Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Biother Radiopharm ; 29(9): 359-67, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25286347

RESUMEN

INTRODUCTION: Gastrin-releasing peptide receptors (GRPR) and GRP-derived analogs have attracted attention due to high receptor expression in frequently occurring human neoplasia. The authors recently synthesized a series of GRPR-affine peptide analogs based on the 27-mer GRP and derivatized with the DOTA chelator at the N-terminus for (111)In-labeling. In this study, the authors evaluated the most promising from these series, DOTA-GRP(13-27), after radiolabeling with (177)Lu for future therapeutic applications. In addition, to improve in vivo stability of the peptide against in vivo degradation by the protease neutral endopeptidase (NEP), the authors coinjected [(177)Lu]DOTA-GRP(13-27) with the potent NEP inhibitor phosphoramidon (PA). The authors also aimed at reducing renal uptake by coadministration of lysine. METHODS: In vivo stability studies were performed in Swiss albino mice. Biodistribution studies were conducted in NMRI nu/nu mice bearing prostate cancer (PC)-3 xenografts. Ex vivo autoradiography was performed using frozen sections from PC-3 xenografts and kidneys. RESULTS AND DISCUSSION: Coadministration of PA significantly increased the percentage of intact radiopeptide in the mouse circulation. From biodistribution and ex vivo autoradiography studies, coadministration of both lysine and PA with [(177)Lu]DOTA-GRP(13-27) appeared to induce a clear improvement of tumor uptake as well as lower levels of renal radioactivity, causing a promising ninefold increase in tumor/kidney ratios.


Asunto(s)
Glicopéptidos/farmacología , Neprilisina/antagonistas & inhibidores , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/metabolismo , Radiofármacos/farmacocinética , Receptores de Bombesina/biosíntesis , Animales , Línea Celular Tumoral , Péptido Liberador de Gastrina/farmacocinética , Xenoinjertos , Humanos , Lutecio/química , Masculino , Ratones , Ratones Desnudos , Neprilisina/metabolismo , Fragmentos de Péptidos/farmacocinética , Neoplasias de la Próstata/enzimología , Inhibidores de Proteasas/farmacología , Radioisótopos/química , Cintigrafía , Receptores de Bombesina/metabolismo , Distribución Tisular
2.
EJNMMI Res ; 4(1): 47, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26116111

RESUMEN

BACKGROUND: Targeted radionuclide therapy with high-energy beta-emitters is generally considered suboptimal to cure small tumours (<300 mg). Tumour targeting of the CCK2 receptor-binding minigastrin analogue PP-F11 was determined in a tumour-bearing mouse model at increasing peptide amounts. The optimal therapy was analysed for PP-F11 labelled with (90)Y, (177)Lu or (213)Bi, accounting for the radionuclide specific activities (SAs), the tumour absorbed doses and tumour (radio) biology. METHODS: Tumour uptake of (111)In-PP-F11 was determined in nude mice bearing CCK2 receptor-transfected A431 xenografts at 1 and 4 h post-injection for escalating peptide masses of 0.03 to 15 nmol/mouse. The absorbed tumour dose was estimated, assuming comparable biodistributions of the (90)Y, (177)Lu or (213)Bi radiolabelled peptides. The linear-quadratic (LQ) model was used to calculate the tumour control probabilities (TCP) as a function of tumour mass and growth. RESULTS: Practically achievable maximum SAs for PP-F11 labelled with (90)Y and (177)Lu were 400 MBq (90)Y/nmol and 120 MBq(177)Lu/nmol. Both the large elution volume from the 220 MBq (225)Ac generator used and reaction kinetics diminished the maximum achieved (213)Bi SA in practice: 40 MBq (213)Bi/nmol. Tumour uptakes decreased rapidly with increasing peptide amounts, following a logarithmic curve with ED50 = 0.5 nmol. At 0.03 nmol peptide, the (300 mg) tumour dose was 9 Gy after 12 MBq (90)Y-PP-F11, and for (111)In and (177)Lu, this was 1 Gy. A curative dose of 60 Gy could be achieved with a single administration of 111 MBq (90)Y labelled to 0.28 nmol PP-F11 or with 4 × 17 MBq (213)Bi (0.41 nmol) when its α-radiation relative biological effectiveness (RBE) was assumed to be 3.4. Repeated dosing is preferable to avoid complete tumour receptor saturation. Tumours larger than 200 mg are curable with (90)Y-PP-F11; the other radionuclides perform better in smaller tumours. Furthermore, (177)Lu is not optimal for curing fast-growing tumours. CONCLUSIONS: Receptor saturation, specific radiopharmaceutical activities and absorbed doses in the tumour together favour therapy with the CCK2 receptor-binding peptide PP-F11 labelled with (90)Y, despite its longer ß-particle range in tissue, certainly for tumours larger than 300 mg. The predicted TCPs are of theoretical nature and need to be compared with the outcome of targeted radionuclide experiments.

3.
Eur J Nucl Med Mol Imaging ; 38(8): 1426-35, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21528387

RESUMEN

PURPOSE: Stability of radiolabelled cholecystokinin 2 (CCK2) receptor targeting peptides has been a major limitation in the use of such radiopharmaceuticals especially for targeted radionuclide therapy applications, e.g. for treatment of medullary thyroid carcinoma (MTC). The purpose of this study was to compare the in vitro stability of a series of peptides binding to the CCK2 receptor [selected as part of the COST Action on Targeted Radionuclide Therapy (BM0607)] and to identify major cleavage sites. METHODS: Twelve different 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA)-minigastrin/CCK conjugates were provided within an European COST Action (BM0607) by different laboratories and radiolabelled with (177)Lu. Their in vitro stabilities were tested in fresh human serum. Radiochemical yields (RCY) and intact radioligands for half-life calculations were determined by radio-HPLC. Matrix-assisted laser desorption/ionisation time-of-flight mass spectrometry (MALDI-TOF MS) analysis of metabolites was performed to identify cleavage products using conjugates labelled with excess stable (nat)Lu, incubated in serum at 37°C. Urine metabolite analysis after injection in normal mice was performed by radio-HPLC analysis. RESULTS: Variable stability in human serum was found for the different peptides with calculated half-lives between 4.5 ± 0.1 h and 198 ± 0.1 h (n = 2). In urine of normal mice only metabolised peptide fragments were detected even at short times after injection for all peptides. MALDI-TOF MS revealed a major cleavage site of all minigastrin derivatives between Asp and Phe-NH(2) at the C-terminal end. CONCLUSION: Development of CCK2 receptor ligands especially for therapeutic purposes in patients with MTC or small cell lung cancer (SCLC) is still ongoing in different laboratories. This comparative study provided valuable insight into the importance of biological stability especially in the context of other results of this comparative trial within the COST Action BM0607.


Asunto(s)
Conducta Cooperativa , Péptidos/metabolismo , Receptor de Colecistoquinina B/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Femenino , Humanos , Metionina/metabolismo , Ratones , Datos de Secuencia Molecular , Oxidación-Reducción , Péptidos/sangre , Péptidos/química , Unión Proteica , Estabilidad Proteica
4.
Eur J Nucl Med Mol Imaging ; 37(7): 1386-96, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20182713

RESUMEN

PURPOSE: Prostate-specific antigen (PSA)-based screening for prostate cancer (PC) has dramatically increased early diagnosis. Current imaging techniques are not optimal to stage early PC adequately. A promising alternative to PC imaging is peptide-based scintigraphy using radiolabelled bombesin (BN) analogues that bind to gastrin-releasing peptide receptors (GRPR) being overexpressed in PC. When labelled to appropriate radionuclides BN targeting of GRPRs may also provide applications for peptide radionuclide receptor therapy (PRRT). Assessment studies under identical experimental conditions allowing a reliable comparison of the potential of such analogues are lacking. This study was performed to evaluate and directly compare five promising radiolabelled BN analogues for their targeting efficacy for PC under standardised conditions. METHODS: The BN agonists [(111)In]DOTA-PESIN, [(111)In]AMBA, [(111)In]MP2346 and [(111)In]MP2653 and one antagonist [(99m)Tc]Demobesin-1 were evaluated in GRPR-overexpressing human PC-3 tumour-bearing mice to determine peptide stability in vivo, biodistribution and GRPR targeting potential by animal SPECT/CT imaging and ex vivo autoradiography. RESULTS: HPLC analysis of blood showed intact Demobesin-1 at 5 and 15 min after injection (64.1 +/- 1.6% and 41.0 +/- 01%, respectively) being much less for the other compounds. AMBA, the second most stable analogue, showed 36.1 +/- 2.7% and 9.8 +/- 1.1% intact peptide after 5 and 15 min. PC-3 tumour uptake at 1 h was comparable for Demobesin-1, AMBA, PESIN and MP2346 (3.0 +/- 0.4, 2.7 +/- 0.5, 2.3 +/- 0.5 and 2.1 +/- 0.9%ID/g, respectively), but very low for MP2653 (0.9 +/- 0.2%ID/g). In addition, MP2346 showed undesirably high uptake in the kidneys (7.9 +/- 1.9%ID/g) being significantly less for the other analogues. AMBA, MP2346 and PESIN revealed favourable increases in tumour to blood ratios over time while changes in tumour to kidney and pancreas ratios for Demobesin-1 from 1 to 24 h after injection were significantly better than for the other analogues. All analogues visualised PC-3 tumours by SPECT/CT and autoradiography. CONCLUSION: In the present study the BN antagonist Demobesin-1 was the best performing analogue showing superior in vivo stability, highest tumour uptake and retention while pancreatic and renal clearance were rapid. PESIN and AMBA were the best GRP agonists with sufficient in vivo stabilities as well as high tumour uptake and retention. Based on these results all three analogues deserve further evaluation for clinical use in PC patients.


Asunto(s)
Bombesina/análogos & derivados , Bombesina/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Autorradiografía , Bombesina/farmacocinética , Línea Celular Tumoral , Estabilidad de Medicamentos , Estudios de Factibilidad , Humanos , Masculino , Ratones , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/diagnóstico por imagen , Control de Calidad , Coloración y Etiquetado , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X
5.
Int J Cancer ; 126(12): 2826-34, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19876914

RESUMEN

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.


Asunto(s)
Andrógenos/farmacología , Bombesina/farmacocinética , Péptido Liberador de Gastrina/metabolismo , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores de Bombesina/metabolismo , Animales , Autorradiografía , Bombesina/análogos & derivados , Castración , Péptido Liberador de Gastrina/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Radioisótopos de Indio , Masculino , Tasa de Depuración Metabólica , Ratones , Ratones Desnudos , Neoplasias Hormono-Dependientes/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Radiofármacos/farmacocinética , Receptores de Bombesina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testosterona/administración & dosificación , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Acc Chem Res ; 42(7): 873-80, 2009 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-19445476

RESUMEN

Molecular imaging plays an essential role in balancing the clinical benefits and risks of radionuclide-based cancer therapy. To effectively treat individual patients, careful assessment of biodistribution, dosimetry, and toxicity is essential. In this Account, we describe advances that combine features of molecular imaging and radionuclide therapy to provide new avenues toward individualized cancer treatment. Selective receptor-targeting radiopeptides have emerged as an important class of radiopharmaceuticals for molecular imaging and therapy of tumors that overexpress peptide receptors on the cell membrane. After such peptides labeled with gamma-emitting radionuclides bind to their receptors, they allow clinicians to visualize receptor-expressing tumors non-invasively. Peptides labeled with beta-particle emitters could also eradicate receptor-expressing tumors. The somatostatin receptors, which are overexpressed in a majority of neuroendocrine tumors, represent the first and best example of targets for radiopeptide-based imaging and radionuclide therapy. The somatostatin analogue (111)In-octreotide permits the localization and staging of neuroendocrine tumors that express the appropriate somatostatin receptors. Newer modified somatostatin analogues, including Tyr(3)-octreotide and Tyr(3)-octreotate, are successfully being used for tumor imaging and radionuclide therapy. Because there are few effective therapies for patients with inoperable or metastasized neuroendocrine tumors, this therapy is a promising novel treatment option for these patients. Peptide receptor imaging and radionuclide therapy can be combined in a single probe, called a "theranostic". To select patients who are likely to benefit from this type of intervention, we first use a peptide analogue labeled with a diagnostic radionuclide to obtain a scan. Selected patients will be treated using the same or a similar peptide analogue labeled with a therapeutic radionuclide. The development of such theranostics could greatly advance the development of personalized treatments. Apart from patient selection for radionuclide therapy, other imaging applications of targeted radiopeptides include localization of primary tumors, detection of metastatic disease (staging/restaging), dosimetry (prediction of response and radiotoxicity), monitoring effects of surgery, radio(nuclide)therapy or chemotherapy, and detection of progression of disease or relapse (follow up). For further evaluation of tumor receptor expression and to increase the value of cancer targeting using radiopeptides, researchers have introduced and evaluated different radiolabeled analogues of other peptide families, such as cholecystokinin (CCK), gastrin, bombesin, substance P, vasoactive intestinal peptide (VIP), and neuropeptide (NP)-Y analogues. We expect improvements in the development of new peptide analogues: such advances could reduce side effects and allow for the use of combination therapy (for example, combining radiopeptide analogues with chemotherapeutics).


Asunto(s)
Tumores Neuroendocrinos/diagnóstico por imagen , Tumores Neuroendocrinos/radioterapia , Octreótido/análogos & derivados , Radiofármacos/uso terapéutico , Somatostatina/análogos & derivados , Secuencia de Aminoácidos , Animales , Humanos , Octreótido/uso terapéutico , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/radioterapia , Péptidos/química , Tomografía de Emisión de Positrones , Ratas , Receptores de Somatostatina/metabolismo , Somatostatina/uso terapéutico , Tomografía Computarizada de Emisión de Fotón Único
7.
Eur J Nucl Med Mol Imaging ; 36(8): 1265-72, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19266197

RESUMEN

PURPOSE: Cholecystokinin 2 (CCK-2) receptor overexpression has been demonstrated in a high percentage of medullary thyroid carcinomas (MTC). Analogous to somatostatin receptors, CCK-2 receptors might be viable targets for radionuclide scintigraphy and/or radionuclide therapy. Several CCK-2 receptor-binding radiopeptides have been developed, and some have been carried through into clinical studies. However, these studies are mostly limited and difficult to compare. The aim of this study was to evaluate the diagnostic and therapeutic potential of three promising CCK-2 receptor-binding radiopeptides in patients with MTC. METHODS: (111)In-DOTA-(D: )Asp-Tyr-Nle-Gly-Trp-Nle-Asp-Phe-NH(2) ((111)In-DOTA-CCK), a CCK analogue, and the gastrin-based ligands (99m)Tc-N(4)-Gly-(D: )Glu-(Glu)(5)-Ala-Tyr-Gly-Trp-Met-Asp-Phe-NH(2) ((99m)Tc-demogastrin 2) and (111)In-DOTA-(D: )Glu-Ala-Tyr-Gly-Trp-Met-Asp-Phe-NH(2) ((111)In-DOTA-MG11) were each administered to the same group of six patients. Planar images made at 3-5, 7 and 24 h p.i. were used for comparison of tumour visualisation and renal uptake. RESULTS: (99m)Tc-demogastrin 2 scintigraphy visualised all known lesions and new lesions in four of six patients. (111)In-DOTA-CCK and (111)In-DOTA-MG11 on the other hand missed several lesions; tumour uptake of these two radiopharmaceuticals was quite low. Comparison of retention of renal activity showed no major differences between the three radiopeptides. CONCLUSION: (99m)Tc-demogastrin 2 scintigraphy appeared most promising as a diagnostic tool in patients with MTC. Further studies are required to evaluate its value in patient management. Direct comparisons of the compounds studied strongly suggests that (111)In-DOTA-CCK and (111)In-DOTA-MG11 have less potential as imaging agents than (99m)Tc-demogastrin 2. These DOTA-linked compounds are considered unlikely to be useful for radionuclide therapy because of low tumour uptake.


Asunto(s)
Neoplasias del Tronco Encefálico/diagnóstico por imagen , Neoplasias del Tronco Encefálico/metabolismo , Oligopéptidos/metabolismo , Receptor de Colecistoquinina B/metabolismo , Neoplasias de la Tiroides/diagnóstico por imagen , Neoplasias de la Tiroides/metabolismo , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Femenino , Humanos , Radioisótopos de Indio , Marcaje Isotópico , Masculino , Persona de Mediana Edad , Oligopéptidos/química , Oligopéptidos/farmacocinética , Radiactividad , Cintigrafía , Bazo/metabolismo , Distribución Tisular
8.
Eur J Nucl Med Mol Imaging ; 33(7): 831-40, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16568203

RESUMEN

PURPOSE: The aim of this study was to evaluate [(99m)Tc]Demotate 2 ([(99m)Tc-N(4) (0-1),Asp(0),Tyr(3)]octreotate) as a candidate for in vivo imaging of sst(2)-positive tumours and to compare it with [(111)In]DOTA-tate ([(111)In-DOTA(0),Tyr(3)]octreotate). METHODS: Labelling of Demotate 2 with (99m)Tc was performed at room temperature using SnCl(2) as reductant in the presence of citrate at alkaline pH. Radiochemical analysis involved ITLC and HPLC methods. Peptide conjugate affinities for sst(2) were determined by receptor autoradiography on rat brain cortex sections using [DOTA(0),(125)I-Tyr(3)]octreotate as the radioligand. The affinity profile of Demotate 2 for human sst(1)-sst(5) was studied by receptor autoradiography in cell preparations using the universal somatostatin radioligand [(125)I][Leu(8),(D: )Trp(22),Tyr(25)]somatostatin-28. The internalisation rates of [(99m)Tc]Demotate 2 and [(111)In]DOTA-tate were compared in sst(2)-positive and -negative control cell lines. Biodistribution of radiopeptides was studied in male Lewis rats bearing CA20948 tumours. RESULTS: Peptide conjugates showed selectivity and a high affinity binding for sst(2) (Demotate 2 IC(50)=3.2 nM and DOTA-tate IC(50)=5.4 nM). [(99m)Tc]Demotate 2, like [(111)In]DOTA-tate, internalised rapidly in all sst(2)-positive cells tested, but not in sst(2)-negative control cells. After injection in CA20948 tumour-bearing rats both radiopeptides showed high and specific uptake in the sst(2)-positive organs and in the implanted tumour and rapid excretion from non-target tissues via the kidneys. CONCLUSION: [(99m)Tc]Demotate 2, similarly to the known sst(2)-targeting agent [(111)In]DOTA-tate, showed promising biological qualities for application in the scintigraphy of sst(2)-positive tumours.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas de Neoplasias/metabolismo , Octreótido/análogos & derivados , Compuestos Organometálicos/farmacocinética , Compuestos de Organotecnecio/farmacocinética , Neoplasias Pancreáticas/metabolismo , Péptidos Cíclicos/farmacocinética , Receptores de Somatostatina/metabolismo , Animales , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Masculino , Tasa de Depuración Metabólica , Octreótido/farmacocinética , Especificidad de Órganos , Neoplasias Pancreáticas/diagnóstico por imagen , Cintigrafía , Radiofármacos/farmacocinética , Ratas , Ratas Endogámicas Lew , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Distribución Tisular
9.
J Nucl Med ; 47(1): 122-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16391196

RESUMEN

UNLABELLED: Tumor-induced angiogenesis can be targeted by RGD (Arg-Gly-Asp) peptides, which bind to alpha(v)beta(3)-receptors upregulated on angiogenic endothelial cells. RGD-containing peptides are capable of inducing apoptosis through direct activation of procaspase-3 to caspase-3 in cells. Additionally, tumor cells overexpressing somatostatin receptors can be targeted by somatostatin analogs. Radiolabeled somatostatin analogs are successfully used to image and treat such tumors via receptor-targeted scintigraphy and therapy. We combined these 2 peptides, RGD and somatostatin, to synthesize a new hybrid peptide, RGD-diethylenetriaminepentaacetic acid (DTPA)-octreotate (c(Arg-Gly-Asp-D-Tyr-Asp)-Lys(DTPA)-D-Phe-c(Cys-Tyr-D-Trp-Lys-Thr-Cys)-Thr). An earlier study showed that tumor-bearing rats had high receptor-specific uptake of RGD-(111)In-DTPA-octreotate in somatostatin receptor subtype 2-positive tissues and tumors. Furthermore, RGD-(111)In-DTPA-octreotate showed a pronounced tumoricidal effect, which is probably the result of increased apoptosis, as is shown by an increased caspase-3 activity after incubation with (111)In-labeled RGD-DTPA-octreotate in comparison with the 2 monopeptides (111)In-DTPA-RGD and (111)In-DTPA-Tyr(3)-octreotate. In this study, we evaluated the biodistributions of RGD-(111)In-DTPA-octreotate and (125)I-RGD-octreotate and investigated the caspase-3 activation of the unlabeled compound RGD-DTPA-octreotate in vitro. METHODS: Biodistribution studies on tumor-bearing rats were performed with RGD-(111)In-DTPA-octreotate and (125)I-RGD-octreotate. The apoptotic activity, by activation of caspase-3 with RGD-DTPA-octreotate and RGD-octreotate, was examined using colorimetric and immunocytochemical assays. RESULTS: The radiolabeled compound, RGD-(111)In-DTPA-octreotate, showed high uptake and retention in the rats in which rat pancreatic CA20948 tumor had been implanted. A major drawback was high renal uptake. In vitro, the unlabeled peptide RGD-DTPA-octreotate induced a significant increase in caspase-3 levels in various cell lines in comparison with RGD and Tyr(3)-octreotate (P < 0.01). Caspase-3 activation was time dependent. To alter the elimination route, we examined the biodistribution of radioiodinated RGD-octreotate without DTPA [c(Arg-Gly-Asp-D-Tyr-Asp)-D-Phe-c(Cys-Tyr-D-Trp-Lys-Thr-Cys)-Thr], as a model of unlabeled RGD-octreotate, in tumor-bearing rats. (125)I-RGD-octreotate showed a much lower renal uptake than did RGD-(111)In-DTPA-octreotate. Furthermore, the affinity of RGD-octreotate increased in comparison with RGD-DTPA-octreotate (values of 1.4 x 10(-8) mol/L vs. 9.4 x 10(-8) mol/L, respectively, for inhibitory concentration of 50%). Finally, RGD-octreotate was still able to activate caspase-3, as was indicated with immunocytochemistry. CONCLUSION: Because of the high renal uptake, RGD-(111)In-DTPA-octreotate is unsuitable for radionuclide therapy. However, the unlabeled peptides, RGD-DTPA-octreotate and RGD-octreotate, also induced an increase in caspase-3 levels, indicating the therapeutic potential of this compound. Thus, the development of hybrid molecules can become a new approach in the treatment of cancer.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/farmacocinética , Proteínas Reguladoras de la Apoptosis/uso terapéutico , Oligopéptidos/farmacocinética , Oligopéptidos/uso terapéutico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/radioterapia , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Radioisótopos de Indio/farmacocinética , Radioisótopos de Indio/uso terapéutico , Masculino , Tasa de Depuración Metabólica , Especificidad de Órganos , Neoplasias Pancreáticas/patología , Radioinmunoterapia/métodos , Radiofármacos/farmacocinética , Ratas , Ratas Endogámicas Lew , Distribución Tisular , Resultado del Tratamiento
10.
J Nucl Med ; 46 Suppl 1: 13S-7S, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15653647

RESUMEN

UNLABELLED: Peptide receptor-targeted radionuclide therapy of somatostatin receptor-expressing tumors is a promising application of radiolabeled somatostatin analogs. Suitable radionuclides are (90)Y, a pure, high-energy beta-emitter (2.27 MeV), and (177)Lu, a medium-energy beta-emitter (0.5 MeV) with a low-abundance gamma. METHODS: Lewis rats, each bearing both a small (approximately 0.5 cm(2)) and a large (7-9 cm(2)) somatostatin receptor-positive rat pancreatic CA20948 tumor in their flanks, were used. We investigated the radiotherapeutic effects of [(90)Y-tetraazacyclododecanetetraacetic acid (DOTA),Tyr(3)]octreotide, [(90)Y-DOTA,Tyr(3)]octreotate, [(177)Lu-DOTA,Tyr(3)]octreotate, and the combination of (90)Y- and (177)Lu-labeled analogs at the same tumor radiation dose (60 Gy). RESULTS: Radiotherapeutic effects of the (90)Y- and (177)Lu-labeled analogs were found in the rat tumor model. In these animals bearing tumors of different sizes, the antitumor effects of the combination of 50% (177)Lu- plus 50% (90)Y-analogs were superior to those in animals treated with either (90)Y- or (177)Lu- analog alone. In smaller tumors, the (90)Y radiation energy was not completely absorbed in the tumor, whereas in larger tumors the increased number of clonogenic tumor cells at the fixed level of absorbed dose may account for the failure of (177)Lu alone to go completely into remission. CONCLUSION: This study shows the superior antitumor effects of the combination of (177)Lu- and (90)Y-somatostatin analogs when compared with either (90)Y- or (177)Lu-analog alone in animals bearing tumors of various sizes.


Asunto(s)
Lutecio/administración & dosificación , Octreótido/análogos & derivados , Octreótido/administración & dosificación , Compuestos Organometálicos/administración & dosificación , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/radioterapia , Radioisótopos/administración & dosificación , Somatostatina/análogos & derivados , Itrio/administración & dosificación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Línea Celular Tumoral , Humanos , Masculino , Radiofármacos/uso terapéutico , Ratas , Ratas Endogámicas Lew , Somatostatina/administración & dosificación , Análisis de Supervivencia , Resultado del Tratamiento
11.
J Nucl Med ; 45(10): 1716-20, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15471839

RESUMEN

UNLABELLED: Receptor-targeted scintigraphy and radionuclide therapy with radiolabeled somatostatin analogs are successfully applied for somatostatin receptor-positive tumors. The synergistic effects of an apoptosis-inducing factor, for example, the Arg-Gly-Asp (RGD) motif, can increase the radiotherapeutic efficacy of these peptides. Hence, the tumoricidal effects of the hybrid peptide RGD-diethylaminetriaminepentaacetic acid (DTPA)-Tyr3-octreotate (cyclic[c](Arg-Gly-Asp-D-Tyr-Asp)-Lys(DTPA)-D-Phe-c(Cys-Tyr-D-Trp-Lys-Thr-Cys)-Thr), hereafter referred to as RGD-DTPA-octreotate, were evaluated in comparison with those of RGD (c(Arg-Gly-Asp-D-Tyr-Asp)) and Tyr3-octreotate (D-Phe-c(Cys-Tyr-D-Trp-Lys-Thr-Cys)-Thr). METHODS: The therapeutic effects of RGD-111In-DTPA-octreotate, 111In-DTPA-RGD, and 111In-DTPA-Tyr3-octreotate were investigated with various cell lines by use of a colony-forming assay, and caspase-3 activity was also determined. RESULTS: Tumoricidal effects were found with 111In-DTPA-RGD, 111In-DTPA-Tyr3-octreotate, and RGD-111In-DTPA-octreotate, in order from least effective to most effective. Also, the largest increase in caspase-3 levels was found with RGD-111In-DTPA-octreotate. CONCLUSION: RGD-111In-DTPA-octreotate has more pronounced tumoricidal effects than 111In-DTPA-RGD and 111In-DTPA-Tyr3-octreotate, because of increased apoptosis, as indicated by increased caspase-3 activity.


Asunto(s)
Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Radioisótopos de Indio/administración & dosificación , Oligopéptidos/administración & dosificación , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/radioterapia , Péptidos Cíclicos/administración & dosificación , Animales , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Evaluación Preclínica de Medicamentos , Radioinmunoterapia/métodos , Ratas , Resultado del Tratamiento
12.
Cancer Biother Radiopharm ; 18(5): 761-8, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14629824

RESUMEN

Somatostatin analogs promising for peptide receptor scintigraphy (PRS) and peptide receptor radionuclide therapy (PRRT) are D-Phe-c(Cys-Tyr-D-Trp-Lys-Thr-Cys)-Thr(ol) (Tyr 3-octreotide) and D-Phe-c(Cys-Tyr-D-Trp-Lys-Thr-Cys)-Thr (tyr3-octreotate). For radiotherapeutic applications these peptides are being labeled with the beta(-) particle emitters 177Lu or 90Y. We evaluated the therapeutic effects of these analogs chelated with tetra-azacyclododecatatro-acetic acid (DOTA) and labeled with 90Y or 177Lu in an in vitro colony-forming assay using the rat pancreatic tumor cell line CA20948. Furthermore, we investigated the effects of incubation time, radiation dose, and specific activity of [177Lu-DOTA]-D-Phe1-c (Cys-Tyr-D-Trp-Lys-Thr-Cys)-Thr (177Lu-octreotate). 177Lu-octreotate could reduce tumor growth to 100% cell kill and effects were dependent on radiation dose, incubation time, and specific activity used. Similar concentrations of 177Lu-DOTA, which is not bound to the cells, had a less pronounced effect on the tumor cell survival. Both tyr3-octreotide and tyr3-octreotate labeled with either 177Lu or 90Y, using DOTA as chelator, were able to control tumor growth in a dose-dependent manner. In all concentrations used radiolabeled tyr3-octreotate had a higher tumor kill compared to radiolabeled tyr3-octreotide, labeled with 177Lu or 90Y. This is in accordance with the higher affinity of tyr3-octreotate for the subtype 2 (sst2)-receptor compared to tyr3-octreotide, leading to a higher amount of cell-associated radioactivity, resulting in a significantly higher tumor radiation dose. In conclusion, tyr3-octreotate labeled with 177Lu or 90Y is the most promising analog for PRRT.


Asunto(s)
Octreótido/análogos & derivados , Octreótido/farmacología , Péptidos Cíclicos/farmacología , Receptores de Péptidos/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Lutecio , Péptidos Cíclicos/metabolismo , Radioisótopos , Ratas , Itrio
13.
J Nucl Med ; 44(8): 1315-21, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12902423

RESUMEN

UNLABELLED: Human somatostatin (SS) receptor (sst)-positive tumors can be visualized by gamma camera scintigraphy after the injection of [(111)In-diethylenetriaminepentaacetic acid (DTPA)-D-Phe(1)] octreotide. Uptake of [(111)In-DTPA-D-Phe(1)]octreotide is dependent on sst-mediated internalization of the radioligand by the tumor cells. Human sst-positive tumors frequently express multiple sst subtypes. In vitro studies have demonstrated that the 5 sst subtypes (sst(1-5)) differentially internalize sst-bound ligand. The present study was performed to evaluate the role of sst(2) in vivo in determining the uptake of [(111)In-DTPA-D-Phe(1)]octreotide, as well as of the more "universal" ligand [(111)In-DTPA]SS-14, by sst-positive organs expressing multiple sst subtypes. METHODS: Wild-type and sst(2) knockout mice (n = 4 per treatment group) were injected intravenously with 1 MBq (0.1 micro g) [(111)In-DTPA-D-Phe(1)]octreotide or [(111)In-DTPA]SS-14. After 24 h, the animals were sacrificed and radioactivity in the organs under investigation was determined. In addition, the sst subtype messenger RNA (mRNA) expression pattern in these organs was determined by reverse transcriptase polymerase chain reaction (RT-PCR) analysis. RESULTS: RT-PCR analysis demonstrated the presence of all 5 sst subtype mRNAs in the adrenals and pituitary of wild-type mice but no sst(2) in the knockout mice. The thymus expressed mRNA for sst(2) and sst(4) mRNA in wild-type mice, whereas no sst(2) was detected in knockout mice. In wild-type mice, the in vivo uptake values (in percentage injected dose per gram of tissue) of [(111)In-DTPA-D-Phe(1)]octreotide for the pituitary, adrenals, pancreas, and thymus amounted to 1.2 +/- 0.2, 0.26 +/- 0.03, 0.18 +/- 0.03, and 0.30 +/- 0.05, respectively, in wild-type mice. Compared with wild-type mice, sst(2) knockout mice had dramatically lower uptake values in these organs-lower by 97%, 83%, 96%, and 94%, respectively (P < 0.01 vs. wild type). Comparable differences in the uptake of radioactivity between wild-type and knockout mice were found using [(111)In-DTPA]SS-14 as the radiotracer. Interestingly, in some organs expressing sst(2) mRNA (liver, muscle, and peripheral blood mononuclear cells), no specific binding of [(111)In-DTPA-D-Phe(1)]octreotide or [(111)In-DTPA]SS-14 to sst in vivo was found, suggesting that the sst(2) protein expression level was very low in these tissues. CONCLUSION: The uptake of [(111)In-DTPA-D-Phe(1)]octreotide and [(111)In-DTPA]SS-14 in sst-positive organs is determined predominantly by sst(2).


Asunto(s)
Octreótido/análogos & derivados , Octreótido/farmacocinética , Especificidad de Órganos , Ácido Pentético/análogos & derivados , Ácido Pentético/farmacocinética , Receptores de Somatostatina/clasificación , Receptores de Somatostatina/metabolismo , Somatostatina , Animales , Radioisótopos de Indio/farmacocinética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Radiofármacos/farmacocinética , Receptores de Somatostatina/deficiencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Somatostatina/farmacocinética , Distribución Tisular , Recuento Corporal Total
14.
J Nucl Med ; 44(1): 98-104, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12515882

RESUMEN

UNLABELLED: Peptide receptor radionuclide therapy (PRRT) using [(111)In-DTPA(0)]octreotide (where DTPA is diethylenetriaminepentaacetic acid) is feasible because, besides gamma-radiation, (111)In emits both therapeutic Auger and internal conversion electrons having a tissue penetration of 0.02-10 and 200-500 micro m, respectively. The aim of this study was to investigate the therapeutic effects of [(111)In-DTPA(0)]octreotide in a single-cell model including the effects of incubation time, radiation dose, and specific activity of [(111)In-DTPA(0)]octreotide. Finally, we discriminated between the effects of the Auger electrons and internal conversion electrons in PRRT. METHODS: An in vitro, colony-forming assay to study cell survival after PRRT using the sst subtype 2-positive rat pancreatic tumor cell line CA20948 was developed. RESULTS: In this in vitro system [(111)In-DTPA(0)]octreotide can control tumor growth to 0% survival, and the effects were dependent on incubation time, radiation dose, and specific activity used. Similar concentrations of (111)In-DTPA, which is not internalized into sst-positive tumor cells like [(111)In-DTPA(0)]octreotide, did not influence tumor survival. Excess unlabeled octreotide (10(-6) mol/L) could decrease tumor cell survival to 60% of control; the addition of radiolabeled peptide ([(111)In-DTPA(0)]octreotide [10(-9) mol/L] + 10(-6) mol/L octreotide) did not further decrease survival. CONCLUSION: These in vitro studies show that the therapeutic effect of (111)In is dependent on internalization, enabling the Auger electrons with their very short particle range to reach the nucleus. Our results also indicate that the PRRT effects were receptor mediated.


Asunto(s)
Apoptosis/efectos de la radiación , Radioisótopos de Indio/uso terapéutico , Octreótido/análogos & derivados , Octreótido/uso terapéutico , Neoplasias Pancreáticas/radioterapia , Ácido Pentético/análogos & derivados , Ácido Pentético/uso terapéutico , Radiofármacos/uso terapéutico , Animales , Relación Dosis-Respuesta en la Radiación , Electrones , Ratas , Receptores de Péptidos/uso terapéutico , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Células Tumorales Cultivadas
15.
Semin Nucl Med ; 32(2): 110-22, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11965606

RESUMEN

Fifty patients with somatostatin receptor-positive tumors were treated with multiple doses of [(111)In-diethylenetriamine pentaacetic acid(0)]octreotide. Forty patients were evaluable after cumulative doses of at least 20 GBq up to 160 GBq. Therapeutic effects were seen in 21 patients: partial remission in 1 patient, minor remissions in 6 patients, and stabilization of previously progressive tumors in 14 patients. Our results thus underscore the therapeutic potential of Auger-emitting radiolabelled peptides. The toxicity was generally mild bone marrow toxicity, but 3 of the 6 patients who received more than 100 GBq developed a myelodysplastic syndrome or leukemia. Therefore, we consider 100 GBq as the maximal tolerable dose. With a renal radiation dose of 0.45 mGy/MBq (based on previous studies) a cumulative dose of 100 GBq [(111)In-DTPA(0)]octreotide will lead to 45Gy on the kidneys, twice the accepted limit for external beam radiation. However, no development of hypertension, proteinuria, or significant changes in serum creatinine or creatinine clearance were observed in our patients including 2 patients who received 106 and 113 GBq [(111)In-DTPA(0)]octreotide without protection with amino acids, over a follow-up period of respectively 3 and 2 years. These findings show that the radiation of the short-range (maximal 10 microns) Auger electrons originating from the cells of the proximal tubules is not harmful for the renal function. The decrease in serum inhibin B and concomitant increase of serum FSH levels in men indicate that the spermatogenesis was impaired.


Asunto(s)
Neoplasias/radioterapia , Octreótido/análogos & derivados , Octreótido/uso terapéutico , Ácido Pentético/análogos & derivados , Ácido Pentético/uso terapéutico , Radiofármacos/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/química , Neoplasias/diagnóstico por imagen , Tumores Neuroendocrinos/diagnóstico por imagen , Tumores Neuroendocrinos/radioterapia , Octreótido/administración & dosificación , Octreótido/efectos adversos , Ácido Pentético/administración & dosificación , Ácido Pentético/efectos adversos , Cintigrafía , Radiofármacos/administración & dosificación , Radiofármacos/efectos adversos , Dosificación Radioterapéutica , Receptores de Somatostatina/análisis
16.
Semin Nucl Med ; 32(2): 133-40, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11965608

RESUMEN

In preclinical studies in rats we evaluated biodistribution and therapeutic effects of different somatostatin analogs, [(111)In-DTPA]octreotide, [(90)Y-DOTA,Tyr(3)]octreotide and [(177)Lu-DOTA,Tyr(3)]octreotate, currently also being applied in clinical radionuclide therapy studies. [Tyr(3)]octreotide and [Tyr(3)]octreotate, chelated with DTPA or DOTA, both showed high affinity binding to somatostatin receptor subtype 2 (sst(2)) in vitro. The radiolabelled compounds all showed high tumor uptake in sst(2)-positive tumors in vivo in rats, the highest uptake being reached with [(177)Lu-DOTA,Tyr(3)]octreotate. In preclinical therapy studies in vivo in rats, excellent, dose dependent, tumor size responses were found, responses appeared to be dependent on tumor size at therapy start. These preclinical data showed the great promise of radionuclide therapy with radiolabelled somatostatin analogues. They emphasised the concept that especially the combination of somatostatin analogs radiolabeled with different radionuclides, like (90)Y and (177)Lu, is most promising to reach a wider tumor size region of high curability. Furthermore, different phase I clinical studies, using [(111)In-DTPA]octreotide, [(90)Y-DOTA,Tyr(3)]octreotide or [(177)Lu-DOTA, Tyr(3)]octreotate are described. Fifty patients with somatostatin receptor-positive tumors were treated with multiple doses of [(111)In-DTPA(0)]octreotide. Forty patients were evaluable after cumulative doses of at least 20 GBq up to 160 GBq. Therapeutic effects were seen in 21 patients: partial remission in 1 patient, minor remissions in 6 patients, and stabilization of previously progressive tumors in 14 patients. The toxicity was generally mild bone marrow toxicity, but 3 of the 6 patients who received more than 100 GBq developed a myelodysplastic syndrome or leukemia. Radionuclide therapy with [(90)Y-DOTA,Tyr(3)]octreotide started in 3 different phase I trials. Overall, antimitotic effects have been observed: about 20% partial response and 60% stable disease (N = 92) along with complete symptomatic cure of several malignant insulinoma and gastrinoma patients. Maximum cumulative [(90)Y-DOTA,Tyr(3)]octreotide dose was about 26 GBq, without reaching the maximum tolerable dose. New is the use of [(177)Lu-DOTA,Tyr(3)]octreotate, which shows the highest tumor uptake of all tested octreotide analogs so far, with excellent tumor-to-kidney ratios. Radionuclide therapy with this analog in a phase 1 trial started recently in our center in 63 patients (238 administrations), Interim analysis of 18 patients with neuroendocrine tumors was performed very recently. According to the WHO, toxicity criteria no dose limiting toxicity was observed. Minor CT-assessed tumor shrinkage (25% - 50% reduction) was noticed in 6% of 18 patients and partial remission (50% - 100% reduction, SWOG criteria) in 39%. Eleven percent of patients had tumor progression and in 44% no changes were seen. These data show that radionuclide therapy with radiolabelled somatostatin analogs, like [DOTA, Tyr(3)]octreotide and [DOTA, Tyr(3)octreotate is a most promising new treatment modality for patients who have sst(2)-positive tumors.


Asunto(s)
Radioisótopos de Indio/uso terapéutico , Octreótido/análogos & derivados , Octreótido/uso terapéutico , Compuestos Organometálicos/uso terapéutico , Ácido Pentético/análogos & derivados , Ácido Pentético/uso terapéutico , Radiofármacos/uso terapéutico , Radioisótopos de Itrio/uso terapéutico , Animales , Humanos , Neoplasias Hepáticas/radioterapia , Neoplasias Hepáticas/secundario , Tumores Neuroendocrinos/química , Tumores Neuroendocrinos/radioterapia , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/radioterapia , Ratas , Receptores de Somatostatina/análisis , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...