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










Base de dados
Intervalo de ano de publicação
1.
Eur Thyroid J ; 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38241789

RESUMO

BACKGROUND: Intake of potassium iodide (KI) reduces the accumulation of radioactive iodine in the thyroid gland in the event of possible contamination by radioactive iodine released from a nuclear facility. The WHO has stated the need for research for optimal timing, appropriate dosing regimen and safety for repetitive iodine thyroid blocking (ITB). The French PRIODAC project, addressed all these issues, involving prolonged or repeated releases of radioactive iodine. Preclinical studies established an effective dose through pharmacokinetic modeling, demonstrating the safety of repetitive KI treatment without toxicity. SUMMARY: Recent preclinical studies have determined an optimal effective dose for repetitive administration, associated with pharmacokinetic modelling. The results show the safety and absence of toxicity of repetitive treatment with KI. Good laboratory practice level preclinical studies corresponding to individuals > 12 years have shown a safety margin established between animal doses without toxic effect. After approval from the French health authorities, the market authorization of the 2 tablets of KI-65mg/day was defined with a new dosing scheme of a daily repetitive intake of the treatment up to 7 days unless otherwise instructed by the competent authorities for all categories of population except pregnant women, and children under the age of 12 years. CONCLUSIONS: This new marketed authorization resulting from scientific-based evidence obtained as part of the PRIODAC project may serve as an example to further harmonize the application of KI for repetitive ITB in situations of prolonged radioactive release at the European and International levels, under the umbrella of the WHO.

2.
Transl Oncol ; 14(1): 100937, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33217645

RESUMO

For decades, sodium/iodide symporter NIS-mediated iodide uptake has played a crucial role in the radioactive ablation of thyroid cancer cells. NIS-based gene therapy has also become a promising tool for the treatment of tumors of extrathyroidal origin. But its applicability has been hampered by reduced expression of NIS, resulting in a moderated capacity to accumulate 131I and in inefficient ablation. Despite numerous preclinical enhancement strategies, the understanding of NIS expression within tumors remains limited. This study aims at a better understanding of the functional behavior of exogenous NIS expression in the context of malignant solid tumors that are characterized by rapid growth with an insufficient vasculature, leading to hypoxia and quiescence. Using subcutaneous HT29NIS and K7M2NIS tumors, we show that NIS-mediated uptake and NIS expression at the plasma membrane of cancer cells are impaired in the intratumoral regions. For a better understanding of the underlying molecular mechanisms induced by hypoxia and quiescence (separately and in combination), we performed experiments on HT29NIS cancer cells. Hypoxia and quiescence were both found to impair NIS-mediated uptake through mechanisms including NIS mis-localization. Modifications in the expression of proteins and metabolites involved in plasma membrane localization and in energy metabolism were found using untargeted proteomics and metabolomics approaches. In conclusion, our results provide evidence that hypoxia and quiescence impair NIS expression at the plasma membrane, and iodide uptake. Our study also shows that the tumor microenvironment is an important parameter for successful NIS-based cancer treatment.

3.
J Clin Med ; 9(2)2020 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-31979418

RESUMO

(1) Background: We recently showed that iodinated contrast media (ICM) reduced thyroid uptake of iodide independently of free iodide through a mechanism different from that of NaI and involving a dramatic and long-lasting decrease in Na/I symporter expression. The present study aimed at comparing the response of the thyroid to ICM and NaI using a quantitative proteomic approach. (2) Methods: Scintiscans were performed on ICM-treated patients. Micro Single-Photon Emission Computed Tomography (microSPECT/CT) imaging was used to assess thyroid uptakes in ICM- or NaI-treated mice and their response to recombinant human thyroid-stimulating hormone. Total thyroid iodide content and proteome was determined in control, NaI-, or ICM-treated animals. (3) Results: The inhibitory effect of ICM in patients was selectively observed on thyroids but not on salivary glands for up to two months after a systemic administration. An elevated level of iodide was observed in thyroids from NaI-treated mice but not in those from ICM animals. Exposure of the thyroid to NaI modulates 15 cellular pathways, most of which are also affected by ICM treatment (including the elF4 and P706SK cell signaling pathway and INSR identified as an upstream activator in both treatments). In addition, ICM modulates 16 distinct pathways and failed to affect thyroid iodide content. Finally, administration of ICM reduces thyroid-stimulating hormone (TSH) receptor expression which results in a loss of TSH-induced iodide uptake by the thyroid. (4) Conclusions: Common intracellular mechanisms are involved in the ICM- and NaI-induced reduction of iodide uptake. However, ICM fails to affect thyroid iodide content which suggests that the modulation of these common pathways is triggered by separate effectors. ICM also modulates numerous distinct pathways which may account for its long-lasting effect on thyroid uptake. These observations may have implications in the management of patients affected by differentiated thyroid carcinomas who have been exposed to ICM. They also provide the basis for the utilization of ICM-based compounds in radioprotection of the thyroid.

4.
J Cereb Blood Flow Metab ; 40(8): 1709-1723, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31506013

RESUMO

SLC5A8 is a sodium-coupled monocarboxylate and ketone transporter expressed in various epithelial cells. A putative role of SLC5A8 in neuroenergetics has been also hypothesized. To clarify this issue, we studied the cerebral phenotype of SLC5A8-deficient mice during aging. Elderly SLC5A8-deficient mice presented diffuse leukoencephalopathy characterized by intramyelinic oedema without demyelination suggesting chronic energetic crisis. Hypo-metabolism in the white matter of elderly SLC5A8-deficient mice was found using 99mTc-hexamethylpropyleneamine oxime (HMPAO) single-photon emission CT (SPECT). Since the SLC5A8 protein could not be detected in the mouse brain, it was hypothesized that the leukoencephalopathy of aging SLC5A8-deficient mice was caused by the absence of slc5a8 expression in a peripheral organ, i.e. the kidney, where SLC5A8 is strongly expressed. A hyper-excretion of the ketone ß-hydroxybutyrate (BHB) in the urine of SLC5A8-deficient mice was observed and showed that SLC5A8-deficient mice suffered a cerebral BHB insufficiency. Elderly SLC5A8-deficient mice also presented altered glucose metabolism. We propose that the continuous renal loss of BHB leads to a chronic energetic deficiency in the brain of elderly SLC5A8-deficient mice who are unable to counterbalance their glucose deficit. This study highlights the importance of alternative energetic substrates in neuroenergetics especially under conditions of restricted glucose availability.


Assuntos
Envelhecimento/metabolismo , Corpos Cetônicos/urina , Rim/metabolismo , Leucoencefalopatias/metabolismo , Transportadores de Ácidos Monocarboxílicos/deficiência , Substância Branca/metabolismo , Ácido 3-Hidroxibutírico/urina , Envelhecimento/urina , Animais , Glucose/metabolismo , Leucoencefalopatias/urina , Masculino , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Transportadores de Ácidos Monocarboxílicos/genética , Tomografia Computadorizada de Emissão de Fóton Único , Substância Branca/diagnóstico por imagem
5.
Int J Nanomedicine ; 14: 7933-7946, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31686819

RESUMO

BACKGROUND: Human trials combining external radiotherapy (RT) and metallic nanoparticles are currently underway in cancer patients. For internal RT, in which a radioisotope such as radioiodine is systemically administered into patients, there is also a need for enhancing treatment efficacy, decreasing radiation-induced side effects and overcoming radio-resistance. However, if strategies vectorising radioiodine through nanocarriers have been documented, sensitizing the neoplasm through the use of nanotherapeutics easily translatable to the clinic in combination with the standard systemic radioiodine treatment has not been assessed yet. METHOD AND MATERIALS: The present study explored the potential of hybrid poly(methacrylic acid)-grafted gold nanoparticles to improve the performances of systemic 131I-mediated RT on cancer cells and in tumor-bearing mice. Such nanoparticles were chosen based on their ability previously described by our group to safely withstand irradiation doses while exhibiting good biocompatibility and enhanced cellular uptake. RESULTS: In vitro clonogenic assays performed on melanoma and colorectal cancer cells showed that poly(methacrylic acid)-grafted gold nanoparticles (PMAA-AuNPs) could efficiently lead to a marked tumor cell mortality when combined to a low activity of radioiodine, which alone appeared to be essentially ineffective on tumor cells. In vivo, tumor enrichment with PMAA-AuNPs significantly enhanced the killing potential of a systemic radioiodine treatment. CONCLUSION: This is the first report of a simple and reliable nanomedicine-based approach to reduce the dose of radioiodine required to reach curability. In addition, these results open up novel perspectives for using high-Z metallic NPs in additional molecular radiation therapy demonstrating heterogeneous dose distributions.


Assuntos
Ouro/química , Radioisótopos do Iodo/uso terapêutico , Nanopartículas Metálicas/química , Polímeros/química , Animais , Morte Celular , Linhagem Celular Tumoral , Feminino , Humanos , Melanoma Experimental/radioterapia , Nanopartículas Metálicas/administração & dosagem , Nanopartículas Metálicas/ultraestrutura , Camundongos Endogâmicos BALB C , Camundongos Nus , Ácidos Polimetacrílicos/química , Radiossensibilizantes/farmacologia , Dosagem Radioterapêutica , Simportadores/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Radiat Prot Dosimetry ; 182(1): 67-79, 2018 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-30169846

RESUMO

Single dose of potassium iodide (KI) is recommended to prevent the risk of thyroid cancer during nuclear accidents. However in the case of repeated/protracted radioiodine release, a unique dose of KI may not protect efficiently the thyroid against the risk of further developing a radiation-induced cancer. The new WHO guidelines for the use in planning for and responding to radiological and nuclear emergencies identify the need of more data on this subject as one of the four research priorities. The aims of the PRIODAC project are (1) to assess the associated side effects of repeated intakes of KI, (2) to better understand the molecular mechanisms regulating the metabolism of iodine, (3) to revise the regulatory French marketing authorization of 65-mg KI tablets and (4) to develop new recommendations related to the administration of KI toward a better international harmonization. A review of the literature and the preliminary data are presented here.


Assuntos
Radioisótopos do Iodo/efeitos adversos , Neoplasias Induzidas por Radiação/prevenção & controle , Iodeto de Potássio/uso terapêutico , Lesões por Radiação/prevenção & controle , Liberação Nociva de Radioativos , Neoplasias da Glândula Tireoide/prevenção & controle , Relação Dose-Resposta à Radiação , Humanos , Neoplasias Induzidas por Radiação/etiologia , Lesões por Radiação/etiologia , Neoplasias da Glândula Tireoide/etiologia
7.
J Nucl Med ; 59(1): 121-126, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29051343

RESUMO

Perturbation of thyroid iodide uptake is a well-documented side effect of the use of iodinated contrast media (ICM) administered intravenously. This side effect is thought to be mediated by free iodide in ICM formulations, but this hypothesis has never been formally proven. The aim of the present study was to assess the validity of this hypothesis. Methods: We used mass spectrometry analysis to quantify free-iodide contamination in ICM. Established cell lines expressing the Na/I symporter (NIS) were used to quantify the effect of ICM on iodide uptake. SPECT/CT was used to measure the in vivo uptake of 99mTc-pertechnetate and 123I in 2 NIS-expressing mouse tissues, thyroid and salivary glands. Scintiscans of ICM-naïve and ICM-administered patients were compared. Immunohistologic and Western blot analyses were performed to evaluate NIS protein expression in these organs. Results: Although free iodide was present in ICM formulations, in vitro uptake of iodide by NIS-expressing cells was not significantly affected by ICM. In mice, intravenous or sublingual administration of ICM led to a reduction in radiotracer uptake by the thyroid, accompanied by a dramatic reduction in NIS protein expression in this tissue. In the salivary glands, neither radiotracer uptake nor NIS protein expression was affected by ICM. The thyroid-selective effect of ICM was also observed in humans. Administration of potassium iodide as a source of free iodide led to a diminution of 99mTc-pertechnetate uptake in both mouse thyroid and mouse salivary glands. Altogether, these data rule out a direct intervention of free iodide in the perturbation of thyroid uptake and suggest a direct and selective effect of ICM on the thyroid. Conclusion: We demonstrated that ICM reduce thyroid uptake of iodide independently of free iodide. This effect is due to a specific and dramatic decrease in NIS expression in thyrocytes. These data cast serious doubt on the relevance of measuring urinary iodide concentration to evaluate the delay between ICM administration and radioiodine therapy in patients with differentiated thyroid carcinoma. Finally, the ability of ICM to perturb iodide uptake in the thyroid may be used in radioprotection.


Assuntos
Meios de Contraste/química , Meios de Contraste/farmacologia , Halogenação , Iodetos/metabolismo , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Células HT29 , Humanos , Camundongos , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único , Glândula Tireoide/diagnóstico por imagem
8.
Thyroid ; 26(11): 1614-1622, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27349131

RESUMO

BACKGROUND: MicroSPECT/CT imaging was used to quantitatively evaluate how iodide uptake in the mouse thyroid is influenced by (i) route of iodine administration; (ii) injection of recombinant human thyrotropin (rhTSH); and (iii) low iodide diet (LID) in euthyroid and triiodothyronine (T3)-treated mice. METHODS: Pertechnetate (99mTcO4-) and 123I thyroid uptake in euthyroid and T3-treated animals fed either a normal-iodine diet (NID) or an LID, treated or not with rhTSH, and radiotracer administered intravenously, subcutaneously, intraperitoneally or by gavage, were assessed using microSPECT/CT imaging. Western blotting was performed to measure sodium/iodide symporter expression levels in the thyroid. RESULTS: Systemic administration of radioiodide resulted in a higher (2.35-fold in NID mice) accumulation of iodide in the thyroid than oral administration. Mice fed LID with systemic radioiodide administration showed a further two-fold increase in thyroid iodide uptake to yield a ∼5-fold increase in uptake compared to the standard NID/oral route. Although rhTSH injections stimulated thyroid activity in both euthyroid and T3-treated mice fed the NID, uptake levels for T3-treated mice remained low compared with those for the euthyroid mice. Combining LID and rhTSH in T3-treated mice resulted in a 2.8-fold higher uptake compared with NID/T3/rhTSH mice and helped restore thyroid activity to levels equivalent to those of euthyroid animals. CONCLUSIONS: Systemic radioiodide administration results in higher thyroidal iodide levels than oral administration, particularly in LID-fed mice. These data highlight the importance of LID, both in euthyroid and T3-treated, rhTSH-injected mice. Extrapolated to human patients, and in the context of clinical guidelines for the preparation of differentiated thyroid cancer patients, our data indicate that LID can potentiate the efficacy of rhTSH treatment in T3-treated patients.


Assuntos
Radioisótopos do Iodo/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Glândula Tireoide/diagnóstico por imagem , Tri-Iodotironina/farmacocinética , Administração Oral , Animais , Dieta/efeitos adversos , Feminino , Terapia de Reposição Hormonal/efeitos adversos , Humanos , Injeções Intraperitoneais , Injeções Intravenosas , Injeções Subcutâneas , Iodo/administração & dosagem , Iodo/efeitos adversos , Radioisótopos do Iodo/administração & dosagem , Radioisótopos do Iodo/metabolismo , Camundongos Endogâmicos C57BL , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/farmacologia , Pertecnetato Tc 99m de Sódio/metabolismo , Pertecnetato Tc 99m de Sódio/farmacocinética , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/metabolismo , Tireotropina/administração & dosagem , Tireotropina/efeitos adversos , Tireotropina/farmacologia , Distribuição Tecidual , Tomografia Computadorizada de Emissão de Fóton Único , Tri-Iodotironina/administração & dosagem , Tri-Iodotironina/metabolismo
9.
Artigo em Inglês | MEDLINE | ID: mdl-27164586

RESUMO

Three scattering models were examined for characterizing ex vivo canine livers and HT29 mouse tumors in the 10-38- and the 15-42-MHz frequency bandwidth, respectively. The spherical Gaussian model (SGM) and the fluid sphere model (FSM) that were examined are suitable for dealing with sparse media, whereas the structure factor model (SFM) is adapted for characterizing concentrated media. For the canine livers, the scatterer radius and the acoustic concentration estimated with the three models were similar and matched well the nuclear structures obtained from histological analysis (with relative errors less than 7%). These results show that the livers could be considered as a diluted medium and that the nuclei in liver could be a dominant source of scattering. For the homogeneous mouse tumors, containing mostly viable HT29 cells, scatterer radius and volume fraction estimated with the SFM showed good agreement with the whole cell structures obtained from histological analysis (with relative errors less than 15%), whereas the sparse models (the SGM and the FSM) gave no consistent quantitative ultrasound parameters. This suggests that the viable HT29 cell areas have densely packed cellular content and that the whole HT29 cell could be responsible for scattering. For the heterogeneous tumors, the hyperechogenic zones observed in the B-mode images were linked to the presence of small necrotic areas surrounded by viable HT29 cells. Comparison between sparse and concentrated models shows that these hyperechogenic zones could be considered as a concentrated medium.


Assuntos
Fígado/diagnóstico por imagem , Neoplasias Experimentais/diagnóstico por imagem , Ultrassonografia , Acústica , Animais , Linhagem Celular Tumoral , Cães , Humanos , Camundongos , Análise Espectral
10.
Biomaterials ; 45: 10-7, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25662490

RESUMO

In the present study, we evaluated, in mice, the efficacy of the tetrafunctional block copolymer 704 as a nonviral gene delivery vector to the lungs. SPECT/CT molecular imaging of gene expression, biochemical assays, and immunohistochemistry were used. Our dataset shows that the formulation 704 resulted in higher levels of reporter gene expression than the GL67A formulation currently being used in a clinical trial in cystic fibrosis patients. The inflammatory response associated with this gene transfer was lower than that induced by the GL67A formulation, and the 704 formulation was amenable to repeated administrations. The cell types transfected by the 704 formulation were type I and type II pneumocytes, and transgene expression could not be detected in macrophages. These results emphasize the relevance of the 704 formulation as a nonviral gene delivery vector for lung gene therapy. Further studies will be required to validate this vector in larger animals, in which the lungs are more similar to human lungs.


Assuntos
Técnicas de Transferência de Genes , Pulmão/metabolismo , Polímeros/química , Animais , Cloranfenicol O-Acetiltransferase/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Inflamação/patologia , Pulmão/diagnóstico por imagem , Pulmão/patologia , Camundongos Endogâmicos BALB C , Simportadores/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X , Transfecção , Transgenes
11.
PLoS One ; 9(3): e92729, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24663284

RESUMO

Low-energy Auger and conversion electrons deposit their energy in a very small volume (a few nm3) around the site of emission. From a radiotoxicological point of view the effects of low-energy electrons on normal tissues are largely unknown, understudied, and generally assumed to be negligible. In this context, the discovery that the low-energy electron emitter, 99mTc, can induce stunning on primary thyrocytes in vitro, at low absorbed doses, is intriguing. Extrapolated in vivo, this observation suggests that a radioisotope as commonly used in nuclear medicine as 99mTc may significantly influence thyroid physiology. The aims of this study were to determine whether 99mTc pertechnetate (99mTcO4-) is capable of inducing thyroid stunning in vivo, to evaluate the absorbed dose of 99mTcO4- required to induce this stunning, and to analyze the biological events associated/concomitant with this effect. Our results show that 99mTcO4--mediated thyroid stunning can be observed in vivo in mouse thyroid. The threshold of the absorbed dose in the thyroid required to obtain a significant stunning effect is in the range of 20 Gy. This effect is associated with a reduced level of functional Na/I symporter (NIS) protein, with no significant cell death. It is reversible within a few days. At the cellular and molecular levels, a decrease in NIS mRNA, the generation of double-strand DNA breaks, and the activation of the p53 pathway are observed. Low-energy electrons emitted by 99mTc can, therefore, induce thyroid stunning in vivo in mice, if it is exposed to an absorbed dose of at least 20 Gy, a level unlikely to be encountered in clinical practice. Nevertheless this report presents an unexpected effect of low-energy electrons on a normal tissue in vivo, and provides a unique experimental setup to understand the fine molecular mechanisms involved in their biological effects.


Assuntos
Pertecnetato Tc 99m de Sódio/efeitos adversos , Glândula Tireoide/efeitos da radiação , Animais , Transporte Biológico , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Elétrons , Feminino , Regulação da Expressão Gênica/efeitos da radiação , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Radiometria , Pertecnetato Tc 99m de Sódio/metabolismo , Simportadores/genética , Simportadores/metabolismo , Glândula Tireoide/citologia , Glândula Tireoide/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X
12.
PLoS One ; 7(3): e34086, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22470517

RESUMO

The utilisation of the Na/I symporter (NIS) and associated radiotracers as a reporter system for imaging gene expression is now reaching the clinical setting in cancer gene therapy applications. However, a formal assessment of the methodology in terms of normalisation of the data still remains to be performed, particularly in the context of the assessment of activities in individual subjects in longitudinal studies. In this context, we administered to mice a recombinant, replication-incompetent adenovirus encoding rat NIS, or a human colorectal carcinoma cell line (HT29) encoding mouse NIS. We used (99m)Tc pertechnetate as a radiotracer for SPECT/CT imaging to determine the pattern of ectopic NIS expression in longitudinal kinetic studies. Some animals of the cohort were culled and NIS expression was measured by quantitative RT-PCR and immunohistochemistry. The radioactive content of some liver biopsies was also measured ex vivo. Our results show that in longitudinal studies involving datasets taken from individual mice, the presentation of non-normalised data (activity expressed as %ID/g or %ID/cc) leads to 'noisy', and sometimes incoherent, results. This variability is due to the fact that the blood pertechnetate concentration can vary up to three-fold from day to day. Normalisation of these data with blood activities corrects for these inconsistencies. We advocate that, blood pertechnetate activity should be determined and used to normalise the activity measured in the organ/region of interest that expresses NIS ectopically. Considering that NIS imaging has already reached the clinical setting in the context of cancer gene therapy, this normalisation may be essential in order to obtain accurate and predictive information in future longitudinal clinical studies in biotherapy.


Assuntos
Simportadores/análise , Tomografia Computadorizada de Emissão de Fóton Único , Adenoviridae/genética , Animais , Técnicas de Transferência de Genes , Genes Reporter , Células HT29 , Humanos , Imuno-Histoquímica , Cinética , Fígado/metabolismo , Fígado/patologia , Estudos Longitudinais , Camundongos , Camundongos Endogâmicos BALB C , RNA/metabolismo , RNA/normas , Compostos Radiofarmacêuticos/sangue , Reação em Cadeia da Polimerase em Tempo Real/normas , Pertecnetato Tc 99m de Sódio/sangue , Simportadores/genética , Simportadores/metabolismo
13.
Thyroid ; 20(5): 519-26, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20384490

RESUMO

BACKGROUND: (99m)Tc pertechnetate is a well-known anion, used for clinical imaging of thyroid function. This gamma emitter is transported by the sodium iodide symporter but is not incorporated into thyroglobulin. Scintigraphy using (99m)Tc pertechnetate or (123)iodide represents a powerful tool for the study of sodium iodide symporter activity in different organs of living animal models. However, in many studies that have been performed in mice, the thyroid could not be distinguished from the salivary glands. In this work, we have evaluated the use of a clinically dedicated single-photon emission computed tomography (SPECT) camera for thyroid imaging and assessed what improvements are necessary for the development of this technique. METHODS: SPECT of the mouse neck region, with pinhole collimation and geometric calibration, was used for the individual measurement of (99m)Tc pertechnetate uptake in the thyroid and the salivary glands. Uptake in the stomach was studied by planar whole-body imaging. Uptake kinetics and biodistribution studies were performed by sequential imaging. RESULTS: This work has shown that thyroid imaging in living mice can be performed with a SPECT camera originally built for clinical use. Our experiments indicate that (99m)Tc pertechnetate uptake is faster in the thyroid than in the salivary glands and the stomach. The decrease in (99m)Tc pertechnetate uptake after injection of iodide or perchlorate as competitive inhibitors was also studied. The resulting rate decreases were faster in the thyroid than in the salivary glands or the stomach. CONCLUSIONS: We have shown that a clinically dedicated SPECT camera can be used for thyroid imaging. In our experiments, SPECT imaging allowed the analysis of (99m)Tc pertechnetate accumulation in individual organs and revealed differences in uptake kinetics.


Assuntos
Compostos Radiofarmacêuticos/farmacocinética , Pertecnetato Tc 99m de Sódio/farmacocinética , Glândula Tireoide/metabolismo , Animais , Mucosa Gástrica/metabolismo , Processamento de Imagem Assistida por Computador , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pescoço/diagnóstico por imagem , Glândulas Salivares/diagnóstico por imagem , Glândulas Salivares/metabolismo , Iodeto de Sódio , Estômago/diagnóstico por imagem , Glândula Tireoide/diagnóstico por imagem , Distribuição Tecidual , Tomografia Computadorizada de Emissão de Fóton Único , Imagem Corporal Total
14.
Proc Natl Acad Sci U S A ; 103(25): 9643-8, 2006 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-16769891

RESUMO

Platelet-derived lysophosphatidic acid (LPA) supports the progression of breast and ovarian cancer metastasis to bone. The mechanisms through which LPA promotes bone metastasis formation are, however, unknown. Here we report that silencing of the type 1 LPA receptor (LPA(1)) in cancer cells blocks the production of tumor-derived cytokines that are potent activators of osteoclast-mediated bone destruction and significantly reduces the progression of osteolytic bone metastases. Moreover, functional blockade of LPA action on its cognate receptor LPA(1) using a pharmacological antagonist mimics the effects of silencing LPA(1) in tumor cells in vitro and substantially reduces bone metastasis progression in animals. Overall, these results suggest that inhibition of platelet-derived LPA action on LPA(1) expressed by tumor cells may be a promising therapeutic target for patients with bone metastases.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Metástase Neoplásica/tratamento farmacológico , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Receptores de Ácidos Lisofosfatídicos/classificação , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Citocinas/metabolismo , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Isoxazóis/uso terapêutico , Lisofosfolipídeos/farmacologia , Camundongos , Metástase Neoplásica/patologia , Osteoclastos/metabolismo , Propionatos/uso terapêutico , Receptores de Ácidos Lisofosfatídicos/metabolismo
15.
Int J Cancer ; 116(5): 686-91, 2005 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-15838828

RESUMO

The antiangiogenic extracellular matrix protein thrombospondin-1 (TSP-1) inhibits tumor growth and metastasis in animals. However, the clinical relevance of such findings are equivocal as increased stromal TSP-1 expression has been associated with either good or poor prognosis. In an effort to obtain a more integrated understanding of the role of TSP-1 in breast cancer, we first used a breast tumorigenesis model in which tumor-associated stromal fibroblasts were engineered to produce high levels of TSP-1. We demonstrate here that stromal TSP-1 delayed human MDA-MB-231/B02 breast tumor growth. However, this delay in MDA-MB-231/B02 tumor growth upon exposure to TSP-1 was associated with an increased vascular endothelial growth factor (VEGF) expression in tumor cells themselves, leading to a tumor growth rate comparable to that of tumors whose fibroblasts did not overproduce TSP-1. Clinical evidence also suggested that primary breast carcinomas have adapted to escape the effects of stromal TSP-1. TSP-1 was found to be expressed in the stroma of human breast carcinomas where, although its level correlated with decreased vascularization, it was unexpectedly associated with a reduction of relapse-free survival. In metastatic axillary lymph nodes, tumor cells expressed high levels of VEGF and TSP-1 expression were no longer associated with a decreased vascularization. Overall, these results suggest that a resistance may develop early in human breast cancers as a result of high in situ exposure to stromal TSP-1, leading to disease progression.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Neovascularização Patológica/prevenção & controle , Trombospondina 1/fisiologia , Animais , Neoplasias da Mama/patologia , Movimento Celular/efeitos dos fármacos , Progressão da Doença , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Transplante de Neoplasias , Trombospondina 1/análise , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular/análise
16.
Cancer Res ; 65(1): 300-8, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15665307

RESUMO

In order to understand why the angiogenesis inhibitor thrombospondin-1 (TSP1) is often, although not always, associated with prostatic tumors, we have investigated its relationship with the testosterone and the vasculature on which both normal and tumorigenic prostatic epithelia depend. In vivo, androgen withdrawal led to increased TSP1 production and decreased vascularization in the normal rat prostate which was reversed by androgen replacement. Androgen repression of TSP1 production occurred at the transcriptional level and was dependent on the presence of the first intron of the TSP1 gene. In an experimental model of prostate tumorigenesis, TSP1, when delivered by admixed stromal fibroblasts, markedly delayed LNCaP tumor growth and limited tumor vascularization. However, prolonged exposure to TSP1 resulted in the growth of tumors secreting high levels of vascular endothelial growth factor in the bloodstream of tumor-bearing animals and tumor growth was no longer sensitive to TSP1 inhibitory effects. Clinical evidence also suggested that prostate carcinomas are able to adapt to escape the antiangiogenic effects of TSP1. In human androgen-dependent localized prostate carcinomas, TSP1 expression was inversely correlated with blood vessel density. Androgen deprivation in patients with hormone-responsive tumors led to increased TSP1 expression and vascular regression. In contrast, despite a sustained expression in the tumor bed, TSP1 was no longer associated with decreased vascularization in hormone-refractory prostate tumors. Overall, these results suggest that the high in situ TSP1 exposure triggered by androgen deprivation in patients with prostate cancer could lead to early tumor resistance. Such patients could benefit from a combination of androgen deprivation and antiangiogenic therapy in order to minimize the induction of such tumor escape.


Assuntos
Androgênios/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Neovascularização Patológica/prevenção & controle , Neovascularização Fisiológica/efeitos dos fármacos , Neoplasias da Próstata/irrigação sanguínea , Trombospondina 1/genética , Inibidores da Angiogênese/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Nus , Orquiectomia , Próstata , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Ratos , Ratos Sprague-Dawley , Trombospondina 1/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/análise
17.
J Clin Invest ; 114(12): 1714-25, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15599396

RESUMO

The role of lysophosphatidic acid (LPA) in cancer is poorly understood. Here we provide evidence for a role of LPA in the progression of breast cancer bone metastases. LPA receptors LPA(1), LPA(2), and LPA(3) were expressed in human primary breast tumors and a series of human breast cancer cell lines. The inducible overexpression of LPA(1) in MDA-BO2 breast cancer cells specifically sensitized these cells to the mitogenic action of LPA in vitro. In vivo, LPA(1) overexpression in MDA-BO2 cells enhanced the growth of subcutaneous tumor xenografts and promoted bone metastasis formation in mice by increasing both skeletal tumor growth and bone destruction. This suggested that endogenous LPA was produced in the tumor microenvironment. However, MDA-BO2 cells or transfectants did not produce LPA. Instead, they induced the release of LPA from activated platelets which, in turn, promoted tumor cell proliferation and the LPA(1)-dependent secretion of IL-6 and IL-8, 2 potent bone resorption stimulators. Moreover, platelet-derived LPA deprivation in mice, achieved by treatment with the platelet antagonist Integrilin, inhibited the progression of bone metastases caused by parental and LPA(1)-overexpressing MDA-BO2 cells and reduced the progression of osteolytic lesions in mice bearing CHO-beta3wt ovarian cancer cells. Overall, our data suggest that, at the bone metastatic site, tumor cells stimulate the production of LPA from activated platelets, which enhances both tumor growth and cytokine-mediated bone destruction.


Assuntos
Plaquetas/metabolismo , Osso e Ossos/patologia , Neoplasias da Mama/patologia , Lisofosfolipídeos/metabolismo , Animais , Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Reabsorção Óssea , Osso e Ossos/metabolismo , Neoplasias da Mama/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Cricetinae , Meios de Cultura/farmacologia , Citocinas/metabolismo , Progressão da Doença , Relação Dose-Resposta a Droga , Doxiciclina/farmacologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Antígeno Ki-67/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitógenos/metabolismo , Modelos Biológicos , Metástase Neoplásica , Osteoclastos/metabolismo , Osteólise , Fosfolipase D/metabolismo , Ativação Plaquetária , Agregação Plaquetária , RNA/química , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transfecção
18.
Cancer Res ; 63(14): 3919-22, 2003 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12873985

RESUMO

In the past few years, several laboratories have developed antiangiogenic molecules that starve tumors by targeting their vasculature and we have shown that, when produced in tumors, the antiangiogenic molecule thrombospondin-1 (TSP1) reduces the vascularization and delays tumor onset. Yet over time, tumor cells producing active TSP1 do eventually form exponentially growing tumors. These tumors are composed of cells secreting unusually high amounts of the angiogenic stimulator vascular endothelial growth factor (VEGF) that are sufficient to overcome the inhibitory TSP1. Here, we use short double-stranded RNA (siRNA) to trigger RNA interference and thereby impair the synthesis of VEGF and ask if this inability to produce VEGF prevents the development of TSP1 resistance. Systemic in vivo administration of crude anti-VEGF siRNA reduced the growth of unaltered fibrosarcoma tumor cells, and when the anti-VEGF siRNA was expressed from tumor cells themselves, such inhibition was synergistic with the inhibitory effects derived from TSP1 secretion by the tumor cells. Anti-VEGF siRNA delayed the emergence of TSP1-resistant tumors and strikingly reduced their subsequent growth rate.


Assuntos
Fatores de Crescimento Endotelial/antagonistas & inibidores , Fibrossarcoma/irrigação sanguínea , Linfocinas/antagonistas & inibidores , Neovascularização Patológica/prevenção & controle , RNA Interferente Pequeno/genética , Trombospondina 1/fisiologia , Animais , Divisão Celular/genética , Fatores de Crescimento Endotelial/biossíntese , Fatores de Crescimento Endotelial/genética , Feminino , Fibrossarcoma/genética , Fibrossarcoma/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/genética , Linfocinas/biossíntese , Linfocinas/genética , Camundongos , Camundongos Nus , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Ratos , Trombospondina 1/biossíntese , Trombospondina 1/genética , Transfecção , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
19.
Cancer Res ; 62(22): 6538-44, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12438248

RESUMO

Bisphosphonates (BPs) are used currently in the treatment of patients with bone metastases because these compounds inhibit bone resorption. We examined here the effects of BPs on inhibition of endothelial cell functions in vitro and in vivo. Treatment of endothelial cells with BPs (clodronate, risedronate, ibandronate, and zoledronic acid) reduced proliferation, induced apoptosis, and decreased capillary-like tube formation in vitro. Quantification of blood vessels in bone biopsy specimens from patients with Paget's disease before and after clodronate treatment showed a 40% reduction of the vascularization after BP treatment. However, such a decreased vascularity could be secondary to a reduction of bone resorption. Therefore, the tissue distribution of [14C]BPs in male rats was examined to develop an angiogenesis model in a noncalcified tissue where BPs could accumulate. [14C]BPs (zoledronic acid, ibandronate, and clodronate) not only accumulated in bone but also transiently accumulated in the prostate. The effects of BPs on testosterone-induced revascularization of the prostate gland in castrated rats were then studied. Testosterone in combination with ibandronate or zoledronic acid induced a 17-35% reduction of the prostate weight compared with castrated rats treated with testosterone alone. Blood vessel immunostaining on prostate tissue sections revealed that both ibandronate and zoledronic acid induced a 50% reduction of the revascularization of the prostate gland. Moreover, zoledronic acid did not alter testosterone-induced activity of a luciferase gene reporter construct transfected in androgen-dependent prostatic cells, indicating that this BP did not directly interfere with testosterone. In conclusion, BPs have in vivo antiangiogenic properties, which could be of relevance to improve therapy and prevention of bone metastasis. In addition, our results extend the potential clinical use of BPs to patients with early prostate cancer.


Assuntos
Inibidores da Angiogênese/farmacologia , Difosfonatos/farmacologia , Ácido Etidrônico/análogos & derivados , Neovascularização Fisiológica/efeitos dos fármacos , Próstata/irrigação sanguínea , Testosterona/farmacologia , Inibidores da Angiogênese/farmacocinética , Animais , Osso e Ossos/irrigação sanguínea , Osso e Ossos/patologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Ácido Clodrônico/farmacocinética , Ácido Clodrônico/farmacologia , Difosfonatos/farmacocinética , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Ácido Etidrônico/farmacocinética , Ácido Etidrônico/farmacologia , Humanos , Ácido Ibandrônico , Imidazóis/farmacocinética , Imidazóis/farmacologia , Masculino , Orquiectomia , Osteíte Deformante/patologia , Próstata/citologia , Próstata/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ácido Risedrônico , Testosterona/antagonistas & inibidores , Distribuição Tecidual , Ácido Zoledrônico
20.
FASEB J ; 16(10): 1266-8, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12153995

RESUMO

The reasons why tumor cells metastasize to bone remain obscure. There is some evidence to support the theory that integrins (acting as cell surface adhesion receptors) play a role in mediating metastasis in certain organs. Here, we report that overexpression of a functionally active integrin alpha(v)b3 in Chinese hamster ovary (CHO) tumor cells drastically increased the incidence, number, and area of bone metastases in nude mice compared with those observed in mock-transfected CHO cells (CHO dhfr+) or in CHO cells expressing a functionally inactive integrin alpha(v)b3 (CHO beta3Delta744). Moreover, a breast cancer cell line (B02) established from bone metastases caused by MDA-MB-231 cells constitutively overexpressed integrin alpha(v)b3, whereas the cell surface expression level of other integrins remained unchanged. In vivo, the extent of bone metastases in B02-bearing mice was significantly increased compared with that of MDA-MB-231-bearing mice. In vitro, B02 cells and CHO cells expressing a functionally active integrin alpha(v)b3 exhibited substantially increased invasion of and adhesion to mineralized bone, bone sialoprotein, and collagen compared with those found with MDA-MB-231, CHO dhfr+, and CHO beta3Delta744 cells, respectively. Overall, our findings suggest that integrin alpha(v)b3 expression in tumor cells accelerates the development of osteolytic lesions, presumably through increased invasion of and adhesion to bone.


Assuntos
Neoplasias Ósseas/secundário , Receptores de Vitronectina/metabolismo , Receptores de Vitronectina/fisiologia , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Células CHO , Carcinoma/metabolismo , Carcinoma/patologia , Carcinoma/secundário , Adesão Celular , Divisão Celular , Movimento Celular , Colágeno/metabolismo , Cricetinae , Feminino , Sialoproteína de Ligação à Integrina , Camundongos , Camundongos Nus , Modelos Biológicos , Invasividade Neoplásica , Receptores de Vitronectina/genética , Sialoglicoproteínas/metabolismo , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...