Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Nucl Med Mol Imaging ; 38(4): 623-32, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21170526

RESUMO

PURPOSE: Radiolabelled peptides used for peptide receptor radionuclide therapy are excreted mainly via the kidneys and are partly reabsorbed and retained in the proximal tubular cells. The resulting high renal radiation dose can cause nephrotoxicity, limiting the maximum activity dose and the effectiveness of peptide receptor radionuclide therapy. The mechanisms of kidney reabsorption of these peptides are incompletely understood, but the scavenger receptor megalin has been shown to play a role in the reabsorption of (111)In-octreotide. In this study, the role of megalin in the renal reabsorption of various relevant radiolabelled peptides was investigated. METHODS: Groups of kidney-specific megalin-deficient mice and wild-type mice were injected with (111)In-labelled somatostatin, exendin, neurotensin or minigastrin analogues. Single photon emission computed tomographic (SPECT) images of the kidneys were acquired and analysed quantitatively, or the animals were killed 3 h after injection and the activity concentration in the kidneys was measured. RESULTS: Megalin-deficient mice showed significantly lower uptake of all studied radiolabelled peptides in the kidneys, ranging from 22% ((111)In-octreotide) to 65% ((111)In-exendin) of uptake in wild-type kidneys. Quantitative analysis of renal uptake by SPECT and ex vivo measurements showed a very good correlation. CONCLUSION: Megalin is involved in the renal reabsorption of radiolabelled octreotide, octreotate, exendin, neurotensin and minigastrin. This knowledge may help in the design of strategies to reduce this reabsorption and the resulting nephrotoxicity in peptide receptor radionuclide therapy, enabling more effective therapy. Small-animal SPECT is an accurate tool, allowing in vivo quantification of renal uptake and serial measurements in individual mice.


Assuntos
Rim/diagnóstico por imagem , Rim/metabolismo , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Peptídeos/metabolismo , Peptídeos/farmacocinética , Tomografia Computadorizada de Emissão de Fóton Único , Animais , Feminino , Imuno-Histoquímica , Marcação por Isótopo , Masculino , Camundongos , Especificidade de Órgãos , Peptídeos/uso terapêutico , Transporte Proteico , Distribuição Tecidual
2.
J Nucl Med ; 51(12): 1962-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21078795

RESUMO

UNLABELLED: High kidney radiation doses during clinical peptide receptor radionuclide therapy (PRRT) with ß-particle-emitting radiolabeled somatostatin analogs will lead to renal failure several months after treatment, urging the coinfusion of the cationic amino acids lysine and arginine to reduce the renal radiation dose. In rat PRRT studies, renal protection by the coadministration of lysine was confirmed by histologic examination of kidney specimens indicating nephrotoxicity. In the current study, we investigated dedicated small-animal SPECT/CT renal imaging in rats to monitor renal function in vivo during follow-up of PRRT, with and without lysine. METHODS: The following 3 groups of rats were imaged using a multipinhole SPECT/CT camera: controls (group 1) and rats at more than 90 d after therapy with 460 MBq (15 µg) of (177)Lu-DOTA-Tyr(3)-octreotate without (group 2) or with (group 3) a 400-mg/kg lysine coinjection as kidney protection (n ≥ 6 per group). At 90 and 140 d after therapy, static kidney scintigraphy was performed at 2 h after injection of 25 MBq of (99m)Tc-dimercaptosuccinic acid ((99m)Tc-DMSA). In addition, dynamic dual-isotope renography was performed using 50 MBq of (111)In-diethylenetriaminepentaacetic acid ((111)In-DTPA) and 50 MBq of (99m)Tc-mercaptoacetyltriglycine ((99m)Tc-MAG3) at 100-120 d after therapy. RESULTS: (111)In-DTPA and (99m)Tc-MAG3 studies revealed a time-activity pattern comparable to those in patients, with a peak at 2-6 min followed by a decline of renal radioactivity. Reduced (111)In-DTPA, (99m)Tc-MAG3, and (99m)Tc-DMSA uptake indicated renal damage in group 2, whereas group 3 showed only a decrease of (99m)Tc-MAG3 peak activity. These results indicating nephrotoxicity in group 2 and renal protection in group 3 correlated with levels of urinary protein and serum creatinine and urea and were confirmed by renal histology. CONCLUSION: Quantitative dynamic dual-isotope imaging using both (111)In-DTPA and (99m)Tc-MAG3 and static (99m)Tc-DMSA imaging in rats is feasible using small-animal SPECT, enabling longitudinal monitoring of renal function. (99m)Tc-MAG3 renography, especially, appears to be a more sensitive marker of tubular function after PRRT than serum chemistry or (99m)Tc-DMSA scintigraphy.


Assuntos
Rim/fisiologia , Octreotida/análogos & derivados , Compostos Organometálicos/uso terapêutico , Compostos Radiofarmacêuticos/uso terapêutico , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Albuminas , Animais , Autorradiografia , Creatinina/sangue , Taxa de Filtração Glomerular , Rim/diagnóstico por imagem , Nefropatias/prevenção & controle , Túbulos Renais/fisiologia , Lisina/farmacologia , Masculino , Octreotida/uso terapêutico , Ácido Pentético/análogos & derivados , Proteinúria/metabolismo , Lesões por Radiação/prevenção & controle , Renografia por Radioisótopo , Ratos , Ratos Endogâmicos Lew , Ácido Dimercaptossuccínico Tecnécio Tc 99m , Tecnécio Tc 99m Mertiatida
3.
J Nucl Med ; 51(6): 973-7, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20484435

RESUMO

UNLABELLED: We determined the renal radiation dose of a series of (111)In-labeled peptides using animal SPECT. Because the animals' health deteriorated, renal toxicity was assessed. METHODS: Wild-type and megalin-deficient mice were imaged repeatedly at 3- to 6-wk intervals to quantify renal retention after injection of 40-50 MBq of (111)In-diethylenetriaminepentaacetic acid-labeled peptides (octreotide, exendin, octreotate, neurotensin, and minigastrin analogs), and the absorbed kidney radiation doses were estimated. Body weight, renal function parameters, and renal histology were determined at 16-20 wk after the first scan and compared with those in naive animals. RESULTS: Because of high renal retention, (111)In-diethylenetriaminepentaacetic acid-exendin-4 scans resulted in a 70-Gy kidney radiation dose in wild-type mice. Megalin-deficient kidneys received 20-40 Gy. The other peptides resulted in much lower renal doses. Kidney function monitoring indicated renal damage in imaged animals. CONCLUSION: Micro-SPECT enables longitudinal studies in 1 animal. However, long-term nephrotoxic effects may be induced after high renal radiation doses, even with (111)In-labeled radiotracers.


Assuntos
Radioisótopos de Índio , Rim/efeitos da radiação , Peptídeos , Tomografia Computadorizada de Emissão de Fóton Único/efeitos adversos , Animais , Exenatida , Feminino , Injeções , Rim/patologia , Rim/fisiopatologia , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Masculino , Camundongos , Especificidade de Órgãos , Ácido Pentético/química , Peptídeos/administração & dosagem , Peptídeos/química , Doses de Radiação , Risco , Fatores de Tempo , Tomografia Computadorizada por Raios X , Peçonhas/química
4.
Int J Cancer ; 126(12): 2826-34, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19876914

RESUMO

Human prostate cancer (PC) overexpresses the gastrin-releasing peptide receptor (GRPR). Radiolabeled GRPR-targeting analogs of bombesin (BN) have successfully been introduced as potential tracers for visualization and treatment of GRPR-overexpressing tumors. A previous study showed GRPR-mediated binding of radiolabeled BN analogs in androgen-dependent but not in androgen-independent xenografts representing the more advanced stages of PC. We have further investigated the effect of androgen modulation on GRPR-expression in three androgen-dependent human PC-bearing xenografts: PC295, PC310 and PC82 using the androgen-independent PC3-model as a reference. Effects of androgen regulation on GRPR expression were initially studied on tumors obtained from our biorepository of xenograft tissues performing reverse transcriptase polymerase chain reaction (RT-PCR) and autoradiography ((125)I-universal-BN). A prospective biodistribution study ((111)In-MP2653) and subsequent autoradiography ((125)I-GRP and (111)In-MP2248) was than performed in castrated and testosterone resupplemented tumor-bearing mice. For all androgen-dependent xenografts, tumor uptake and binding decreased drastically after 7 days of castration. Resupplementation of testosterone to castrated animals restored GRPR expression extensively. Similar findings were concluded from the initial autoradiography and RT-PCR studies. Results from RT-PCR, for which human specific primers are used, indicate that variations in GRPR expression can be ascribed to mRNA downregulation and not to castration-induced reduction in the epithelial fraction of the xenograft tumor tissue. In conclusion, expression of human GRPR in androgen-dependent PC xenografts is reduced by androgen ablation and is reversed by restoring the hormonal status of the animals. This knowledge suggests that hormonal therapy may affect GRPR expression in PC tissue making GRPR-based imaging and therapy especially suitable for non-hormonally treated PC patients.


Assuntos
Androgênios/farmacologia , Bombesina/farmacocinética , Peptídeo Liberador de Gastrina/metabolismo , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias da Próstata/metabolismo , Receptores da Bombesina/metabolismo , Animais , Autorradiografia , Bombesina/análogos & derivados , Castração , Peptídeo Liberador de Gastrina/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Radioisótopos de Índio , Masculino , Taxa de Depuração Metabólica , Camundongos , Camundongos Nus , Neoplasias Hormônio-Dependentes/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Compostos Radiofarmacêuticos/farmacocinética , Receptores da Bombesina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testosterona/administração & dosagem , Distribuição Tecidual , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Eur J Nucl Med Mol Imaging ; 36(12): 1968-76, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19585115

RESUMO

PURPOSE: Peptide receptor radionuclide therapy using ß-emitting radiolabelled somatostatin analogues like DOTA,Tyr3-octreotate shows beneficial results in patients suffering from somatostatin receptor overexpressing tumours. However, after high-dose therapy partial renal reabsorption of radiopeptides may lead to nephrotoxicity. Co-infusion of lysine/arginine lowers renal retention of these radiopeptides without affecting tumour uptake. Recently co-administration of Gelofusine has been described to have a comparable kidney-protecting effect in rats. In the present study optimal dosing of Gelofusine co-administration was studied in tumour-bearing rats. METHODS: Doses of 40, 80, 120 or 160 mg/kg Gelofusine were co-injected with 15 µg DOTA,Tyr3-octreotate, labelled with 3 MBq 111In for biodistribution (24 h post-injection, n = 4 per group) and with 60 MBq 111In for microSPECT imaging experiments at 3, 24 and 48 h post-injection. An additional group of rats received 80 mg/kg Gelofusine plus 400 mg/kg lysine co-injection. Biodistribution studies were performed both in older (475 g) and younger (300 g) rats, the latter bearing CA20948 tumours. RESULTS: Co-injection of 40 mg/kg Gelofusine resulted in 40-50% reduction of renal uptake and retention of 111In-DOTA,Tyr3-octreotate, whereas higher doses further increased the reduction to 50-60% in both groups of rats. Combining Gelofusine and lysine caused 70% reduction of renal uptake. The uptake of radiolabelled octreotate both in somatostatin receptor-expressing normal tissues and tumours was not affected by Gelofusine co-injection. CONCLUSION: In rats co-injection of 80 mg/kg Gelofusine resulted in maximum reduction of renal retention of 111In-DOTA,Tyr3-octreotate, which was further improved when combined with lysine. Tumour uptake of radiolabelled octreotate was not affected, resulting in an increased tumour to kidney ratio.


Assuntos
Rim/efeitos dos fármacos , Rim/metabolismo , Neoplasias Pancreáticas/patologia , Peptídeos Cíclicos/metabolismo , Poligelina/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Relação Dose-Resposta a Droga , Regulação Neoplásica da Expressão Gênica , Marcação por Isótopo , Masculino , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/metabolismo , Peptídeos Cíclicos/farmacocinética , Ratos , Receptores de Somatostatina/metabolismo , Distribuição Tecidual/efeitos dos fármacos , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X
6.
Bioconjug Chem ; 20(7): 1323-31, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19537708

RESUMO

The somatostatin analogue Tyr(3)-octreotide, which has a high binding affinity for the SSTR2 receptor (somatostatin receptor subtype 2) expressed on tumor cells, is used clinically for the diagnosis and treatment of a variety of neuroendocrine tumors and gastrointestinal disorders. There is growing interest in the development of multivalent peptide systems, because they may have enhanced binding affinity compared to monovalent analogues. In this report, we describe the design and synthesis of a series of Tyr(3)-octreotide-containing monomeric, dimeric, and tetrameric dendrimeric conjugates. These multivalent dendrimeric cyclic peptides were obtained using Cu(I)-catalyzed 1,3-dipolar cycloaddition between peptidyl azides and dendrimeric alkynes. Their affinities for the SSTR2 receptor were determined by a competitive binding assay on rat brain sections.


Assuntos
Alcinos/química , Azidas/química , Dendrímeros/química , Octreotida/análogos & derivados , Octreotida/farmacologia , Receptores de Somatostatina/metabolismo , Alcinos/síntese química , Animais , Azidas/síntese química , Ligação Competitiva , Encéfalo/metabolismo , Catálise , Cobre/química , Ciclização , Dendrímeros/síntese química , Octreotida/síntese química , Ligação Proteica , Ratos
7.
Cancer Biother Radiopharm ; 23(2): 137-57, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18454684

RESUMO

Somatostatin receptor-targeting peptides are widely used for the imaging and therapy of neuroendocrine tumors. Peptide-receptor radionuclide therapy (PRRT) in neuroendocrine tumor patients with radiolabeled somatostatin analogs has resulted in symptomatic improvement, prolonged survival, and enhanced quality of life. The side-effects of PRRT are few and mostly mild, certainly when using kidney protective agents. If a more widespread use of PRRT is possible, such therapy might become the therapy of first choice in patients with metastasized or inoperable neuroendocrine gastroenteropancreatic tumors. Yet, much profit can be gained from improving the receptor-targeting strategies available and developing new strategies. This review presents an overview of several options to optimize receptor-targeted imaging and radionuclide therapy. These include the optimization of peptide analogs, increasing the number of receptors on the tumor site, and combining PRRT with other treatment strategies. The development of new peptide analogs with increased receptor-binding affinity and improved stability might lead to a higher accumulation of radioactivity inside tumor cells. Analogs of somatostatin have been widely studied. However, much profit can be gained in improving peptide analogs targeting other tumor-related receptors, including gastrin-releasing peptide (GRP) receptors, neurotensin (NT) receptors, cholecystokinin (CCK) receptors, and glucagon-like peptide-1 (GLP-1) receptors. Several peptide analogs targeting these receptors are well on their way to clinical utilization. The literature shows that it is possible to increase the receptor density on tumor cells by using different methods, which results in higher binding and internalization rates and thus a higher contrast during peptide-receptor scintigraphy. In PRRT treatment, this would enable the administration of higher therapeutic doses to tumors, which might lead to a higher cure rate in patients. Combinations of radionuclide therapy with other treatment modalities, such as chemotherapy or pretreatment with radiosensitizers, might increase the impact of the treatment. Further, the administration of higher dosages of radioactivity to the patient, enabled by combinations of PRRT with strategies reducing the radiation dose to healthy organs, will improve the outcome of tumor treatment. Also, targeting one or several tumor-specific receptors by using combinations of therapeutic agents, as well as by reducing nontarget uptake of radioactivity, will enlarge the therapeutic window of PRRT. Clinical studies will provide more insight in the effects of combining treatment strategies in cancer patients.


Assuntos
Cintilografia/métodos , Receptores de Peptídeos/metabolismo , Animais , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/radioterapia , Peptídeos/síntese química , Peptídeos/química , Peptídeos/uso terapêutico , Radioisótopos/uso terapêutico , Compostos Radiofarmacêuticos
8.
Nucl Med Biol ; 34(6): 633-41, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17707803

RESUMO

INTRODUCTION: During therapy with radiolabeled peptides, the kidney is most often the critical organ. Newly developed peptides are evaluated preclinically in different animal models before their application in humans. In this study, the renal retention of several radiolabeled peptides was compared in male and female rats and mice. METHODS: After intravenous injection of radiolabeled peptides [somatostatin, cholecystokinin (CCK), minigastrin, bombesin and neurotensin analogues], renal uptake was determined in both male and female Lewis rats and C57Bl mice. In addition, ex vivo autoradiography of renal sections was performed to localize accumulated radioactivity. RESULTS: An equal distribution pattern of renal radioactivity was found for all peptides: high accumulation in the cortex, lower accumulation in the outer medulla and no radioactivity in the inner medulla of the kidneys. In both male rats and mice, an increasing renal uptake was found: [(111)In-DTPA]CCK8<[(111)In-DTPA-Pro(1),Tyr(4)]bombesin approximately [(111)In-DTPA]neurotensin<[(111)In-DTPA]octreotide<<[(111)In-DTPA]MG0. Renal uptake of [(111)In-DTPA]octreotide in rats showed no gender difference, and renal radioactivity was about constant over time. In mice, however, renal uptake in females was significantly higher than that in males and decreased rapidly over time in both genders. Moreover, renal radioactivity in female mice injected with [(111)In-DTPA]octreotide showed a different localization pattern. CONCLUSIONS: Regarding the renal uptake of different radiolabeled peptides, both species showed the same ranking order. Similar to findings in patients, rats showed comparable and constant renal retention of radioactivity in both genders, in contrast to mice. Therefore, rats appear to be the more favorable species for the study of the renal retention of radioactivity.


Assuntos
Bombesina/farmacocinética , Colecistocinina/análogos & derivados , Colecistocinina/farmacocinética , Gastrinas/farmacocinética , Antagonistas de Hormônios/farmacocinética , Rim/diagnóstico por imagem , Rim/metabolismo , Neurotensina/farmacocinética , Somatostatina/farmacocinética , Animais , Autorradiografia , Feminino , Imuno-Histoquímica , Técnicas In Vitro , Radioisótopos de Índio , Marcação por Isótopo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Ácido Pentético/farmacocinética , Cintilografia , Ratos , Ratos Endogâmicos Lew , Caracteres Sexuais , Especificidade da Espécie , Distribuição Tecidual
9.
Bioconjug Chem ; 18(3): 724-30, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17378600

RESUMO

Bombesin is a tetradecapeptide neurohormone that binds to gastrin-releasing peptide receptors (GRPR). GRPRs have been found in a variety of cancers including invasive breast and prostate tumors. The peptide MP2346 (DOTA-(Pro(1),Tyr(4))-bombesin(1-14)) was designed to bind to these GRP receptors. This study was undertaken to evaluate radiolabeled MP2346 as a positron emission tomography (PET) imaging agent. MP2346 was radiolabeled, in high radiochemical purity, with the positron-emitting nuclides (64)Cu (t(1/2) = 12.7 h, beta+ = 19.3%, E(avg) = 278 keV) and (86)Y (t(1/2) = 14.7 h, beta+ = 33%, E(avg) = 664 keV). (64)Cu-MP2346 and (86)Y-MP2346 were studied in vitro for cellular internalization by GRPR-expressing PC-3 (human prostate adenocarcinoma) cells. Both (64)Cu- and (86)Y-MP2346 were studied in vivo for tissue distribution in nude mice with PC-3 tumors. Biodistribution in PC3 tumor-bearing mice demonstrated higher tumor uptake, but lower liver retention, in animals injected with (86)Y-MP2346 compared to (64)Cu-MP2346. Receptor-mediated uptake was confirmed by a significant reduction in uptake in the PC-3 tumor and other receptor-rich tissues by coinjection of a blockade. Small animal PET/CT imaging was carried out in mice bearing PC-3 tumors and rats bearing AR42J tumors. It was possible to delineate PC-3 tumors in vivo with (64)Cu-MP2346, but superior (86)Y-MP2346-PET images were obtained due to lower uptake in clearance organs and lower background activity. The (86)Y analogue demonstrated excellent PET image quality in models of prostate cancer for the delineation of the GRPR-rich tumors and warrants further investigation.


Assuntos
Bombesina/análogos & derivados , Compostos Organometálicos/farmacocinética , Tomografia por Emissão de Pósitrons , Neoplasias da Próstata/diagnóstico por imagem , Compostos Radiofarmacêuticos/farmacocinética , Receptores da Bombesina/análise , Animais , Bombesina/análise , Bombesina/farmacocinética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Compostos Organometálicos/análise , Neoplasias da Próstata/química , Neoplasias da Próstata/metabolismo , Compostos Radiofarmacêuticos/análise , Ratos , Ratos Endogâmicos , Receptores da Bombesina/metabolismo , Distribuição Tecidual
10.
J Nucl Med ; 48(1): 88-93, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17204703

RESUMO

UNLABELLED: Human prostate cancers (PC) overexpress gastrin-releasing peptide (GRP) receptors. This observation suggests that GRP receptors may be used as new visualization and treatment modalities for these tumors. Radiolabeled GRP receptor-targeting analogs of GRP and bombesin (BN) have successfully been developed for these purposes. Expression of GRP receptors in human prostate tumors is, however, primarily evaluated in early stages of tumor development and information on expression in the more progressive prostate tumors is uncertain. To evaluate GRP receptor expression in all stages of PC, we investigated GRP receptor expression using a panel of 12 established human PC xenograft models representing the different stages of human PC and the effect of antiandrogen treatment (castration). METHODS: Human PC xenografts were grown in male nude mice, and GRP receptor density in the tumors was evaluated using displacement receptor autoradiography with the universal BN receptor analog (125)I-[D-Tyr(6),beta-Ala(11),Phe(13),Nle(14)]BN(6-14) and the BN analog (111)In-[DTPA-Pro(1),Tyr(4)]BN (DTPA is diethylenetriaminepentaacetic acid) before and after castration. RESULTS: Autoradiography showed high-density GRP receptor expression in the androgen-dependent tumors (3/12 models), whereas only very low receptor expression was found in the androgen-responsive and -independent tumors (9/12 models). Castration resulted in GRP receptor downregulation (11%-36% of initial values) in the 3 androgen-dependent tumors. CONCLUSION: High GRP receptor density was only observed in androgen-dependent PC xenografts, indicating high GRP receptor expression in the early, androgen-dependent, stages of prostate tumor development and not in later stages. In addition, castration strongly reduced GRP receptor expression in androgen-dependent tumors, indicating that GRP receptor expression in human PC is androgen-regulated.


Assuntos
Androgênios/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/metabolismo , Animais , Autorradiografia , Bombesina/farmacologia , Castração , Linhagem Celular Tumoral , Peptídeo Liberador de Gastrina/química , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Peptídeos/química , Ligação Proteica
11.
Eur J Nucl Med Mol Imaging ; 34(2): 219-27, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17021812

RESUMO

PURPOSE AND METHODS: Studies on peptide receptor radionuclide therapy (PRRT) using radiolabelled somatostatin analogues have shown promising results with regard to tumour control. The efficacy of PRRT is limited by uptake and retention in the proximal tubules of the kidney, which might lead to radiation nephropathy. We investigated the long-term renal toxicity after different doses of [(177)Lu-DOTA(0),Tyr(3)]octreotate and the effects of dose fractionation and lysine co-injection in two tumour-bearing rat models. RESULTS: Significant renal toxicity was detected beyond 100 days after start of treatment as shown by elevated serum creatinine and proteinuria. Microscopically, tubules were strongly dilated with flat epithelium, containing protein cylinders. Creatinine levels rose significantly after 555 MBq [(177)Lu-DOTA(0),Tyr(3)]octreotate, but were significantly lower after 278 MBq (single injection) or two weekly doses of 278 MBq. Renal damage scores were maximal after 555 MBq and significantly lower in the 278 and 2x278 MBq groups. Three doses of 185 MBq [(177)Lu-DOTA(0),Tyr(3)]octreotate with intervals of a day, a week or a month significantly influenced serum creatinine (469+/-18, 134+/-70 and 65+/-15 micromol/l, respectively; p<0.001). Renal histological damage scores were not significantly influenced by dose fractionation. Lysine co-administration with three weekly treatments of 185 MBq significantly lowered serum creatinine and proteinuria. CONCLUSION: Injection of high doses of [(177)Lu-DOTA(0),Tyr(3)]octreotate resulted in severe renal damage in rats as indicated by proteinuria, elevated serum creatinine and histological damage. This damage was dose dependent and became overt between 100 and 200 days after treatment. Dose fractionation had significant beneficial effects on kidney function. Also, lysine co-injection successfully prevented functional damage.


Assuntos
Doenças da Medula Óssea/etiologia , Medula Óssea/efeitos da radiação , Nefropatias/etiologia , Rim/efeitos da radiação , Octreotida/análogos & derivados , Compostos Organometálicos/toxicidade , Lesões por Radiação/etiologia , Animais , Doenças da Medula Óssea/diagnóstico , Relação Dose-Resposta a Droga , Nefropatias/diagnóstico , Dose Letal Mediana , Estudos Longitudinais , Masculino , Octreotida/toxicidade , Lesões por Radiação/diagnóstico , Compostos Radiofarmacêuticos/toxicidade , Ratos , Ratos Endogâmicos Lew
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...