Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Eur J Nucl Med Mol Imaging ; 37(7): 1335-44, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20157707

ABSTRACT

PURPOSE: Functionally critically located gliomas represent a challenging subgroup of intrinsic brain neoplasms. Standard therapeutic recommendations often cannot be applied, because radical treatment and preservation of neurological function are contrary goals. The successful targeting of gliomas with locally injected beta radiation-emitting (90)Y-DOTAGA-substance P has been shown previously. However, in critically located tumours, the mean tissue range of 5 mm of (90)Y may seriously damage adjacent brain areas. In contrast, the alpha radiation-emitting radionuclide (213)Bi with a mean tissue range of 81 microm may have a more favourable toxicity profile. Therefore, we evaluated locally injected (213)Bi-DOTA-substance P in patients with critically located gliomas as the primary therapeutic modality. METHODS: In a pilot study, we included five patients with critically located gliomas (WHO grades II-IV). After diagnosis by biopsy, (213)Bi-DOTA-substance P was locally injected, followed by serial SPECT/CT and MR imaging and blood sampling. Besides feasibility and toxicity, the functional outcome was evaluated. RESULTS: Targeted radiopeptide therapy using (213)Bi-DOTA-substance P was feasible and tolerated without additional neurological deficit. No local or systemic toxicity was observed. (213)Bi-DOTA-substance P showed high retention at the target site. MR imaging was suggestive of radiation-induced necrosis and demarcation of the tumours, which was validated by subsequent resection. CONCLUSION: This study provides proof of concept that targeted local radiotherapy using (213)Bi-DOTA-substance P is feasible and may represent an innovative and effective treatment for critically located gliomas. Primarily non-operable gliomas may become resectable with this treatment, thereby possibly improving the prognosis.


Subject(s)
Alpha Particles/therapeutic use , Glioma/radiotherapy , Heterocyclic Compounds, 1-Ring/therapeutic use , Organometallic Compounds/therapeutic use , Substance P/analogs & derivatives , Adult , Feasibility Studies , Glioma/metabolism , Heterocyclic Compounds, 1-Ring/administration & dosage , Heterocyclic Compounds, 1-Ring/adverse effects , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Humans , Injections , Middle Aged , Organometallic Compounds/administration & dosage , Organometallic Compounds/adverse effects , Organometallic Compounds/pharmacokinetics , Pilot Projects , Substance P/administration & dosage , Substance P/adverse effects , Substance P/pharmacokinetics , Substance P/therapeutic use , Treatment Outcome
2.
Nucl Med Biol ; 35(8): 839-49, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19026945

ABSTRACT

Medullary thyroid carcinoma (MTC) expresses CCK-2 receptors. (111)In-labeled DOTA-DGlu-Ala-Tyr-Gly-Trp-Met-Asp-Phe-NH(2) (DOTA-MG11), DOTA-DAsp-Tyr-Nle-Gly-Trp-Nle-Asp-Phe-NH(2) (DOTA-CCK), and (99m)Tc-labeled N(4)-Gly-DGlu-(Glu)(5)-Ala-Tyr-Gly-Trp-Met-Asp-Phe-NH(2) ((99m)Tc-Demogastrin 2) are analogs developed for CCK-2 receptor-targeted scintigraphy. All 3 radiolabeled analogs were selected on the basis of their high CCK-2 receptor affinity and their good in vitro serum stability, with in vitro serum t(1/2) values of several hours. Radiolabeling of DOTA-peptides with (111)In requires a heating procedure, typically in the range of 80 degrees -100 degrees C up to 30 min. Following this procedure with DOTA-MG11 resulted in a >98 % incorporation of (111)In, however, with a radiochemical purity (RCP) of <50 %. The decrease in RCP was found to be due to oxidation of the methionine residue in the molecule. Moreover, this oxidized compound lost its CCK-2 receptor affinity. Therefore, conditions during radiolabeling were optimised: labeling of DOTA-MG11 and DOTA-CCK with (111)In involved 5 min heating at 80 degrees C and led to an incorporation of (111)In of >98 %. In addition, all analogs were radiolabeled in the presence of quenchers to prevent radiolysis and oxidation resulting in a RCP of >90 %. All 3 radiolabeled analogs were i.v. administered to 6 MTC patients: radioactivity cleared rapidly by the kidneys, with no significant differences in the excretion pattern of the 3 radiotracers. All 3 radiolabeled analogs exhibited a low in vivo stability in patients, as revealed during analysis of blood samples, with the respective t(1/2) found in the order of minutes. In patient blood, the rank of radiopeptide in vivo stability was: (99m)Tc-Demogastrin 2 (t(1/2) 10-15 min)>(111)In-DOTA-CCK (t(1/2) approximately 5-10 min)>(111)In-DOTA-MG11 (t(1/2)<5 min).


Subject(s)
Carcinoma, Medullary/diagnostic imaging , Isotope Labeling , Radioligand Assay , Radiopharmaceuticals/metabolism , Receptor, Cholecystokinin B/metabolism , Thyroid Neoplasms/diagnostic imaging , Adolescent , Adult , Aged , Autoradiography , Chromatography, High Pressure Liquid , Drug Stability , Female , Gastrins/metabolism , Humans , Male , Middle Aged , Radionuclide Imaging , Radiopharmaceuticals/chemistry , Receptor, Cholecystokinin B/analysis
3.
Br J Cancer ; 93(10): 1144-51, 2005 Nov 14.
Article in English | MEDLINE | ID: mdl-16251870

ABSTRACT

Somatostatin receptor (sstr)-mediated radiation therapy is a new therapeutic modality for neuroendocrine (NE) tumours. High expression of sstr in NE tumours leads to tumour-specific uptake of radiolabelled somatostatin analogues and high absorbed doses. In this study, we present the first optimised radiation therapy via sstr using [(177)Lu-DOTA(0)-Tyr(3)]-octreotate given to nude mice xenografted with the human midgut carcinoid GOT1. The tumours in 22 out of 23 animals given therapeutic amounts showed dose-dependent, rapid complete remission. The diagnostic amount (0.5 MBq [(177)Lu-DOTA(0)-Tyr(3)]-octreotate) did not influence tumour growth and was rapidly excreted. In contrast, the therapeutic amount (30 MBq [(177)Lu-DOTA(0)-Tyr(3)]-octreotate) induced rapid tumour regression and entrapment of (177)Lu so that the activity concentration of (177)Lu remained high, 7 and 13 days after injection. The entrapment phenomenon increased the absorbed dose to tumours from 1.6 to 4.0 Gy MBq(-1) and the tumours in animals treated with 30 MBq received 120 Gy. Therapeutic amounts of [(177)Lu-DOTA(0)-Tyr(3)]-octreotate rapidly induced apoptosis and gradual development of fibrosis in grafted tumours. In conclusion, human midgut carcinoid xenografts can be cured by receptor-mediated radiation therapy by optimising the uptake of radioligand and taking advantage of the favourable change in biokinetics induced by entrapment of radionuclide in the tumours.


Subject(s)
Carcinoid Tumor/metabolism , Carcinoid Tumor/radiotherapy , Receptors, Somatostatin/metabolism , Animals , Carcinoid Tumor/drug therapy , Humans , Mice , Mice, Nude , Octreotide/analogs & derivatives , Octreotide/pharmacokinetics , Octreotide/therapeutic use , Xenograft Model Antitumor Assays
4.
Eur J Nucl Med Mol Imaging ; 31(8): 1097-104, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15029459

ABSTRACT

68Ge/68Ga generators provide cyclotron-independent access to positron emission tomography (PET) radiopharmaceuticals. We describe a system which allows the safe and efficient handling of 68Ge/68Ga generator eluates for labelling of DOTA-derivatised peptide ligands. The system comprises concentration and purification of the 68Ga eluate as well as labelling and purification steps for peptides, and can be used with different 68Ge/68Ga generator types. The suitability and efficiency were tested with two different DOTA-derivatised somatostatin derivatives and a DOTA-derivatised bombesin derivative. Amounts of 10-20 nmol of the peptides were sufficient and resulted in labelling yields of 50% for all peptides. The built-in safety precautions have proven to be appropriate in allowing use of the method for routine clinical applications. The system was set up and operated in a "hot lab" by personnel with no previous experience in the preparation of PET radiopharmaceuticals.


Subject(s)
Gallium Radioisotopes/chemistry , Heterocyclic Compounds, 1-Ring/chemistry , Isotope Labeling/instrumentation , Isotope Labeling/methods , Peptides/chemistry , Radiopharmaceuticals/chemistry , Bombesin/analogs & derivatives , Bombesin/isolation & purification , Gallium Radioisotopes/isolation & purification , Heterocyclic Compounds, 1-Ring/chemical synthesis , Heterocyclic Compounds, 1-Ring/isolation & purification , Ligands , Peptides/chemical synthesis , Peptides/isolation & purification , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/isolation & purification , Somatostatin/analogs & derivatives , Somatostatin/isolation & purification , Specimen Handling/instrumentation , Specimen Handling/methods
5.
J Inorg Biochem ; 97(4): 315-23, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14568235

ABSTRACT

In spite of the fact that Group 13 metal ions (Al(3+), Ga(3+), In(3+) and Tl(+/3+)) show no main biological role, they are NMR-active nuclides which can be used in magnetic resonance spectroscopy of biologically relevant systems. The fact that these metal ions are quadrupolar (with the exception of thallium) means that they are particularly sensitive to ligand type and coordination geometry. The line width of the NMR signals of their complexes shows a strong dependence on the symmetry of coordination, which constitutes an effective tool in the elucidation of structures. Here we report published NMR studies of this family of elements, applied to systems of biological importance. Special emphasis is given to binding studies of these cations to biological molecules, such as proteins, and to chelating agents of radiopharmaceutical interest. The possibility of in vivo NMR studies is also stressed, with extension to (27)Al-based MRI (magnetic resonance imaging) experiments.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Metals/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Aluminum/chemistry , Aluminum/metabolism , Animals , Cations/chemistry , Gallium/chemistry , Gallium/metabolism , Gallium Isotopes , Humans , Indium/chemistry , Indium/metabolism , Isotopes , Ligands , Metals/metabolism , Proteins/chemistry , Proteins/metabolism , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/metabolism , Thallium/chemistry , Thallium/metabolism
6.
Cancer Biother Radiopharm ; 17(5): 527-33, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12470422

ABSTRACT

Distribution profiles and elimination pathways in rats of two new octreotate derivatives radiolabeled with yttrium, namely Y-DOTAGA-tate and Y-DOTA-t-GA-tate, were compared with those of Y-DOTA-octreotide and Y-DOTA-Tyr(3)-octreotide. All synthetic somatostatin analogues under study were rapidly cleared from the blood and most organs of rats. The main elimination pathway for all peptides under study was urine excretion. High and long-term uptakes of radioactivity in the kidneys and also in organs with high density of somatostatin receptors (the adrenals and pancreas) were found. Radioactivity concentrations in these somatostatin receptor-rich organs were substantially higher for octreotate derivatives in comparison with octreotide analogues; the highest values for Y-DOTAGA-tate were determined. The octreotate derivatives under study appear to be specific ligands for treatment of somatostatin receptor-positive tumors if some mechanism to decrease their kidney retention is provided.


Subject(s)
Octreotide/analogs & derivatives , Octreotide/pharmacokinetics , Animals , Blood Proteins/metabolism , Male , Rats , Rats, Wistar , Receptors, Somatostatin/metabolism , Yttrium Radioisotopes
7.
Eur J Nucl Med ; 28(12): 1743-50, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11734910

ABSTRACT

The somatostatin analogue (90)Y-DOTATOC (yttrium-90 DOTA- D-Phe(1)-Tyr(3)-octreotide) is used for treatment of patients with neuroendocrine tumours. Accurate pretherapeutic dosimetry would allow for individual planning of the optimal therapeutic strategy. In this study, the biodistribution and resulting dosimetric calculation for therapeutic exposure of critical organs and tumour masses based on the positron emission tomography (PET) tracer (86)Y-DOTATOC, which is chemically identical to the therapeutic agent, were compared with results based on the tracer commonly used for somatostatin receptor scintigraphy, (111)In-DTPA-octreotide (indium-111 DTPA- D-Phe(1)-octreotide, OctreoScan). Three patients with metastatic carcinoid tumours were investigated. Dynamic and static PET studies with 77-186 MBq (86)Y-DOTATOC were performed up to 48 h after injection. Serum and urinary activity were measured simultaneously. Within 1 week, but not sooner than 5 days, patients were re-investigated by conventional scintigraphy with (111)In-DTPA-octreotide (110-187 MBq) using an equivalent protocol. Based on the regional tissue uptake kinetics, residence times were calculated and doses for potential therapy with (90)Y-DOTATOC were estimated. Serum kinetics and urinary excretion of both tracers showed no relevant differences. Estimated liver doses were similar for both tracers. Dose estimation for organs with the highest level of radiation exposure, the kidneys and spleen, showed differences of 10.5%-20.1% depending on the tracer. The largest discrepancies in dose estimation, ranging from 23.1% to 85.9%, were found in tumour masses. Furthermore, there was a wide inter-subject variability in the organ kinetics. Residence times (tau(organs)) for (90)Y-DOTATOC therapy were: tau(liver) 1.59-2.79 h; tau(spleen) 0.07-1.68 h; and tau(kidneys) 0.55-2.46 h (based on (86)Y-DOTATOC). These data suggest that dosimetry based on (86)Y-DOTATOC and (111)In-DTPA-octreotide yields similar organ doses, whereas there are relevant differences in estimated tumour doses. Individual pretherapeutic dosimetry for (90)Y-DOTATOC therapy appears necessary considering the large differences in organ doses between individual patients. If possible, the dosimetry should be performed with the chemically identical tracer (86)Y-DOTATOC.


Subject(s)
Carcinoid Tumor/diagnostic imaging , Carcinoid Tumor/radiotherapy , Indium Radioisotopes/therapeutic use , Octreotide/analogs & derivatives , Octreotide/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Somatostatin/pharmacokinetics , Yttrium Radioisotopes/therapeutic use , Aged , Humans , Male , Middle Aged , Octreotide/therapeutic use , Radiation Dosage , Radiometry , Radiopharmaceuticals/therapeutic use , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Tissue Distribution , Tomography, Emission-Computed
8.
Thyroid ; 11(7): 647-59, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11484893

ABSTRACT

Despite the fact that several recent studies report an expression of somatostatin receptors in nonmedullary thyroid cancer (non-MTC), there is still no consensus concerning the diagnostic and therapeutic usefulness of radionuclide-labeled somatostatin analogues in non-MTC. We present the results of 50 scintigraphic studies with (111)In-Pentetreotide ((111)In-P) in 48 patients with metastasizing non-MTC (n = 9 papillary, n = 9 follicular, n = 29 Hurthle cell, n = 1 insular carcinoma). The findings were compared with histology and with other imaging modalities. (111)In-P provided unequivocally positive results in 37 of 50 (74%) of the patients (27% in the 11 patients with current thyroglobulin levels <10 ng/mL and 85% in the patients with thyroglobulin >10 ng/mL). Histopathology demonstrated that maximal uptake was observed in Hurthle cell carcinoma (95% positive examinations if thyroglobulin exceeds 10 ng/mL). We also describe for the first time dosimetric and clinical data from the courses of 90Y-DOTATOC therapy in three patients with progressive, somatostatin-receptor-positive non-MTC (up to 9.3 GBq per 4 cycles). Tumor progression could not be stopped in any of the patients treated with 90Y-DOTATOC. We conclude that (111)In-P is a promising tool for whole-body diagnosis in nonradioiodine-accumulating non-MTC, especially in Hürthle cell cancer, and if 2-[18F]fluorodeoxyglucose-positron emission tomography (FDG-PET) is not available. Although the number of patients treated with 90Y-DOTATOC is still limited, our applied treatment protocol appears to be ineffective in metastasizing non-MTC.


Subject(s)
Octreotide/analogs & derivatives , Octreotide/therapeutic use , Radiopharmaceuticals/therapeutic use , Somatostatin/analogs & derivatives , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/drug therapy , Yttrium Radioisotopes/therapeutic use , Adult , Aged , Aged, 80 and over , Female , Humans , Indium Radioisotopes , Male , Middle Aged , Radionuclide Imaging
9.
J Nucl Med ; 42(7): 1053-6, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11438627

ABSTRACT

UNLABELLED: Imaging of somatostatin receptors (SSTRs) using [111In]diethylenetriaminepentaacetic-acid-octreotide (DTPAOC) has proven to be helpful in the differentiation of meningiomas, neurinomas or neurofibromas, and metastases as well as in the follow-up of meningiomas. A drawback of the SPECT method is its limited sensitivity in detecting small meningiomas. Because of PET's increased spatial resolution and its ability to absolutely quantify biodistribution, a PET tracer for SSTR imaging would be desirable. METHODS: 1,4,7,10-tetraazacyclododecane-N,N',N",N"'-tetraacetic-acid-D-Phe1-Tyr3-octreotide (DOTATOC) was labeled using the positron-emitting generator nuclide 68Ga. We acquired dynamic PET images over 120 min after intravenous injection of 175 MBq [68Ga]DOTATOC in 3 patients suffering from 8 meningiomas (WHO I degrees; 7- to 25-mm diameter). Patients' heads had been fixed using individually shaped fiber masks equipped with an external stereotactic localizer system to match PET, CT, and MRI datasets. RESULTS: [68Ga]DOTATOC was rapidly cleared from the blood (half-life alpha, 3.5 min; half-life beta, 63 min). Standardized uptake values (SUVs) of meningiomas increased immediately after injection and reached a plateau 60-120 min after injection (mean SUV, 10.6). No tracer could be found in the surrounding healthy brain tissue. All meningiomas (even the 3 smallest [7- to 8-mm diameter]) showed high tracer uptake and could be visualized clearly. Tracer boundaries showed a good correspondence with the matched CT and MRI images. PET provided valuable additional information regarding the extent of meningiomas located beneath osseous structures, especially at the base of the skull. CONCLUSION: According to our initial experiences, [68Ga]DOTATOC seems to be a very promising new PET tracer for imaging SSTRs even in small meningiomas, offering excellent imaging properties and a very high tumor-to-background ratio.


Subject(s)
Gallium Radioisotopes , Meningeal Neoplasms/diagnostic imaging , Meningioma/diagnostic imaging , Octreotide/analogs & derivatives , Radiopharmaceuticals , Receptors, Somatostatin/analysis , Tomography, Emission-Computed , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Meningeal Neoplasms/chemistry , Meningioma/chemistry , Pilot Projects , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed
10.
Eur J Nucl Med ; 28(4): 426-34, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11357492

ABSTRACT

A newly developed somatostatin radioligand, DOTA-[D-Phe1-Tyr3]-octreotide (DOTATOC), has been synthesised for therapeutic purposes, because of its stable and easy labelling with yttrium-90. The aim of this study was to determine the dosage, safety profile and therapeutic efficacy of 90Y-DOTATOC in patients with cancers expressing somatostatin receptors. We recruited 30 patients with histologically confirmed cancer. The main inclusion criterion was the presence of somatostatin receptors as documented by 111In-DOTATOC scintigraphy. 90Y-DOTATOC was injected intravenously using a horizontal protocol: patients received equivalent-activity doses in each of three cycles over 6 months. The first six patients received 1.11 GBq per cycle and the four successive groups of six patients received doses increasing in 0.37-GBq steps. Toxicity was evaluated according to WHO criteria. No patient had acute or delayed adverse reactions up to 2.59 GBq 90Y-DOTATOC per cycle (total 7.77 GBq). After a total dose of 3.33 GBq, one patient developed grade II renal toxicity 6 months later. The maximum tolerated dose per cycle has not yet been reached, although transient lymphocytopenia has been observed. Total injectable activity is limited by the fact that the maximum dose tolerated by the kidneys has been estimated at 20-25 Gy. Complete or partial tumour mass reduction occurred in 23% of patients; 64% had stable and 13% progressive disease. It is concluded that high activities of 90Y-DOTATOC can be administered with a low risk of myelotoxicity, although the cumulative radiation dose to the kidneys is a limiting factor and requires careful evaluation. Objective therapeutic responses have been observed.


Subject(s)
Neoplasms/radiotherapy , Octreotide/analogs & derivatives , Octreotide/therapeutic use , Radiopharmaceuticals/therapeutic use , Receptors, Somatostatin/drug effects , Somatostatin/metabolism , Adult , Aged , Female , Humans , Indicators and Reagents , Male , Middle Aged , Octreotide/adverse effects , Octreotide/pharmacokinetics , Radiometry , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Tomography, X-Ray Computed , Yttrium Radioisotopes/therapeutic use
11.
Bioorg Med Chem Lett ; 10(18): 2133-5, 2000 Sep 18.
Article in English | MEDLINE | ID: mdl-10999487

ABSTRACT

New DOTA-based bifunctional prochelators, e.g., 1-(1-carboxy-3-carbotertbutoxypropyl)-4,7,10-(carbotertbutoxyme thyl)-1,4,7,10-tetraazacyclodode-cane (DOTAGA(tBu)4), (6d) for a broad application in the modification of biomolecules with metal ions were prepared. The five-step synthesis of 6d has an overall yield of about 20%. The coupling of 6d to a bioactive peptide on solid-phase was exemplified with use of a CCK-B (cholecystokinin) analogue.


Subject(s)
Chelating Agents/chemical synthesis , Prodrugs/chemical synthesis , Cholecystokinin/chemical synthesis , Molecular Mimicry , Oligopeptides/chemical synthesis , Organometallic Compounds/chemical synthesis , Radioisotopes/chemistry
12.
Endocrinology ; 141(9): 3304-12, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10965902

ABSTRACT

Somatostatin analogs labeled with radionuclides are of considerable interest in nuclear oncology as diagnostic or therapeutic tools for somatostatin receptor (SSTR)-expressing tumors. We investigated the suitability of DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) as a replacement for the widely used diethylenetriaminepentaacetic acid, to enable stable labeling of somatostatin analogs with both therapeutic (90Y) and diagnostic (111In) radionuclides. The three clinically relevant somatostatin agonists, octreotide, vapreotide, and lanreotide, together with the newly designed Tyr3-octreotide (TyrOc), were conjugated to DOTA and labeled with 90Y or 111In. For all DOTA-somatostatin analogs tested, irrespective of the incorporated radionuclide, we observed favorable biodistribution profiles in AR4-2J tumor-bearing mice: 1) a rapid clearance from all SSTR-negative tissues except kidney; 2) a specific uptake in SSTR-positive tissues, including tumor; and 3) an excellent tumor penetration. The main route of excretion was via the kidneys. Nevertheless, DOTATOC was clearly superior to the other DOTA-somatostatin analogs tested, as well as OctreoScan, as indicated by the highest tumor-to-nontarget-tissue ratio, including the tumor-to-SSTR-positive-tissue ratios. The presence of different SSTR subtypes in the SSTR-positive tissues possibly contributes to these differential uptakes. We assume that the very favorable behavior of DOTATOC in our mouse model makes this radioligand very promising for future applications in nuclear oncology.


Subject(s)
Heterocyclic Compounds, 1-Ring/pharmacology , Hormone Antagonists/pharmacology , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/radiotherapy , Radiopharmaceuticals/therapeutic use , Somatostatin/pharmacology , Animals , Chelating Agents/pharmacology , Chromatography, High Pressure Liquid , Female , Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Hormone Antagonists/pharmacokinetics , Indium Radioisotopes/therapeutic use , Kidney/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Messenger/biosynthesis , Receptors, Somatostatin/drug effects , Somatostatin/analogs & derivatives , Somatostatin/chemistry , Tissue Distribution , Yttrium Radioisotopes/therapeutic use
13.
Eur J Nucl Med ; 27(3): 273-82, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10774879

ABSTRACT

In vivo somatostatin receptor scintigraphy using Octreoscan is a valuable method for the visualisation of human endocrine tumours and their metastases. Recently, several new, alternative somatostatin radioligands have been synthesised for diagnostic and radiotherapeutic use in vivo. Since human tumours are known to express various somatostatin receptor subtypes, it is mandatory to assess the receptor subtype affinity profile of such somatostatin radiotracers. Using cell lines transfected with somatostatin receptor subtypes sst1, sst2, sst3, sst4 and sst5, we have evaluated the in vitro binding characteristics of labelled (indium, yttrium, gallium) and unlabelled DOTA-[Tyr3]-octreotide, DOTA-octreotide, DOTA-lanreotide, DOTA-vapreotide, DTPA-[Tyr3]-octreotate and DOTA-[Tyr3]-octreotate. Small structural modifications, chelator substitution or metal replacement were shown to considerably affect the binding affinity. A marked improvement of sst2 affinity was found for Ga-DOTA-[Tyr3]-octreotide (IC50 2.5 nM) compared with the Y-labelled compound and Octreoscan. An excellent binding affinity for sst2 in the same range was also found for In-DTPA-[Tyr3]-octreotate (IC50 1.3 nM) and for Y-DOTA-[Tyr3]-octreotate (IC50 1.6 nM). Remarkably, Ga-DOTA-[Tyr3]-octreotate bound at sst2 with a considerably higher affinity (IC50 0.2 nM). An up to 30-fold improvement in sst3 affinity was observed for unlabelled or Y-labelled DOTA-octreotide compared with their Tyr3-containing analogue, suggesting that replacement of Tyr3 by Phe is crucial for high sst3 affinity. Substitution in the octreotide molecule of the DTPA by DOTA improved the sst3 binding affinity 14-fold. Whereas Y-DOTA-lanreotide had only low affinity for sst3 and sst4, it had the highest affinity for sst5 among the tested compounds (IC50 16 nM). Increased binding affinity for sst3 and sst5 was observed for DOTA-[Tyr3]-octreotide, DOTA-lanreotide and DOTA-vapreotide when they were labelled with yttrium. These marked changes in subtype affinity profiles are due not only to the different chemical structures but also to the different charges and hydrophilicity of these compounds. Interestingly, even the coordination geometry of the radiometal complex remote from the pharmacophoric amino acids has a significant influence on affinity profiles as shown with Y-DOTA versus Ga-DOTA in either [Tyr3]-octreotide or [Tyr3]-octreotate. Such changes in sst affinity profiles must be identified in newly designed radiotracers used for somatostatin receptor scintigraphy in order to correctly interpret in vivo scintigraphic data. These observations may represent basic principles relevant to the development of other peptide radioligands.


Subject(s)
Radiopharmaceuticals , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Animals , Autoradiography , Binding, Competitive , Humans , In Situ Hybridization , Octreotide/chemistry , Peptides, Cyclic/chemistry , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/therapeutic use , Rats , Receptors, Somatostatin/classification , Somatostatin/chemistry , Somatostatin/therapeutic use
14.
Praxis (Bern 1994) ; 88(31-32): 1263-8, 1999 Aug 15.
Article in German | MEDLINE | ID: mdl-10479965

ABSTRACT

Current concepts for the treatment of somatostatin receptor positive tumours have not been very motivating up to now. A promising alternative could be the new peptidic vector DOTA-D-Phe1-Tyr3-Octreotide (DOTATOC) recently developed in Basel. It may be labelled with the beta-emitter yttrium-90 (90Y) for internal radiotherapy after systemic application. Pilot therapy studies have shown convincing results with this new radiopharmaceutical. These studies are presented with regard to efficacy and possible toxicity. In summary, the new receptor-mediated 90Y-DOTATOC therapy led to tumour response in the majority of patients, and only in some receiving high cumulative doses of > 200 mCi per m2 body surface renal and hematological toxicity due to irradiation occurred. For the reduction of renal accretion, concepts with concomitant amino acid infusions containing L-lysine in a higher concentration are currently under way.


Subject(s)
Neuroendocrine Tumors/radiotherapy , Octreotide/analogs & derivatives , Yttrium Radioisotopes/therapeutic use , Female , Follow-Up Studies , Humans , Middle Aged , Neuroendocrine Tumors/chemistry , Neuroendocrine Tumors/diagnostic imaging , Octreotide/adverse effects , Octreotide/therapeutic use , Pilot Projects , Prospective Studies , Radiation Injuries/etiology , Radionuclide Imaging , Radiotherapy Dosage , Receptors, Somatostatin/analysis , Treatment Outcome , Yttrium Radioisotopes/adverse effects
15.
Cancer Res ; 59(15): 3652-7, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10446977

ABSTRACT

Octreotide is a somatostatin analogue that is widely used for cancer therapy and tumor imaging. Its efficacy in tumors depends mainly on the expression of the somatostatin receptor type 2 (sst 2). Desensitization and down-regulation of sst 2 after agonist exposure can have important consequences for patients under ongoing octreotide therapy because it may induce temporary tumor unresponsiveness and impair sst 2-based tumor scintigraphy. Therefore, we have investigated the effect of octreotide on sst 2 expression in vitro, as well as in a tumor mouse model. In vitro, short exposure to octreotide induced rapid dose-dependent down-regulation of sst 2 in the rat pancreatic AR4-2J cell line. Within 0.5 h, 80% of sst 2 had disappeared from the cell surface. A total recovery required 24 h and was shown to depend on protein synthesis, but not on new sst 2 mRNA transcription, indicating that sst 2 was probably degraded during the down-regulation process. Similar results were obtained in vivo. On the other hand, long-term continuous release of octreotide for 7 days, as achieved with octreotide-containing osmotic minipumps, caused sst 2 up-regulation in vivo, but not in vitro. Furthermore, this up-regulation of sst 2 in tumor-bearing scid mice was shown to depend on constant exposure of the animals to octreotide, as it was not observed when octreotide was given discontinuously in two s.c. daily injections. These results demonstrate that the continuous release of a small amount of octreotide, which in cancer therapy may be achieved with long-acting release formulations of the peptide, can induce sst 2 up-regulation on cancer cells. This may improve the efficacy of both tumor imaging and long-term octreotide therapy.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/biosynthesis , Octreotide/therapeutic use , Pancreatic Neoplasms/pathology , Receptors, Somatostatin/biosynthesis , Somatostatin/physiology , Up-Regulation/drug effects , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Mice , Mice, SCID , Neoplasm Proteins/drug effects , Neoplasm Proteins/genetics , Nucleic Acid Synthesis Inhibitors/pharmacology , Octreotide/pharmacology , Pancreatic Neoplasms/metabolism , Protein Synthesis Inhibitors/pharmacology , Rats , Receptors, Somatostatin/drug effects , Receptors, Somatostatin/genetics , Tumor Cells, Cultured
16.
Eur J Nucl Med ; 26(7): 693-8, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10398816

ABSTRACT

Radiolabelled tumour receptor-binding peptides can be used for in vivo scintigraphic imaging. Recently, the somatostatin analogue [Tyr3]octreotide (D-Phe-c(Cys-Tyr-D-Trp-Lys-Thr-Cys)-Thr(ol)) was derivatized with the chelator DOTA (tetra-azacyclododecane-tetra-acetic acid), enabling stable radiolabelling with both the high-energy beta particle-emitter yttrium-90 and the Auger electron-emitter indium-111. The thus produced radiolabelled compounds are promising for peptide receptor radionuclide therapy. Our previous in vitro and in vivo (rat) experiments with these radiolabelled compounds showed favourable binding and biodistribution characteristics with high uptake and retention in the target organs. We also demonstrated receptor-specific, time- and temperature-dependent internalization of radiolabelled [DOTA0,Tyr3]octreotide in somatostatin receptor subtype 2 (sst2)-positive rat pancreatic tumour cell lines. In this study we have investigated the effects of differences in the amount of injected peptide on tissue distribution of 111In-labelled [DOTA0, Tyr3]octreotide in normal, i.e. non-tumour-bearing, and CA20948 tumour-bearing rats. This was done in order to find the amount of peptide at which the highest uptake in target tissues is achieved, and thereby to increase the potential of radionuclide therapy while simultaneously ensuring the lowest possible radiotoxicity in normal organs. Uptake of radiolabelled [DOTA0,Tyr3]octreotide in sst2-positive organs showed different bell-shaped functions of the amount of injected peptide, being highest at 0.05 (adrenals), 0.05-0. 1 (pituitary and stomach) and 0.25 (pancreas) microg. Uptake in the tumour was highest at 0.5 microg injected peptide. The highest uptake was found at peptide amounts that were lower than those reported for [111In-DTPA0]octreotide ((D-Phe-c(Cys-Phe-D-Trp-Lys-Thr-Cys)-Thr(ol), DTPA = diethylene-triamine-penta-acetic acid), consistent with the higher receptor affinity of the first compound. Our observations of mass-dependent differences in uptake of radiolabelled [DOTA0, Tyr3]octreotide, being the resultant of a positive effect of increasing amounts of peptide on, for example, receptor clustering and a negative effect of receptor saturation, are of consequence for rat radionuclide therapy studies with radiolabelled peptides and may also be of consequence for human radionuclide therapy studies with this compound.


Subject(s)
Indium Radioisotopes , Octreotide/analogs & derivatives , Pancreatic Neoplasms/diagnostic imaging , Radiopharmaceuticals , Animals , Octreotide/pharmacokinetics , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Inbred Lew , Tissue Distribution
17.
Clin Cancer Res ; 5(5): 1025-33, 1999 May.
Article in English | MEDLINE | ID: mdl-10353735

ABSTRACT

Human gliomas, especially of low-grade type, have been shown to express high-affinity somatostatin receptor type 2 (J-C. Reubi et al., Am. J. Pathol, 134: 337-344, 1989). We enrolled seven low-grade and four anaplastic glioma patients in a pilot study using the diffusible peptidic vector 90Y-labeled DOTA0-D-Phe1-Tyr3-octreotide (DOTATOC) for receptor targeting. The radiopharmakon was locoregionally injected into a stereotactically inserted Port-a-cath. DOTATOC competes specifically with somatostatin binding to somatostatin receptor type 2 in the low nanomolar range as shown by a displacement curve of 125I-[Tyr3]-octreotide in tumor tissue sections. Diagnostic (111)In-labeled DOTATOC-scintigraphy following local injection displayed homogeneous to nodular intratumoral vector distribution. The cumulative activity of regionally injected peptide-bound 90Y amounted to 370-3300 MBq, which is equivalent to an effective dose range between 60 +/- 15 and 550 +/- 110 Gy. Activity was injected in one to four fractions according to tumor volumes; 1110 MBq of 90Y-labeled DOTATOC was the maximum activity per single injection. We obtained six disease stabilizations and shrinking of a cystic low-grade astrocytoma component. The only toxicity observed was secondary perifocal edema. The activity:dose ratio (MBq:Gy) represents a measure for the stability of peptide retention in receptor-positive tissue and might predict the clinical course. We conclude that SR-positive human gliomas, especially of low-grade type, can be successfully targeted by intratumoral injection of the metabolically stable small regulatory peptide DOTATOC.


Subject(s)
Brain Neoplasms/radiotherapy , Glioma/radiotherapy , Neoplasm Proteins/antagonists & inhibitors , Octreotide/analogs & derivatives , Radioisotopes/therapeutic use , Radiopharmaceuticals/therapeutic use , Receptors, Somatostatin/antagonists & inhibitors , Ytterbium/therapeutic use , Adult , Astrocytoma/metabolism , Astrocytoma/pathology , Astrocytoma/radiotherapy , Binding, Competitive , Brain Edema/chemically induced , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Diffusion , Disease Progression , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/radiotherapy , Glioma/metabolism , Glioma/pathology , Humans , Injections, Intralesional , Male , Middle Aged , Octreotide/administration & dosage , Octreotide/adverse effects , Octreotide/pharmacokinetics , Octreotide/therapeutic use , Oligodendroglioma/metabolism , Oligodendroglioma/pathology , Oligodendroglioma/radiotherapy , Pilot Projects , Radioisotopes/administration & dosage , Radioisotopes/adverse effects , Radioisotopes/pharmacokinetics , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/pharmacokinetics , Radiotherapy Dosage , Stereotaxic Techniques , Tissue Distribution , Ytterbium/administration & dosage , Ytterbium/adverse effects , Ytterbium/pharmacokinetics
18.
J Nucl Med ; 40(5): 762-7, 1999 May.
Article in English | MEDLINE | ID: mdl-10319747

ABSTRACT

UNLABELLED: Scintigraphy with [111In-diethylenetriamine pentaacetic acid0-D-Phe1]-octreotide (DTPAOC) is used to demonstrate neuroendocrine and other somatostatin-receptor-positive tumors. Despite encouraging results, this 111In-labeled compound is not well suited for peptide-receptor-mediated radiotherapy of somatostatin-receptor-positive tumors. Another somatostatin analog, [1,4,7,10-tetraazacyclododecane-N,N',N",N'''-tetraacetic acid0, D-Phe1, Tyr3]-octreotide (DOTATOC), can be labeled with the beta-emitter 90Y in a stable manner. METHODS: We compared the distribution, kinetics and dosimetry of 111In-DTPAOC and 111In-DOTATOC in eight patients to predict the outcomes of these parameters in patients who will be treated with 90Y-DOTATOC. RESULTS: Serum radioactivity levels for the radiopharmaceuticals did not differ significantly 2-24 h after injection (P>0.05). Up to 2 h postinjection they were slightly, but significantly, lower after administration of 111In-DOTATOC (P < 0.01 at most time points). The percentage of peptide-bound radioactivity in serum did not differ after administration of either compound. Urinary excretion was significantly lower after administration of 111In-DOTATOC (P < 0.01). The visualization of known somatostatin-receptor-positive organs and tumors was clearer after administration of 111In-DOTATOC than after administration of 111In-DTPAOC. This was confirmed by significantly higher calculated uptakes in the pituitary gland and spleen. The uptake in the tumor sites did not differ significantly (P > 0.05), although in three of the four patients in whom tumor uptake could be calculated, it was higher after administration of 111In-DOTATOC. CONCLUSION: The distribution and excretion pattern of 111In-DOTATOC resembles that of 111In-DTPAOC, and the uptake in somatostatin-receptor-positive organs and most tumors is higher for 111In-DOTATOC. If 90Y-DOTATOC shows an uptake pattern similar to 111In-DOTATOC, it is a promising radiopharmaceutical for peptide-receptor-mediated radiotherapy in patients with somatostatin-receptor-positive tumors.


Subject(s)
Indium Radioisotopes , Neuroendocrine Tumors/diagnostic imaging , Octreotide/analogs & derivatives , Somatostatin/analogs & derivatives , Adult , Aged , Female , Humans , Indium Radioisotopes/pharmacokinetics , Male , Middle Aged , Neuroendocrine Tumors/metabolism , Octreotide/pharmacokinetics , Octreotide/therapeutic use , Radiation Dosage , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Receptors, Somatostatin/analysis , Somatostatin/pharmacokinetics , Tissue Distribution , Yttrium Radioisotopes/therapeutic use
19.
J Recept Signal Transduct Res ; 19(1-4): 167-80, 1999.
Article in English | MEDLINE | ID: mdl-10071756

ABSTRACT

The clinical importance of somatostatin type-2 receptors (SSTR2) and the study of novel analogues of somatostatin such as OctreoScan or [Tyr3]-octreotide containing DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) as metal chelator led us to develop a methodology to monitor the expression of SSTR2 on tumours of pancreatic origin (e.g. rat AR4-2J cancer cells). Usual binding assay protocols using the commercial [125I][Tyr1]-somatostatin radioligand failed, even in the presence of a cocktail of protease inhibitors with a broad spectrum of activity, possibly due to the high susceptibility of this tracer to proteases expressed in pancreatic cells. We prepared our own radioligand [125I][Tyr2]-octreotide which was shown to be much more resistant to degradation after incubation with AR4-2J plasma membranes. As expected, the increased stability of [125I][Tyr3]-octreotide was associated with good binding to SSTR2. Addition of appropriate protease inhibitors further increased the specific binding of [125I][Tyr3]-octreotide to AR4-2J plasma membranes without affecting the stability of the tracer, suggesting that the protease inhibitors also protect the integrity of SSTR2. Optimal conditions (time, temperature, medium) were developed for a binding assay in 96-well plates using AR4-2J plasma membranes in order to make the assay suitable for high-throughput analysis. This protocol was the basis for studying the in vivo regulation of SSTR2 expression in AR4-2J cells implanted into scid mice after exposure to different compounds.


Subject(s)
Radioligand Assay/methods , Receptors, Somatostatin/metabolism , Animals , Hormones/metabolism , Iodine Radioisotopes , Mice , Mice, SCID , Neoplasm Transplantation , Octreotide/analogs & derivatives , Octreotide/metabolism , Pancreatic Neoplasms/metabolism , Radiopharmaceuticals , Rats , Receptors, Somatostatin/analysis , Transplantation, Heterologous , Tumor Cells, Cultured
20.
Nuklearmedizin ; 38(1): 15-23, 1999.
Article in German | MEDLINE | ID: mdl-9987777

ABSTRACT

AIM: Recent in-vitro and in-vivo studies demonstrated a somatostatin receptor expression in some non-medullary thyroid carcinomas. In this study we investigated the somatostatin receptor status for this particular tumor entity in a larger patient group. SUBJECT AND METHODS: We compared 131-iodine with 111-In-pentetreotide scans in 24 patients with metastasizing, non-medullary thyroid cancer. The findings were correlated with other imaging modalities. Additionally, we performed receptor autoradiography in one patient, octreotide therapy in another patient and administration of 90-Y- and 111-In-DOTATOC in 2 consecutive patients. RESULTS: In the 15 patients with papillary or follicular carcinoma, 111-In-pentetreotide was inferior to 131-I in 8/15, equal in 1/15, and superior in 6/15 patients. In 8/9 of the patients with Hürthle cell carcinoma, metastases showed a 111-In-pentetreotide accumulation of various intensity, while 131-iodine scans were negative except for one patient. 111-In-pentetreotide was equal or superior compared to 201-Tl or 99m-Tc-sestamibi, but for the most part inferior in comparison with 18-F-FDG-PET. The findings of 111-In-pentetreotide scintigraphy correlated well with the receptor autoradiography and the accumulation of DOTATOC, but not with the therapeutic effect of "cold" octreotide on the thyroid cancer metastases. CONCLUSIONS: Several metastases of papillary and follicular carcinoma, and the majority of Hürthle cell cancer metastases can express somatostatin receptors. 111-In-pentetreotide scintigraphy is a promising tool for localization of metastases especially in Hürthle cell cancer or if PET is not available, and may be useful for selection of possible candidates, if therapeutic effective beta-emitting somatostatin analogues will be available for routine application.


Subject(s)
Adenocarcinoma, Follicular/diagnostic imaging , Carcinoma, Papillary/diagnostic imaging , Radiopharmaceuticals , Receptors, Somatostatin/analysis , Thyroid Neoplasms/diagnostic imaging , Adenocarcinoma, Follicular/metabolism , Adenocarcinoma, Follicular/pathology , Adult , Aged , Autoradiography , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Female , Humans , Indium Radioisotopes/pharmacokinetics , Iodine Radioisotopes/pharmacokinetics , Male , Middle Aged , Octreotide/analogs & derivatives , Octreotide/pharmacokinetics , Octreotide/therapeutic use , Radiopharmaceuticals/pharmacokinetics , Somatostatin/analogs & derivatives , Somatostatin/pharmacokinetics , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Tomography, Emission-Computed , Yttrium Radioisotopes
SELECTION OF CITATIONS
SEARCH DETAIL
...