaging to simulate the dispersion of 211At in a lab room to eventually prevent user's risk of internal exposure in terms of radiation safety. METHODS: 211At radiation sources with two chemical properties (Na211At and Free 211At) were prepared. The solutions of 211At were placed onto a dish with paper, and then, it was placed in a small animal cage for 3 h. After removing the dish, an imaging plate with attaching reference sources was placed at four walls of the cage for 15 h in a lead box. Imaging plates were read, and all pixel data were calculated using Microsoft Excel 2016 to obtain three-dimensional (3D) distribution. Calculated results were depicted using a 3D sphere model. RESULTS: The mean activity of Free 211At was 2.3 times higher than that of Na211At on all autoradiography images. In the cage, the shape of the dispersion of Na211At was almost homogeneous, whereas that of Free 211At was more heterogeneous. CONCLUSION: We found that the solution of 211At vaporized naturally and was distributed heterogeneously in the cage, and the chemical properties of 211At influenced their behaviors. These results must be considered to minimize the risks of radiation safety.


Assuntos
Astato , Radioimunoterapia , Autorradiografia , Humanos
4.
Sci Rep ; 11(1): 6668, 2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33758250

RESUMO

Developmental delay, epilepsy, and neonatal diabetes (DEND) syndrome, the most severe end of neonatal diabetes mellitus, is caused by mutation in the ATP-sensitive potassium (KATP) channel. In addition to diabetes, DEND patients present muscle weakness as one of the symptoms, and although the muscle weakness is considered to originate in the brain, the pathological effects of mutated KATP channels in skeletal muscle remain elusive. Here, we describe the local effects of the KATP channel on muscle by expressing the mutation present in the KATP channels of the DEND syndrome in the murine skeletal muscle cell line C2C12 in combination with computer simulation. The present study revealed that the DEND mutation can lead to a hyperpolarized state of the muscle cell membrane, and molecular dynamics simulations based on a recently reported high-resolution structure provide an explanation as to why the mutation reduces ATP sensitivity and reveal the changes in the local interactions between ATP molecules and the channel.


Assuntos
Diabetes Mellitus/genética , Epilepsia/genética , Doenças do Recém-Nascido/genética , Canais KATP/química , Canais KATP/genética , Músculo Esquelético/metabolismo , Mutação , Transtornos Psicomotores/genética , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Animais , Sítios de Ligação , Cálcio/metabolismo , Expressão Gênica , Glucose/metabolismo , Canais KATP/metabolismo , Potenciais da Membrana , Camundongos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Desenvolvimento Muscular , Fibras Musculares Esqueléticas , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
5.
Lung Cancer ; 153: 134-142, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33508526

RESUMO

OBJECTIVES: Delta-like 1 homolog (DLK1) is a non-canonical Notch ligand known to be expressed in several cancers but whose role in lung cancer is not yet fully understood. We sought to confirm DLK1 expression in small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC), and to examine DLK1's clinical significance. Furthermore, we examined the possible utility of DLK1 as a novel target in radioimmunotherapy (RIT). METHODS: We retrospectively assessed the correlation between clinical features and DLK1 expression by immunohistochemistry in resected specimens from 112 patients with SCLC and 101 patients with NSCLC. Moreover, we performed cell and animal experiments, and examined the possibility of RIT targeting DLK1 in SCLC using iodine-125 (125I) -labeled anti-DLK1 antibody, knowing that 125I can be replaced with the alpha-particle-emitter astatine-211 (211At). RESULTS: In SCLC and NSCLC, 20.5 % (23/112) and 16.8 % (17/101) of patients (respectively) had DLK1-positive tumors. In NSCLC, DLK1 expression was associated with recurrence-free survival (P < 0.01) but not with overall survival. In SCLC, there was no association between DLK1 expression and survival. In addition, 125I-labeled anti-DLK1 antibody specifically targeted DLK1 on human SCLC tumor cell lines. Furthermore, 125I-labeled anti-DLK1 antibody was incorporated into tumor tissue in a mouse model. CONCLUSION: A proportion of SCLC and NSCLC exhibits DLK1 expression. As a clinical feature, DLK1 expression could be a promising prognostic factor for recurrence in patients with resected NSCLC. In addition, DLK1 could serve as a new therapeutic target, including RIT, as suggested by our pilot study using a radiolabeled anti-DLK1 antibody in SCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Proteínas de Ligação ao Cálcio , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Humanos , Radioisótopos do Iodo , Neoplasias Pulmonares/radioterapia , Proteínas de Membrana/metabolismo , Recidiva Local de Neoplasia , Projetos Piloto , Radioimunoterapia , Estudos Retrospectivos
6.
EJNMMI Phys ; 7(1): 58, 2020 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-32960387

RESUMO

BACKGROUND: 211At is one of the ideal nuclides for targeted radionuclide therapies (TRTs). Meta-[211At]astatobenzylguanidine (211At-MABG) has been proposed for the treatment of pheochromocytoma. To effectively use these radiopharmaceuticals, dosimetry must be performed. It is important to determine the absorbed doses of free 211At and 211At-MABG to determine the organs that may be at risk when using TRTs. The aim of this study was to estimate human dosimetry from preclinical biodistribution of free 211At and 211At-MABG in various organs in normal mice. METHODS: Male C57BL/6 N mice were administered 0.13 MBq of free 211At or 0.20 MBq of 211At-MABG by tail-vein injection. The mice were sacrificed at 5 min, and at 1, 3, 6, and 24 h after the injection (n = 5 for each group). The percentage of injected activity per mass in organs and blood (%IA/g) was determined. The human absorbed doses of free 211At and 211At-MABG were calculated using the Organ Level INternal Dose Assessment/EXponential Modeling (OLINDA/EXM) version 2.0 and IDAC-Dose 2.1. RESULTS: High uptake of free 211At was observed in the lungs, spleen, salivary glands, stomach, and thyroid. The absorbed doses of free 211At in the thyroid and several tissues were higher than those of 211At-MABG. The absorbed doses of 211At-MABG in the adrenal glands, heart wall, and liver were higher than those of free 211At. CONCLUSIONS: The absorbed doses of 211At-MABG in organs expressing the norepinephrine transporter were higher than those of free 211At. In addition, the biodistribution of free 211At was different from that of 211At-MABG. The absorbed dose of free 211At may help predict the organs potentially at risk during TRTs using 211At-MABG due to deastatination.

7.
Chem Pharm Bull (Tokyo) ; 68(6): 538-545, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32475858

RESUMO

There are various diagnostic and therapeutic agents for prostate cancer using bombesin (BBN) derivatives, but astatine-211 (211At)-labeled BBN derivatives have yet to be studied. This study presented a preliminary evaluation of 211At-labeled BBN derivative. Several nonradioactive iodine-introduced BBN derivatives (IB-BBNs) with different linkers were synthesized and their binding affinities measured. Because IB-3 exhibited a comparable affinity to native BBN, [211At]AB-3 was synthesized and the radiochemical yields of [211At]AB-3 was 28.2 ± 2.4%, with a radiochemical purity of >90%. The stability studies and cell internalization/externalization experiments were performed. [211At]AB-3 was taken up by cells and internalized; however, radioactivity effluxed from cells over time. In addition, the biodistribution of [211At]AB-3, with and without excess amounts of BBN, were evaluated in PC-3 tumor-bearing mice. Despite poor stability in murine plasma, [211At]AB-3 accumulated in tumor tissue (4.05 ± 0.73%ID/g) in PC-3 tumor-bearing mice, which was inhibited by excess native BBN (2.56 ± 0.24%ID/g). Accumulated radioactivity in various organs is probably due to free 211At. Peptide degradation in murine plasma and radioactivity efflux from cells are areas of improvement. The development of 211At-labeled BBN derivatives requires modifying the BBN sequence and preventing deastatination.


Assuntos
Antineoplásicos/farmacologia , Astato/química , Bombesina/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Compostos Radiofarmacêuticos/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Bombesina/análogos & derivados , Bombesina/síntese química , Bombesina/química , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia , Células PC-3 , Neoplasias da Próstata/patologia , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/química , Distribuição Tecidual , Células Tumorais Cultivadas
8.
Sci Rep ; 10(1): 6810, 2020 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-32321944

RESUMO

To explore stem-cell-targeted radioimmunotherapy with α-particles in acute myelogenous leukemia (AML), pharmacokinetics and dosimetry of the 211At-labeled anti-C-X-C chemokine receptor type 4 monoclonal antibody (211At-CXCR4 mAb) were conducted using tumor xenografted mice. The biological half-life of 211At-CXCR4 mAb in blood was 15.0 h. The highest tumor uptake of 5.05%ID/g with the highest tumor-to-muscle ratio of 8.51 ± 6.14 was obtained at 6 h. Radiation dosimetry estimated with a human phantom showed absorbed doses of 0.512 mGy/MBq in the bone marrow, 0.287 mGy/MBq in the kidney, and <1 mGy/MBq in other major organs except bone. Sphere model analysis revealed 22.8 mGy/MBq in a tumor of 10 g; in this case, the tumor-to-bone marrow and tumor-to-kidney ratios were 44.5 and 79.4, respectively. The stem-cell-targeted α-particle therapy using 211At-CXCR4 mAb for AML appears possible and requires further therapeutic studies.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Astato/uso terapêutico , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/terapia , Células-Tronco Neoplásicas/patologia , Radioimunoterapia , Receptores CXCR4/imunologia , Animais , Humanos , Radioisótopos do Iodo , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Músculos/patologia , Especificidade de Órgãos , Doses de Radiação , Distribuição Tecidual , Células U937 , Ensaios Antitumorais Modelo de Xenoenxerto
9.
EJNMMI Res ; 9(1): 51, 2019 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-31161539

RESUMO

BACKGROUND: Eribulin, an inhibitor of microtubule dynamics, shows antitumor potency against a variety of solid cancers through its antivascular activity and remodeling of tumor vasculature. 18F-Fluoromisonidazole (18F-FMISO) is the most widely used PET probe for imaging tumor hypoxia. In this study, we utilized 18F-FMISO to clarify the effects of eribulin on the tumor hypoxic condition in comparison with histological findings. MATERIAL AND METHODS: Mice bearing a human cancer cell xenograft were intraperitoneally administered a single dose of eribulin (0.3 or 1.0 mg/kg) or saline. Three days after the treatment, mice were injected with 18F-FMISO and pimonidazole (hypoxia marker for immunohistochemistry), and intertumoral 18F-FMISO accumulation levels and histological characteristics were determined. PET/CT was performed pre- and post-treatment with eribulin (0.3 mg/kg, i.p.). RESULTS: The 18F-FMISO accumulation levels and percent pimonidazole-positive hypoxic area were significantly lower, whereas the number of microvessels was higher in the tumors treated with eribulin. The PET/CT confirmed that 18F-FMISO distribution in the tumor was decreased after the eribulin treatment. CONCLUSIONS: Using 18F-FMISO, we demonstrated the elimination of the tumor hypoxic condition by eribulin treatment, concomitantly with the increase in microvessel density. These findings indicate that PET imaging using 18F-FMISO may provide the possibility to detect the early treatment response in clinical patients undergoing eribulin treatment.

10.
Nucl Med Biol ; 70: 39-45, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30836255

RESUMO

INTRODUCTION: Hypoxia, a common feature of most solid tumors, is an important predictor of tumor progression and resistance to radiotherapy. We developed a novel hypoxia imaging probe with optimal biological characteristics for use in clinical settings. METHODS: We designed and synthesized several new hypoxia probes with additional hydrophilic characteristics compared to [18F]fluoromisonidazole ([18F]FMISO). These were 1-(2,2-Dihydroxy-methyl-3-[18F]-Fluoropropyl) azomycin ([18F]DiFA, formerly [18F]HIC101) and its analogs ([18F]F1 and [18F]F2). Biodistribution studies with EMT6 mammary carcinoma cell-bearing mice were performed 1 and 2 h after injection of each probe. Small-animal positron emission tomography (PET) imaging studies were conducted using [18F]DiFA and [18F]FMISO in the same mice. Tumoral hypoxia was confirmed via pimonidazole staining. Ex vivo digital autoradiographs were obtained for confirming the co-localization of [18F]DiFA and pimonidazole in the tumor tissues. RESULTS: The EMT6 tumors used had pimonidazole-positive regions. In biodistribution studies, the tumor-to-blood ratio and tumor-to-muscle ratio of [18F]DiFA was significantly higher than the respective [18F]FMISO ratios 1 h after injection. Hence, we selected [18F]DiFA as the best hypoxia probe among those tested. Small-animal PET imaging studies showed time-dependent increases in the tumor-to-normal tissue ratio of [18F]DiFA uptake. Rapid clearance from the rest of the body was observed primarily via the renal system. Ex vivo autoradiography showed a positive correlation between [18F]DiFA uptake and the regions of pimonidazole distribution, indicating that [18F]DiFA selectively accumulated in the tumor tissue's hypoxic region. CONCLUSIONS: A better contrast image and a shorter waiting time may be obtained with [18F]DiFA than with [18F]FMISO. ADVANCES IN KNOWLEDGE: By optimizing LogP based on the [18F]FMISO structure, we demonstrated that [18F]DiFA could detect tumor hypoxia regions at an early time point. IMPLICATIONS FOR PATIENT CARE: [18F]DiFA imaging facilitates the evaluation of various cancer hypoxic states due to the lower uptake of normal tissues and could contribute to novel treatment development.


Assuntos
Radioisótopos de Flúor/química , Radioisótopos de Flúor/farmacocinética , Misonidazol/análogos & derivados , Tomografia por Emissão de Pósitrons/normas , Hipóxia Tumoral , Animais , Autorradiografia , Linhagem Celular Tumoral , Camundongos , Nitroimidazóis/química , Nitroimidazóis/farmacocinética , Radioquímica , Padrões de Referência , Distribuição Tecidual
11.
Mol Imaging Biol ; 21(1): 122-129, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29845425

RESUMO

PURPOSE: Hypoxia in tumor has close relationship with angiogenesis and tumor progression. Previously, we developed 2,2-dihydroxymethyl-3-[18F]fluoropropyl-2-nitroimidazole ([18F]DiFA) as a novel positron emission tomography (PET) probe for diagnosis of hypoxia. In this study, we elucidated whether the accumulation of [18F]DiFA in cells is dependent on the hypoxic state and revealed how [18F]DiFA accumulates in hypoxic cells in combination with imaging mass spectrometry (IMS). PROCEDURES: FaDu human head and neck cancer cells were treated with [18F]DiFA and then incubated under normoxia (21% O2) or hypoxia (1% O2) for 2 h. The cells were extracted using methanol, and the radioactivities of the precipitates (macromolecule fraction) and supernatants (low-molecular-weight fraction) were measured. FaDu-bearing mice were injected intravenously with [18F]DiFA and with pimonidazole 1 h later. The tumors were excised 2 h after the injection of [18F]DiFA. Autoradiography, IMS, and immunohistochemical (IHC) staining for pimonidazole were performed with serial tumor sections. RESULTS: In the in vitro study, the radioactivity of FaDu cells was significantly higher under hypoxia than that under normoxia (0.53 ± 0.02 vs. 0.27 ± 0.02 %dose/mg protein, p < 0.05). The radioactivity of the low-molecular-weight fraction was 66.3 ± 0.6% in the hypoxic cell. In the in vivo study, [18F]DiFA accumulated in the tumor tissues existed mainly as low-molecular-weight compounds (90.4 ± 0.9%). In addition, the glutathione conjugate of reductive DiFA metabolite (amino-DiFA-GS) existed in tumor tissues revealed by the IMS study, and the distribution pattern of amino-DiFA-GS was very similar to that of the radioactivity and the positive staining area of pimonidazole. CONCLUSIONS: Our results suggest that [18F]DiFA undergoes the glutathione conjugation reaction following reductive metabolism in hypoxic cells, which leads hypoxia-specific PET imaging with [18F]DiFA.


Assuntos
Radioisótopos de Flúor , Glutationa/metabolismo , Hipóxia/metabolismo , Nitroimidazóis/farmacocinética , Tomografia por Emissão de Pósitrons/métodos , Hipóxia Tumoral , Animais , Neoplasias de Cabeça e Pescoço/diagnóstico , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Xenoenxertos , Humanos , Hipóxia/diagnóstico , Hipóxia/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nitroimidazóis/química , Oxirredução , Estresse Oxidativo/fisiologia , Distribuição Tecidual , Células Tumorais Cultivadas , Hipóxia Tumoral/efeitos dos fármacos
12.
Oncol Lett ; 14(2): 2341-2346, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28781672

RESUMO

A mechanistic dissociation exists between tumor starvation and vascular normalization after antiangiogenic therapy. Thus, improved understanding of tumor responses (tumor starvation or vascular normalization) is important for optimizing treatment strategies. 18F-fluoromisonidazole (18F-FMISO) is widely used for imaging tumor hypoxia. To clarify the tumor response to the antiangiogenic drug sorafenib, the present study evaluated the changes in the tumor oxygen state using 18F-FMISO in mice bearing a renal cell carcinoma xenograft (A498). Mice bearing A498 xenografts were assigned to the control and three sorafenib-treatment groups and administered sorafenib (0, 10, 20 or 40 mg/kg/day, per os) once daily for 3 days. Following one day after the final administration, the mice were injected with 18F-FMISO and pimonidazole (a hypoxia marker). 18F-FMISO accumulation in the tumor was determined by autoradiography. Immunohistochemistry of pimonidazole and cluster of differentiation (CD)31 (a vascular marker) was also performed. 18F-FMISO accumulation levels in the tumor significantly increased by 4.3-, 8.4- and 8.6-fold compared with in the control group following 10, 20 and 40 mg/kg sorafenib treatments, respectively [0.07±0.04, 0.32±0.11, 0.62±0.15 and 0.63±0.23 (%ID/m2) × kg for the control, and 10, 20 and 40 mg treatments, respectively; all P<0.0083 vs. the control]. The number of pimonidazole-positive cells also significantly increased by 6.8-, 12.3- and 20.2-fold compared with in the control group following 10, 20 and 40 mg/kg sorafenib treatments, respectively (0.78±0.79, 5.36±2.29, 9.66±1.58 and 15.85±4.59% pimonidazole-positive cells; all P<0.0083 vs. the control). The number of microvessels in tumors markedly decreased to 33.5, 17.6, and 14.0% of the control following 10, 20 and 40 mg/kg sorafenib treatments, respectively (17.1±2.5, 5.7±1.0, 3.0±1.0 and 2.4±0.3 vessels/mm2; P<0.0083 vs. the control). The 18F-FMISO expression level in the tumor increased sorafenib-dose-dependently, which is consistent with the increase in the number of pimonidazole-positive cells and decrease in the number of microvessels. These findings indicated that the present sorafenib treatment protocol induces 'tumor hypoxia/starvation' in the renal cell carcinoma xenograft (A498) due to its antiangiogenic properties.

13.
Nucl Med Biol ; 54: 18-26, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28821003

RESUMO

INTRODUCTION: 111In-DTPA-d-Phe1-octreotide scintigraphy is an important method of detecting neuroendocrine tumors. We previously reported that a new derivative of 111In-DTPA-d-Phe1-octreotide, 111In-DTPA-d-Phe-1-Asp0-d-Phe1-octreotide, accomplished the reduction of prolonged renal accumulation of radioactivity. The aim of this study was to evaluate the tumor accumulation of 111In-DTPA-d-Phe-1-Asp0-d-Phe1-octreotide in vitro and in vivo by comparing it with 111In-DTPA-d-Phe1-octreotide. METHODS: The tumor accumulation of this octreotide derivative was determined by measuring its uptake using cultured AR42J cells in vitro and biodistribution studies in vivo. The distribution of the radiotracer and the extent of somatostatin receptor-specific uptake in the tumor were estimated by a counting method using AR42J tumor-bearing mice. The radioactive metabolite species in the tumor and kidney were identified by HPLC analyses at 3 and 24h post-injection of the 111In-DTPA-conjugated peptide. RESULTS: In both cases, in vitro and in vivo, the tumor radioactivity levels of 111In-DTPA-d-Phe-1-Asp0-d-Phe1-octreotide were approximately 2-4 times higher than those of 111In-DTPA-d-Phe1-octreotide. On in vitro cellular uptake inhibition and radioreceptor assay, 111In-DTPA-d-Phe-1-Asp0-d-Phe1-octreotide exhibited a binding affinity to somatostatin receptor highly similar to that of 111In-DTPA-d-Phe1-octreotide. As the additional cellular uptake of 111In-DTPA-d-Phe-1-Asp0-d-Phe1-octreotide was significantly lower at low temperature than at 37°C, it was considered that a cellular uptake pathway is involved in energy-dependent endocytotic processes. In the radiometabolite analysis of 111In-DTPA-d-Phe-1-Asp0-d-Phe1-octreotide, 111In-DTPA-d-Phe-Asp-OH was a major metabolite in the tumor at 24h post-injection. CONCLUSION: 111In-DTPA-d-Phe-1-Asp0-d-Phe1-octreotide exhibited higher tumor accumulation and persistence of tumor radioactivity than 111In-DTPA-d-Phe1-octreotide. We reasoned that this higher tumor accumulation would not be based on the receptor affinity but on a receptor-mediated endocytotic process involved in temperature-dependent cellular uptake. The present study demonstrated the great potential of the pharmaceutical development of a new radiolabeled peptide with high tumor accumulation and low renal radioactivity by the chemical modification of 111In-DTPA-d-Phe1-octreotide.


Assuntos
Ácido Aspártico/química , Radioisótopos de Índio , Rim/metabolismo , Octreotida/química , Octreotida/metabolismo , Ácido Pentético/química , Fenilalanina/química , Animais , Transporte Biológico , Linhagem Celular Tumoral , Rim/diagnóstico por imagem , Camundongos , Octreotida/farmacocinética , Ratos , Receptores de Somatostatina/metabolismo , Distribuição Tecidual
14.
Ann Nucl Med ; 31(8): 596-604, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28695498

RESUMO

OBJECTIVE: 18F-fluoromisonidazole (FMISO), a well-known PET imaging probe for diagnosis of hypoxia, is believed to accumulate in hypoxic cells via covalent binding with macromolecules after reduction of the nitro group. Previously, we showed the majority of 18F-FMISO was incorporated into low-molecular-weight metabolites in hypoxic tumors, and the glutathione conjugate of reduced FMISO (amino-FMISO-GS) distributed in the tumor hypoxic regions as revealed by imaging mass spectrometry (IMS). The present study was conducted to clarify whether FMISO is metabolized to amino-FMISO-GS within tumor cells and how amino-FMISO-GS contributes to FMISO accumulation in hypoxic cells. We also evaluated the relationship between FMISO accumulation and the glutathione conjugation-related factors in the cells. METHODS: Tumor cells (FaDu, LOVO, and T24) were treated with 18F-FMISO and incubated under normoxic or hypoxic conditions for 4 h. The FMISO metabolites were analyzed with LC-ESI-MS. Several glutathione conjugation-related factors of tumor cells were evaluated in vitro. FaDu tumor-bearing mice were intravenously injected with 18F-FMISO and the tumors were excised at 4 h post-injection. Autoradiography, IMS and histologic studies were performed. RESULTS: Amino-FMISO-GS was the main contributor to FMISO incorporated in hypoxic FaDu cells in vitro and in vivo. Total FMISO uptake levels and amino-FMISO-GS levels were highest in FaDu, followed by LOVO, and then T24 (total uptake: 0.851 ± 0.009 (FaDu), 0.617 ± 0.021 (LOVO) and 0.167 ± 0.006 (T24) % dose/mg protein; amino-FMISO-GS: 0.502 ± 0.035 (FaDu), 0.158 ± 0.013 (LOVO), and 0.007 ± 0.001 (T24) % dose/mg protein). The glutathione level of FaDu was significantly higher than those of LOVO and T24. The enzyme activity of glutathione-S-transferase catalyzing the glutathione conjugation reaction in FaDu was similar levels to that in LOVO, and was higher than that in T24. Quantitative RT-PCR analysis revealed that the expression levels of efflux transporters of the glutathione conjugate (multidrug resistance-associated protein 1) were lowest in FaDu, followed by LOVO, and then T24. CONCLUSIONS: FMISO accumulates in hypoxic cells through reductive metabolism followed by glutathione conjugation. We illustrated the possibility that increased production and decreased excretion of amino-FMISO-GS contribute to FMISO accumulation in tumor cells under hypoxic conditions.


Assuntos
Glutationa/metabolismo , Misonidazol/análogos & derivados , Neoplasias Experimentais/metabolismo , Oxigênio/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Hipóxia Tumoral , Linhagem Celular Tumoral , Humanos , Taxa de Depuração Metabólica , Misonidazol/farmacocinética , Neoplasias Experimentais/diagnóstico por imagem , Ligação Proteica , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual
15.
PLoS One ; 12(4): e0175976, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28410399

RESUMO

Diabetes mellitus accelerates atherosclerosis that causes most cardiovascular events. Several metabolic pathways are considered to contribute to the development of atherosclerosis, but comprehensive metabolic alterations to atherosclerotic arterial cells remain unknown. The present study investigated metabolic changes and their relationship to vascular histopathological changes in the atherosclerotic arteries of rabbits with alloxan-induced diabetes. Diabetic atherosclerosis was induced in rabbit ilio-femoral arteries by injecting alloxan (100 mg/kg), injuring the arteries using a balloon, and feeding with a 0.5% cholesterol diet. We histologically assessed the atherosclerotic lesion development, cellular content, pimonidazole positive-hypoxic area, the nuclear localization of hypoxia-inducible factor-1α, and apoptosis. We evaluated comprehensive arterial metabolism by performing metabolomic analyses using capillary electrophoresis-time of flight mass spectrometry. We evaluated glucose uptake and its relationship to vascular hypoxia using 18F-fluorodeoxyglucose and pimonidazole. Plaque burden, macrophage content, and hypoxic areas were more prevalent in arteries with diabetic, than non-diabetic atherosclerosis. Metabolomic analyses highlighted 12 metabolites that were significantly altered between diabetic and non-diabetic atherosclerosis. A half of them were associated with glycolysis metabolites, and their levels were decreased in diabetic atherosclerosis. The uptake of glucose evaluated as 18F-fluorodeoxyglucose in atherosclerotic lesions increased according to increased macrophage content or hypoxic areas in non-diabetic, but not diabetic rabbits. Despite profound hypoxic areas, the nuclear localization of hypoxia-inducible factor-1α decreased and the number of apoptotic cells increased in diabetic atherosclerotic lesions. Altered glycolysis metabolism and an impaired response to hypoxia in atherosclerotic lesions under conditions of insulin-dependent diabetes might be involved in the development of diabetic atherosclerosis.


Assuntos
Aloxano/toxicidade , Aterosclerose/patologia , Diabetes Mellitus Experimental/induzido quimicamente , Glucose/metabolismo , Hipóxia , Animais , Apoptose , Aterosclerose/diagnóstico por imagem , Aterosclerose/etiologia , Autorradiografia , Peso Corporal , Análise por Conglomerados , Diabetes Mellitus Experimental/complicações , Dieta Hiperlipídica , Eletroforese Capilar , Artéria Femoral/diagnóstico por imagem , Artéria Femoral/patologia , Fluordesoxiglucose F18/química , Fluordesoxiglucose F18/metabolismo , Glicólise , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imuno-Histoquímica , Macrófagos/metabolismo , Macrófagos/patologia , Espectrometria de Massas , Metabolômica , Microscopia de Fluorescência , Nitroimidazóis/química , Nitroimidazóis/metabolismo , Análise de Componente Principal , Coelhos
16.
Nucl Med Biol ; 48: 16-25, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28160666

RESUMO

INTRODUCTION: Radiolabeled octreotide derivatives have been studied as diagnostic and therapeutic agents for somatostatin receptor-positive tumors. To prevent unnecessary radiation exposure during their clinical application, the present study aimed to develop radiolabeled peptides which could reduce radioactivity levels in the kidney at both early and late post-injection time points by introducing a negative charge with an acidic amino acid such as L-aspartic acid (Asp) at a suitable position in 111In-DTPA-conjugated octreotide derivatives. METHODS: Biodistribution of the radioactivity was evaluated in normal mice after administration of a novel radiolabeled peptide by a counting method. The radiolabeled species remaining in the kidney were identified by comparing their HPLC data with those obtained by alternative synthesis. RESULTS: The designed and synthesized radiolabeled peptide 111In-DTPA-d-Phe-1-Asp0-d-Phe1-octreotide exhibited significantly lower renal radioactivity levels than those of the known 111In-DTPA-d-Phe1-octreotide at 3 and 24h post-injection. The radiolabeled species in the kidney at 24h after the injection of new octreotide derivative represented 111In-DTPA-d-Phe-OH and 111In-DTPA-d-Phe-Asp-OH as the metabolites. Their radiometabolites and intact 111In-DTPA-conjugated octreotide derivative were observed in urine within 24h post-injection. CONCLUSION: The present study provided a new example of an 111In-DTPA-conjugated octreotide derivative having the characteristics of both reduced renal uptake and shortened residence time of radioactivity in the kidney. It is considered that this kinetic control was achieved by introducing a negative charge on the octreotide derivative thereby suppressing the reabsorption in the renal tubules and affording the radiometabolites with appropriate lipophilicity.


Assuntos
Desenho de Fármacos , Rim/efeitos da radiação , Octreotida/análogos & derivados , Ácido Pentético/análogos & derivados , Animais , Estabilidade de Medicamentos , Marcação por Isótopo , Rim/metabolismo , Camundongos , Octreotida/química , Octreotida/farmacocinética , Octreotida/urina , Ácido Pentético/química , Ácido Pentético/farmacocinética , Ácido Pentético/urina , Radioatividade , Distribuição Tecidual
17.
Mol Imaging Biol ; 19(4): 531-539, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27981470

RESUMO

PURPOSE: Vulnerable plaques are key factors for ischemic diseases. Thus, their precise detection is necessary for the diagnosis of such diseases. Immunoglobulin G (IgG)-based imaging probes have been developed for imaging biomolecules related to plaque formation for the diagnosis of atherosclerosis. However, IgG accumulates nonspecifically in atherosclerotic regions, and its accumulation mechanisms have not yet been clarified in detail. Therefore, we explored IgG accumulation mechanisms in atherosclerotic lesions and examined images of radiolabeled IgG for the diagnosis of atherosclerosis. PROCEDURES: Mouse IgG without specificity to biomolecules was labeled with technetium-99m via 6-hydrazinonicotinate to yield [99mTc]IgG. ApoE-/- or C57BL/6J mice were injected intravenously with [99mTc]IgG, and their aortas were excised 24 h after injection. After radioactivity measurement, serial aortic sections were autoradiographically and histopathologically examined. RAW264.7 macrophages were polarized into M1 or M2 and then treated with [99mTc]IgG. The radioactivities in the cells were measured after 1 h of incubation. [99mTc]IgG uptake in M1 macrophages was also evaluated after the pretreatment with an anti-Fcγ receptor (FcγR) antibody. The expression levels of FcγRs in the cells were measured by western blot analysis. RESULTS: [99mTc]IgG accumulation levels in the aortas were significantly higher in apoE-/- mice than in C57BL/6J mice (5.1 ± 1.4 vs 2.8 ± 0.5 %ID/g, p < 0.05). Autoradiographic images showed that the accumulation areas highly correlated with the macrophage-infiltrated areas. M1 macrophages showed significantly higher levels of [99mTc]IgG than M2 or M0 (nonpolarized) macrophages [2.2 ± 0.3 (M1) vs 0.5 ± 0.1 (M2), 0.4 ± 0.1 (M0) %dose/mg protein, p < 0.01] and higher expression levels of FcγRI and FcγRII. [99mTc]IgG accumulation in M1 macrophages was suppressed by pretreatment with the anti-FcγR antibody [2.2 ± 0.3 (nonpretreatment) vs 1.2 ± 0.2 (pretreatment) %ID/mg protein, p < 0.01]. CONCLUSIONS: IgG accumulated in pro-inflammatory M1 macrophages via FcγRs in atherosclerotic lesions. Thus, the target biomolecule-independent imaging of active inflammation should be taken into account in the diagnosis of atherosclerosis using IgG-based probes.


Assuntos
Imunoglobulina G/metabolismo , Macrófagos/metabolismo , Imagem Molecular/métodos , Sondas Moleculares/metabolismo , Placa Aterosclerótica/patologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Autorradiografia , Polaridade Celular , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/metabolismo , Células RAW 264.7 , Receptores de IgG/metabolismo , Tecnécio/metabolismo , Distribuição Tecidual
18.
EJNMMI Res ; 6(1): 90, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27957722

RESUMO

BACKGROUND: Sorafenib, an oral multikinase inhibitor, has anti-proliferative and anti-angiogenic activities and is therapeutically effective against renal cell carcinoma (RCC). Recently, we have evaluated the tumor responses to sorafenib treatment in a RCC xenograft using [Methyl-3H(N)]-3'-fluoro-3'-deoxythythymidine ([3H]FLT). Contrary to our expectation, the FLT level in the tumor significantly increased after the treatment. In this study, to clarify the reason for the elevated FLT level, dynamic 3'-[18F]fluoro-3'-deoxythymidine ([18F]FLT) positron emission tomography (PET) and kinetic studies were performed in mice bearing a RCC xenograft (A498). The A498 xenograft was established in nude mice, and the mice were assigned to the control (n = 5) and treatment (n = 5) groups. The mice in the treatment group were orally given sorafenib (20 mg/kg/day p.o.) once daily for 3 days. Twenty-four hours after the treatment, dynamic [18F]FLT PET was performed by small-animal PET. Three-dimensional regions of interest (ROIs) were manually defined for the tumors. A three-compartment model fitting was carried out to estimate four rate constants using the time activity curve (TAC) in the tumor and the blood clearance rate of [18F]FLT. RESULTS: The dynamic pattern of [18F]FLT levels in the tumor significantly changed after the treatment. The rate constant of [18F]FLT phosphorylation (k3) was significantly higher in the treatment group (0.111 ± 0.027 [1/min]) than in the control group (0.082 ± 0.009 [1/min]). No significant changes were observed in the distribution volume, the ratio of [18F]FLT forward transport (K1) to reverse transport (k2), between the two groups (0.556 ± 0.073 and 0.641 ± 0.052 [mL/g] in the control group). CONCLUSIONS: Our dynamic PET studies indicated that the increase in FLT level may be caused by the phosphorylation of FLT in the tumor after the sorafenib treatment in the mice bearing a RCC xenograft. Dynamic PET studies with kinetic modeling could provide improved understanding of the biochemical processes involved in tumor responses to therapy.

19.
PLoS One ; 11(8): e0161639, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27580239

RESUMO

Hypoxia, or low oxygen concentration, is a key factor promoting tumor progression and angiogenesis and resistance of cancer to radiotherapy and chemotherapy. 2-Nitroimidazole-based agents have been widely used in pathological and nuclear medicine examinations to detect hypoxic regions in tumors; in particular, pimonidazole is used for histochemical staining of hypoxic regions. It is considered to accumulate in hypoxic cells via covalent binding with macromolecules or by forming reductive metabolites after reduction of its nitro group. However, the detailed mechanism of its accumulation remains unknown. In this study, we investigated the accumulation mechanism of pimonidazole in hypoxic tumor tissues in a mouse model by mass spectrometric analyses including imaging mass spectrometry (IMS). Pimonidazole and its reductive metabolites were observed in the tumor tissues. However, their locations in the tumor sections were not similar to the positively stained areas in pimonidazole-immunohistochemistry, an area considered hypoxic. The glutathione conjugate of reduced pimonidazole, a low-molecular-weight metabolite of pimonidazole, was found in tumor tissues by LC-MS analysis, and our IMS study determined that the intratumor localization of the glutathione conjugate was consistent with the area positively immunostained for pimonidazole. We also found complementary localization of the glutathione conjugate and reduced glutathione (GSH), implying that formation of the glutathione conjugate occurred in the tumor tissue. These results suggest that in hypoxic tumor cells, pimonidazole is reduced at its nitro group, followed by conjugation with GSH.


Assuntos
Glutationa/metabolismo , Espectrometria de Massas , Neoplasias Experimentais , Nitroimidazóis , Animais , Hipóxia Celular/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Nitroimidazóis/farmacocinética , Nitroimidazóis/farmacologia
20.
J Nucl Med ; 57(8): 1276-81, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27056620

RESUMO

UNLABELLED: Recently, companion diagnostics with nuclear medicine techniques have been anticipated as more suitable means than biopsy for predicting treatment efficacy. The anticancer effect of capecitabine, an orally administered chemotherapeutic agent activated by thymidine phosphorylase (TP), is positively associated with tumor TP expression levels. This study aimed to assess whether TP imaging using a radiolabeled uracil derivative, (123)I-5-iodo-6-[(2-iminoimidazolidinyl)methyl]uracil ((123)I-IIMU), could predict the efficacy of capecitabine treatment. METHODS: Sensitivity to doxifluridine, a metabolite of capecitabine and direct substrate for TP, was assessed by water-soluble tetrazolium salt assays in vitro for 3 human colon cancer cell lines with different TP expression profiles. The intracellular uptake and retention of (123)I-IIMU were evaluated. Mice inoculated with each cell line were treated with capecitabine for 2 wk, and tumor growth was compared. In vivo distribution studies and SPECT/CT imaging of (123)I-IIMU were performed in inoculated mice. RESULTS: In vitro experiments showed a positive relation between TP expression levels and doxifluridine sensitivity. In vitro studies revealed that intracellular uptake and retention of (123)I-IIMU were dependent on TP expression levels. In vivo experiments in inoculated mice showed that (123)I-IIMU accumulation in tumor tissue was in line with TP expression levels and susceptibility to capecitabine treatment. Moreover, SPECT/CT imaging of (123)I-IIMU in tumor-inoculated mice showed that (123)I-IIMU reflects TP expression levels in tumor tissues. CONCLUSION: (123)I-IIMU could be used as an in vivo companion diagnostic for predicting the efficacy of capecitabine treatment.


Assuntos
5'-Nucleotidase/metabolismo , Capecitabina/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/enzimologia , Monitoramento de Medicamentos/métodos , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único/métodos , Animais , Capecitabina/farmacocinética , Linhagem Celular Tumoral , Neoplasias do Colo/diagnóstico por imagem , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Imagem Molecular/métodos , Prognóstico , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA