Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Mol Ther Oncolytics ; 29: 44-58, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37180034

ABSTRACT

Tumor-specific overexpression of receptors enables a variety of targeted cancer therapies, exemplified by peptide-receptor radiotherapy (PRRT) for somatostatin receptor (SSTR)-positive neuroendocrine tumors. While effective, PRRT is restricted to tumors with SSTR overexpression. To overcome this limitation, we propose using oncolytic vaccinia virus (vvDD)-mediated receptor gene transfer to permit molecular imaging and PRRT in tumors without endogenous SSTR overexpression, a strategy termed radiovirotherapy. We hypothesized that vvDD-SSTR combined with a radiolabeled somatostatin analog could be deployed as radiovirotherapy in a colorectal cancer peritoneal carcinomatosis model, producing tumor-specific radiopeptide accumulation. Following vvDD-SSTR and 177Lu-DOTATOC treatment, viral replication and cytotoxicity, as well as biodistribution, tumor uptake, and survival, were evaluated. Radiovirotherapy did not alter virus replication or biodistribution, but synergistically improved vvDD-SSTR-induced cell killing in a receptor-dependent manner and significantly increased the tumor-specific accumulation and tumor-to-blood ratio of 177Lu-DOTATOC, making tumors imageable by microSPECT/CT and causing no significant toxicity. 177Lu-DOTATOC significantly improved survival over virus alone when combined with vvDD-SSTR but not control virus. We have therefore demonstrated that vvDD-SSTR can convert receptor-negative tumors into receptor-positive tumors and facilitate molecular imaging and PRRT using radiolabeled somatostatin analogs. Radiovirotherapy represents a promising treatment strategy with potential applications in a wide range of cancers.

2.
Eur J Nucl Med Mol Imaging ; 49(4): 1113-1126, 2022 03.
Article in English | MEDLINE | ID: mdl-34625828

ABSTRACT

PURPOSE: The ߯-emitting terbium-161 also emits conversion and Auger electrons, which are believed to be effective in killing single cancer cells. Terbium-161 was applied with somatostatin receptor (SSTR) agonists that localize in the cytoplasm (DOTATOC) and cellular nucleus (DOTATOC-NLS) or with a SSTR antagonist that localizes at the cell membrane (DOTA-LM3). The aim was to identify the most favorable peptide/terbium-161 combination for the treatment of neuroendocrine neoplasms (NENs). METHODS: The capability of the 161Tb- and 177Lu-labeled somatostatin (SST) analogues to reduce viability and survival of SSTR-positive AR42J tumor cells was investigated in vitro. The radiopeptides' tissue distribution profiles were assessed in tumor-bearing mice. The efficacy of terbium-161 compared to lutetium-177 was investigated in therapy studies in mice using DOTATOC or DOTA-LM3, respectively. RESULTS: In vitro, [161Tb]Tb-DOTA-LM3 was 102-fold more potent than [177Lu]Lu-DOTA-LM3; however, 161Tb-labeled DOTATOC and DOTATOC-NLS were only 4- to fivefold more effective inhibiting tumor cell viability than their 177Lu-labeled counterparts. This result was confirmed in vivo and demonstrated that [161Tb]Tb-DOTA-LM3 was significantly more effective in delaying tumor growth than [177Lu]Lu-DOTA-LM3, thereby, prolonging survival of the mice. A therapeutic advantage of terbium-161 over lutetium-177 was also manifest when applied with DOTATOC. Since the nuclear localizing sequence (NLS) compromised the in vivo tissue distribution of DOTATOC-NLS, it was not used for therapy. CONCLUSION: The use of membrane-localizing DOTA-LM3 was beneficial and profited from the short-ranged electrons emitted by terbium-161. Based on these preclinical data, [161Tb]Tb-DOTA-LM3 may outperform the clinically employed [177Lu]Lu-DOTATOC for the treatment of patients with NENs.


Subject(s)
Neuroendocrine Tumors , Receptors, Somatostatin , Animals , Humans , Mice , Neuroendocrine Tumors/pathology , Octreotide , Radioisotopes , Receptors, Somatostatin/metabolism , Terbium/therapeutic use , Tissue Distribution
3.
Sci Rep ; 6: 18537, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26727049

ABSTRACT

Overexpresssion of HER-2 in the MDA-MB-435/LCC6 (LCC6(HER-2)) tumour model is associated with significantly increased hypoxia and reduced necrosis compared to isogenic control tumours (LCC6(Vector)); this difference was not related to tumour size or changes in vascular architecture. To further evaluate factors responsible for HER-2-associated changes in the tumour microenvironment, small animal magnetic resonance imaging (MRI) and positron emission tomography (PET) were used to measure tumour tissue perfusion and metabolism, respectively. The imaging data was further corroborated by analysis of molecular markers pertaining to energy homeostasis, and measurements of hypoxia and glucose consumption. The results showed a strong trend towards higher perfusion rates (~58% greater, p = 0.14), and significantly higher glucose uptake in LCC6(HER-2) (~2-fold greater; p = 0.025), relative to control tumours. The expression of proteins related to energy stress (P-AMPK, P-ACC) and glucose transporters (GLUT1) were lower in LCC6(HER-2) tumours (~2- and ~4-fold, respectively). The in vitro analysis showed that LCC6(HER-2) cells become more hypoxic in 1% oxygen and utilise significantly more glucose in normoxia compared to LCC6(Vector)cells (p < 0.005). Amalgamation of all the data points suggests a novel metabolic adaptation driven by HER-2 overexpression where higher oxygen and glucose metabolic rates produce rich energy supply but also a more hypoxic tumour mass.


Subject(s)
Energy Metabolism , Gene Expression , Neoplasms/genetics , Neoplasms/metabolism , Receptor, ErbB-2/genetics , Stress, Physiological , Animals , Cell Line, Tumor , Cell Survival/genetics , Disease Models, Animal , Female , Glucose/metabolism , Humans , Hypoxia , Magnetic Resonance Imaging , Neoplasms/diagnosis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Oxygen Consumption , Positron-Emission Tomography
5.
Eur J Nucl Med Mol Imaging ; 39(12): 1868-75, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22940856

ABSTRACT

PURPOSE: The biokinetics and dosimetry of (111)In-DOTA-NOC-ATE (NOCATE), a high-affinity ligand of SSTR-2 and SSTR-5, and (111)In-DTPA-octreotide (Octreoscan™, OCTREO) were compared in the same patients. METHODS: Seventeen patients (10 men, 7 women; mean age 60 years), referred for an OCTREO scan for imaging of a neuroendocrine tumour (15), thymoma (1) or medullary thyroid carcinoma (1), agreed to undergo a second study with NOCATE. Whole-body anterior-posterior scans were recorded 0.5 (100 % reference scan), 4, 24 and 48 h (17 patients) and 120 h (5 patients) after injection. In 16 patients the OCTREO scan (178 ± 15 MBq) was performed 16 ± 5 days before the NOCATE scan (108 ± 14 MBq) with identical timing; 1 patient had the NOCATE scan before the OCTREO scan. Blood samples were obtained from 14 patients 5 min to 48 h after injection. Activities expressed as percent of the initial (reference) activity in the whole body, lung, kidney, liver, spleen and blood were fitted to biexponential or single exponential functions. Dosimetry was performed using OLINDA/EXM. RESULTS: Initial whole-body, lung and kidney activities were similar, but retention of NOCATE was higher than that of OCTREO. Liver and spleen uptakes of NOCATE were higher from the start (p < 0.001) and remained so over time. Whole-body activity showed similar α and ß half-lives, but the ß fraction of NOCATE was double that of OCTREO. Blood T (1/2)ß for NOCATE was longer (19 vs. 6 h). As a result, the effective dose of NOCATE (105 µSv/MBq) exceeded that of OCTREO (52 µSv/MBq), and the latter result was similar to the ICRP 106 value of 54 µSv/MBq. Differential activity measurement in blood cells and plasma showed an average of <5 % of NOCATE and OCTREO attached to globular blood components. CONCLUSION: NOCATE showed a slower clearance from normal tissues and its effective dose was roughly double that of OCTREO.


Subject(s)
Neuroendocrine Tumors/diagnostic imaging , Octreotide/analogs & derivatives , Organometallic Compounds/pharmacokinetics , Pentetic Acid/analogs & derivatives , Radiopharmaceuticals/pharmacokinetics , Thymoma/diagnostic imaging , Thymus Neoplasms/diagnostic imaging , Thyroid Neoplasms/diagnostic imaging , Adult , Aged , Carcinoma, Neuroendocrine , Female , Humans , Male , Middle Aged , Octreotide/pharmacokinetics , Pentetic Acid/pharmacokinetics , Radiometry , Radionuclide Imaging , Tissue Distribution
6.
Mol Endocrinol ; 24(2): 436-46, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20051480

ABSTRACT

Pasireotide (SOM230) is currently under clinical evaluation as a successor compound to octreotide for the treatment of acromegaly, Cushing's disease, and carcinoid tumors. Whereas octreotide acts primarily via the sst(2A) somatostatin receptor, pasireotide was designed to exhibit octreotide-like sst(2A) activity combined with enhanced binding to other somatostatin receptor subtypes. In the present study, we used phophosite-specific antibodies to examine agonist-induced phosphorylation of the rat sst(2A) receptor. We show that somatostatin and octreotide stimulate the complete phosphorylation of a cluster of four threonine residues within the cytoplasmic (353)TTETQRT(359) motif in a variety of cultured cell lines in vitro as well as in intact animals in vivo. This phosphorylation was mediated by G protein-coupled receptor kinases (GRK) 2 and 3 and followed by rapid cointernalization of the receptor and ss-arrestin into the same endocytic vesicles. In contrast, pasireotide failed to promote substantial phosphorylation and internalization of the rat sst(2A) receptor. In the presence of octreotide or SS-14, SOM230 showed partial agonist behavior, inhibiting phosphorylation, and internalization of sst(2A). Upon overexpression of GRK2 or GRK3, pasireotide stimulated selective phosphorylation of Thr356 and Thr359 but not of Thr353 or Thr354 within the (353)TTETQRT(359) motif. Pasireotide-mediated phosphorylation led to the formation of relatively unstable beta-arrestin-sst(2A) complexes that dissociated at or near the plasma membrane. Thus, octreotide and pasireotide are equally active in inducing classical G protein-dependent signaling via the sst(2A) somatostatin receptor. Yet, we find that they promote strikingly different patterns of sst(2A) receptor phosphorylation and, hence, stimulate functionally distinct pools of beta-arrestin.


Subject(s)
Octreotide/pharmacology , Protein Processing, Post-Translational/drug effects , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Somatostatin/agonists , Animals , Antibodies, Phospho-Specific/isolation & purification , Arrestins/genetics , Arrestins/metabolism , Cell Line , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Endocytosis/drug effects , Humans , Ligands , Male , Pancreas/drug effects , Pancreas/pathology , Phosphorylation/drug effects , Pituitary Gland/drug effects , Pituitary Gland/pathology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Processing, Post-Translational/genetics , Rats , Rats, Wistar , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors , Receptors, Somatostatin/genetics , Somatostatin/antagonists & inhibitors , Somatostatin/pharmacology , Threonine/genetics , Threonine/metabolism , beta-Adrenergic Receptor Kinases/antagonists & inhibitors , beta-Adrenergic Receptor Kinases/genetics , beta-Adrenergic Receptor Kinases/metabolism , beta-Arrestins
7.
J Pharm Sci ; 99(6): 2839-53, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20091826

ABSTRACT

Cationic liposomes exhibit a propensity to selectively target tumor-associated blood vessels demonstrating potential value as anti-cancer drug delivery vehicles. Their utility however, is hampered by their biological instability and rapid elimination following i.v. administration. Efforts to circumvent rapid plasma elimination have, to date, focused on decreasing cationic lipid content and incorporating polyethylene glycol (PEG)-modified lipids. In this study we wanted to determine whether highly charged cationic liposomes with surface-associated PEG could be designed to exhibit extended circulation lifetimes, while retaining tumor vascular targeting properties in an HT29 colorectal cancer xenograft model. Cationic liposomes prepared of DSPC, cationic lipids (DODAC, DOTAP, or DC-CHOL), and DSPE-PEG(2000) were studied. Our results demonstrate that formulations prepared with 50 mol% DODAC or DC-CHOL, and 20 mol% DSPE-PEG(2000) exhibited circulation half-lives ranging from 6.5 to 12.5 h. Biodistribution studies demonstrated that DC-CHOL formulations prepared with DSPE-PEG(2000) accumulated threefold higher in s.c. HT29 tumors than its PEG-free counterpart. Fluorescence microscopy studies suggested that the presence of DSPE-PEG(2000) did not adversely affect liposomal tumor vasculature targeting. We show for the first time that it is achievable to design highly charged, highly pegylated (20 mol% DSPE-PEG(2000)) cationic liposomes which exhibit both extended circulation lifetimes and tumor vascular targeting properties.


Subject(s)
Cations/therapeutic use , Drug Delivery Systems/methods , Liposomes/blood , Animals , Antineoplastic Agents , Chemistry, Pharmaceutical , Cholesterol/analogs & derivatives , Dosage Forms , Female , Lipids , Mice , Phosphatidylethanolamines , Polyethylene Glycols , Raloxifene Hydrochloride , Tissue Distribution
8.
Clin Cancer Res ; 14(7): 2019-27, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18381940

ABSTRACT

PURPOSE: Somatostatin receptor (sst) targeting is an established method to image and treat sst-positive tumors. Particularly, neuroendocrine tumors express the receptor subtype 2 in high density, but sst1, sst3, sst4, and sst5 are also expressed to some extent in different human tumors. Currently used targeting peptides mainly have sst2 affinity. We aimed at developing (radio)peptides that bind with high affinity to all receptor subtypes. EXPERIMENTAL DESIGN: Carbocyclic octapeptides were coupled with macrocyclic chelators for radiometal labeling. Affinity, internalization, and agonist potencies were determined on sst1- to sst5-expressing cell lines. Biodistribution was determined on nude mice bearing HEK-sst2 or AR4-2J and HEK-sst3 tumors. RESULTS: High affinity to all receptor subtypes was found. Y(III)-KE88 showed agonistic properties at all five sst receptor subtypes as it inhibits forskolin-stimulated cyclic AMP production. Surprisingly, very low or even absent sst2 receptor internalization was found compared with currently clinically established octapeptides, whereas the sst3 internalization was very efficient. Biodistribution studies of [(111)In]KE88 and [(67)Ga]KE88/[(68)Ga]KE88 reflected the in vitro data. In nude mice with s.c. implanted sst2 (HEK-sst2, AR4-2J)-expressing and sst3 (HEK-sst3)-expressing tumors, high and persistent uptake was found in sst3-expressing tumors, whereas the uptake in the sst2-expressing tumors was lower and showed fast washout. The kidney uptake was high but blockable by coinjection of lysine. CONCLUSION: This peptide family shows pansomatostatin potency. As radiopeptides, they are the first to show a full pansomatostatin profile. Despite some drawback, they should be useful for imaging sst2-expressing tumors with short-lived radiometals, such as (68)Ga, at early time points and for sst3-expressing tumors at later time points with longer-lived radiometals, such as (64)Cu or (86)Y.


Subject(s)
Peptides/chemistry , Peptides/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/chemical synthesis , Somatostatin/chemistry , Somatostatin/pharmacokinetics , Animals , Binding, Competitive , Cell Line , Chromatography, Affinity , Humans , Ligands , Mice , Mice, Nude , Neoplasms/diagnostic imaging , Peptides/pharmacokinetics , Protein Binding/physiology , Radioisotopes/chemistry , Radioisotopes/pharmacokinetics , Radioligand Assay , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Rats , Receptors, Somatostatin/metabolism , Structure-Activity Relationship , Tissue Distribution
9.
Bioconjug Chem ; 18(1): 84-92, 2007.
Article in English | MEDLINE | ID: mdl-17226960

ABSTRACT

Radiolabeled somatostatin analogues have been successfully used for targeted radiotherapy and for imaging of somatostatin receptor (sst1-5)-positive tumors. Nevertheless, these analogues are subject to improving their tumor-to-nontarget ratio to enhance their diagnostic or therapeutic properties, preventing nephrotoxicity. In order to understand the influence of lipophilicity and charge on the pharmacokinetic profile of [1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)]-somatostatin-based radioligands such as [DOTA,1-Nal3]-octreotide (DOTA-NOC), different spacers (X) based on 8-amino-3,6-dioxaoctanoic acid (PEG2), 15-amino-4,7,10,13-tetraoxapentadecanoic acid (PEG4), N-acetyl glucosamine (GlcNAc), triglycine, beta-alanine, aspartic acid, and lysine were introduced between the chelator DOTA and the peptide NOC. All DOTA-X-NOC conjugates were synthesized by Fmoc solid-phase synthesis. The partition coefficient (log D) at pH = 7.4 indicated that higher hydrophilicity than [111In-DOTA]-NOC was achieved with the introduction of the mentioned spacers, except with triglycine and beta-alanine. The high affinity of [InIII-DOTA]-NOC for human sst2 (hsst2) was preserved with the structural modifications, while an overall drop for hsst3 affinity was observed, except in the case of [InIII-DOTA]-beta-Ala-NOC. The new conjugates preserved the good affinity for hsst5, except for [InIII-DOTA]-Asn(GlcNAc)-NOC, which showed decreased affinity. A significant 1.2-fold improvement in the specific internalization rate in AR4-2J rat pancreatic tumor cells (sst2 receptor expression) at 4 h was achieved with the introduction of Asp as a spacer in the parent compound. In sst3-expressing HEK cells, the specific internalization rate at 4 h for [111In-DOTA]-NOC (13.1% +/- 0.3%) was maintained with [111In-DOTA]-beta-Ala-NOC (14.0% +/- 1.8%), but the remaining derivatives showed <2% specific internalization. Biodistribution studies were performed with Lewis rats bearing the AR4-2J rat pancreatic tumor. In comparison to [111In-DOTA]-NOC (2.96% +/- 0.48% IA/g), the specific uptake in the tumor at 4 h p.i. was significantly improved for the 111In-labeled sugar analogue (4.17% +/- 0.46% IA/g), which among all the new derivatives presented the best tumor-to-kidney ratio (1.9).


Subject(s)
Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 1-Ring/therapeutic use , Somatostatin/chemistry , Animals , Cell Line , Humans , Hydrophobic and Hydrophilic Interactions , Male , Molecular Structure , Neoplasms/drug therapy , Rats , Receptors, Somatostatin/metabolism , Serum , Somatostatin/analogs & derivatives , Somatostatin/metabolism
10.
Chem Biol ; 13(10): 1081-90, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17052612

ABSTRACT

The prototypes for tumor targeting with radiolabeled peptides are derivatives of somatostatin. Usually, they primarily have high affinity for somatostatin receptor subtype 2 (sst2), and they have moderate affinity for sst5. We aimed at developing analogs that recognize different somatostatin receptor subtypes for internal radiotherapy in order to extend the present range of accessible tumors. We synthesized DOTA-octapeptides based on octreotide by replacing Phe3 mainly with unnatural amino acids. The affinity profile was determined by using cell lines transfected with sst1-5. Internalization was determined by using AR42J, HEK-sst3, and HEK-sst5 cell lines, and biodistribution was studied in rat tumor models. Two of the derivatives thus obtained showed an improved binding affinity profile, enhanced internalization into cells expressing sst2 and sst3, respectively, and better tumor:kidney ratios in animals.


Subject(s)
Drug Design , Heterocyclic Compounds, 1-Ring/chemistry , Octreotide/chemical synthesis , Pancreatic Neoplasms/metabolism , Somatostatin/chemical synthesis , Animals , Cell Line , Disease Models, Animal , Drug Screening Assays, Antitumor , Heterocyclic Compounds, 1-Ring/chemical synthesis , Heterocyclic Compounds, 1-Ring/pharmacology , Humans , Indium Radioisotopes , Male , Molecular Conformation , Neoplasm Transplantation , Octreotide/analogs & derivatives , Octreotide/pharmacokinetics , Rats , Rats, Inbred Lew , Receptors, Somatostatin/drug effects , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Stereoisomerism , Time Factors , Tissue Distribution , Tumor Cells, Cultured , Yttrium Radioisotopes
11.
Proc Natl Acad Sci U S A ; 103(44): 16436-41, 2006 Oct 31.
Article in English | MEDLINE | ID: mdl-17056720

ABSTRACT

Targeting neuroendocrine tumors expressing somatostatin receptor subtypes (sst) with radiolabeled somatostatin agonists is an established diagnostic and therapeutic approach in oncology. While agonists readily internalize into tumor cells, permitting accumulation of radioactivity, radiolabeled antagonists do not, and they have not been considered for tumor targeting. The macrocyclic chelator 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) was coupled to two potent somatostatin receptor-selective peptide antagonists [NH(2)-CO-c(DCys-Phe-Tyr-DAgl(8)(Me,2-naphthoyl)-Lys-Thr-Phe-Cys)-OH (sst(3)-ODN-8) and a sst(2)-selective antagonist (sst(2)-ANT)], for labeling with (111/nat)In. (111/nat)In-DOTA-sst(3)-ODN-8 and (111/nat)In-DOTA-[4-NO(2)-Phe-c(DCys-Tyr-DTrp-Lys-Thr-Cys)-DTyr-NH(2)] ((111/nat)In-DOTA-sst(2)-ANT) showed high sst(3)- and sst(2)-binding affinity, respectively. They did not trigger sst(3) or sst(2) internalization but prevented agonist-stimulated internalization. (111)In-DOTA-sst(3)-ODN-8 and (111)In-DOTA-sst(2)-ANT were injected intravenously into mice bearing sst(3)- and sst(2)-expressing tumors, and their biodistribution was monitored. In the sst(3)-expressing tumors, strong accumulation of (111)In-DOTA-sst(3)-ODN-8 was observed, peaking at 1 h with 60% injected radioactivity per gram of tissue and remaining at a high level for >72 h. Excess of sst(3)-ODN-8 blocked uptake. As a control, the potent agonist (111)In-DOTA-[1-Nal(3)]-octreotide, with strong sst(3)-binding and internalization properties showed a much lower and shorter-lasting uptake in sst(3)-expressing tumors. Similarly, (111)In-DOTA-sst(2)-ANT was injected into mice bearing sst(2)-expressing tumors. Tumor uptake was considerably higher than with the highly potent sst(2)-selective agonist (111)In-diethylenetriaminepentaacetic acid-[Tyr(3),Thr(8)]-octreotide ((111)In-DTPA-TATE). Scatchard plots showed that antagonists labeled many more sites than agonists. Somatostatin antagonist radiotracers therefore are preferable over agonists for the in vivo targeting of sst(3)- or sst(2)-expressing tumors. Antagonist radioligands for other peptide receptors need to be evaluated in nuclear oncology as a result of this paradigm shift.


Subject(s)
Neoplasms/metabolism , Receptors, Peptide/metabolism , Receptors, Somatostatin/antagonists & inhibitors , Receptors, Somatostatin/metabolism , Animals , Humans , Mice , Mice, Nude , Molecular Structure , Neoplasms/pathology , Radioligand Assay , Receptors, Somatostatin/agonists , Receptors, Somatostatin/chemistry , Somatostatin/analogs & derivatives , Somatostatin/metabolism
12.
J Nucl Med ; 47(3): 502-11, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16513620

ABSTRACT

UNLABELLED: The uptake of radiolabeled somatostatin analogs by tumor cells through receptor-mediated internalization is a critical process for the in vivo targeting of tumoral somatostatin receptors. In the present study, the somatostatin receptor internalization induced by a variety of somatostatin analogs was measured with new immunocytochemical methods that allow characterization of trafficking of the somatostatin receptor subtype 2 (sst2), somatostatin receptor subtype 3 (sst3), and somatostatin receptor subtype 5 (sst5) in vitro at the protein level. METHODS: Human embryonic kidney 293 (HEK293) cells expressing the sst2, sst3, or the sst5 were used in a morphologic immunocytochemical internalization assay using specific sst2, sst3 and sst5 antibodies to qualitatively and quantitatively determine the capability of somatostatin agonists or antagonists to induce somatostatin receptor internalization. In addition, the internalization properties of a selection of these agonists have been compared and quantified in sst2-expressing CHO-K1 cells using an ELISA. RESULTS: Agonists with a high sst2-binding affinity were able to induce sst2 internalization in the HEK293 and CHO-K1 cell lines. New sst2 agonists, such as Y-DOTA-TATE, Y-DOTA-NOC, Lu-DOTA-BOC-ATE (where DOTA is 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid; TATE is [Tyr3, Thr8]-octreotide; NOC is [1-NaI3]-octreotide; and BOC-ATE is [BzThi3, Thr8]-octreotide), iodinated sugar-containing octreotide analogs, or BIM-23244 were considerably more potent in internalizing sst2 than was DTPA-octreotide (where DTPA is diethylenetriaminepentaacetic acid). Similarly, compounds with high sst3 affinity such as KE108 were able to induce sst3 internalization. In sst2- or sst3-expressing cell lines, agonist-induced receptor internalization was efficiently abolished by sst2- or sst3-selective antagonists, respectively. Antagonists alone had no effect on sst2 or sst3 internalization. We also showed that somatostatin-28 and somatostatin-14 can induce sst5 internalization. Unexpectedly, however, potent sst5 agonists such as KE108, BIM-23244, and L-817,818 were not able to induce sst5 internalization under the same conditions. CONCLUSION: Using sensitive and reproducible immunocytochemical methods, the ability of various somatostatin analogs to induce sst2, sst3, and sst5 internalization has been qualitatively and quantitatively determined. Whereas all agonists triggered sst2 and sst3 internalization, sst5 internalization was induced by natural somatostatin peptides but not by synthetic high-affinity sst5 agonists. Such assays will be of considerable help for the future characterization of ligands foreseen for nuclear medicine applications.


Subject(s)
Drug Delivery Systems/methods , Iodine Radioisotopes/pharmacokinetics , Kidney/diagnostic imaging , Receptors, Somatostatin/metabolism , Somatostatin/agonists , Somatostatin/antagonists & inhibitors , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Humans , Kidney/metabolism , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Somatostatin/analogs & derivatives
13.
J Nucl Med ; 46(12): 2097-103, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16330576

ABSTRACT

UNLABELLED: Auger electron-emitting radionuclides have potential for the therapy of small-size cancers because of their high level of cytotoxicity, low-energy, high linear energy transfer, and short-range biologic effectiveness. Biologic effects are critically dependent on the subcellular (and even subnuclear) localization of these radionuclides. Our goals were the design, synthesis, and in vitro preclinical assessment of new trifunctional conjugates of somatostatin that should aim at the nucleus and, therefore, ensure a longer retention time in the cell, a close approximation to the DNA, and the success of Auger electron emitters in targeted radionuclide therapy as well as also improve other targeted therapy strategies. METHODS: Three trifunctional derivatives of [(1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid)0,Tyr3]octreotide (DOTA-TOC) bearing the nuclear localization signal (NLS) (of simian virus 40 large-T antigen) PKKKRKV in 3 different positions relative to the somatostatin analog sequence were synthesized using solid and solution phase peptide synthesis. These compounds together with DOTA-TOC and DOTA-NLS derivatives were labeled with 111In and tested for binding affinity, internalization, externalization, and nuclei localization on AR4-2J cells and on human embryonic cells stably transfected with sst2A. RESULTS: The two N-terminal derivatives preserved the sstr2A binding affinity. Their rate of internalization in all tested sstr-expressing cell lines was always superior for the trifunctional derivatives in comparison with the parent compound. A 6-fold increase in cellular retention from the total internalized activity and a 45-fold higher accumulation in the cell nuclei were found for one of the N-terminally modified compounds compared with [111In]-DOTA-TOC. The C-terminal conjugate was inferior in all tests compared with the parent compound. CONCLUSION: These encouraging results support our hypothesis that an additional NLS sequence to the DOTA-TOC could not only provide a better carrier for Auger electron-emitting radionuclides but also ensure a longer radioactivity retention time in the tumor cell.


Subject(s)
Electrons , Radiotherapy/methods , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Cell Line , Cell Nucleus/metabolism , Humans , Indium Radioisotopes/pharmacology , Kinetics , Neoplasms/radiotherapy , Octreotide/analogs & derivatives , Octreotide/pharmacology , Peptides/chemistry , Protein Structure, Tertiary , Radioisotopes/pharmacology , Receptors, G-Protein-Coupled/chemistry , Somatostatin/chemistry , Time Factors , Transfection
14.
Clin Cancer Res ; 11(3): 1136-45, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15709181

ABSTRACT

PURPOSE: Molecular imaging and targeted radiotherapy are emerging fields in nuclear oncology. Five human somatostatin receptors (hsstr1-hsstr5) are known to be overexpressed to some degree on various tumors, sstr2 being the most important one. Clinically used somatostatin based radiopeptides target exclusively sstr2. The aim of this study was to develop novel analogues with a broader sstr profile for diagnostic (positron emission tomography and single-photon emission computed tomography) and radiotherapeutic applications. EXPERIMENTAL DESIGN: The following promising structures emerged from a parallel synthetic approach: [1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA(0)),1-Nal(3),Thr(8)]-octreotide (1, DOTA-NOC-ATE) and [DOTA(0),BzThi(3),Thr(8)]-octreotide (2, DOTA-BOC-ATE). The conjugates were labeled with cold and radioactive (111)In. Pharmacologic properties were compared with [(111)In-DOTA,Tyr(3)]-octreotide ([(111)In-DOTA]-TOC). RESULTS: The receptor affinity profile showed high affinity of both peptides to hsstr2, hsstr3, and hsstr5 and some intermediate affinity to hsstr4, whereas [(111)In-DOTA]-TOC shows affinity only to sstr2. The internalization is fast in sstr2 expressing AR4-2J and in transfected sstr3 expressing human embryonic kidney 293 cells. Both radiopeptides internalize much more efficiently than [(111)In-DOTA]-TOC. Animal biodistribution studies showed very high and specific uptake of [(111)In]-1 and [(111)In]-2 in s.c. implanted AR4-2J tumors (Lewis rats) and in somatostatin receptor expressing normal tissues. The uptake was at least 2-fold higher in these tissues and in the tumor compared with [(111)In-DOTA]-TOC. In addition, the kidney uptake was significantly lower for both radiopeptides. CONCLUSIONS: These data suggest that the novel radiopeptides are superior to [(111)In/(90)Y-DOTA]-TOC and show great promise for the clinical application in the imaging of somatostatin receptor-positive tumors and their targeted radiotherapy.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacokinetics , Octreotide/pharmacokinetics , Organometallic Compounds/pharmacokinetics , Animals , Antineoplastic Agents, Hormonal/chemistry , Antineoplastic Agents, Hormonal/metabolism , Binding, Competitive , CHO Cells , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Drug Evaluation, Preclinical , Endocytosis , Humans , Indium Radioisotopes/pharmacokinetics , Male , Molecular Structure , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/radiotherapy , Octreotide/analogs & derivatives , Octreotide/metabolism , Organometallic Compounds/metabolism , Positron-Emission Tomography , Rats , Rats, Inbred Lew , Receptors, Somatostatin/metabolism , Tissue Distribution , Tomography, Emission-Computed, Single-Photon
16.
Met Ions Biol Syst ; 42: 109-42, 2004.
Article in English | MEDLINE | ID: mdl-15206101

ABSTRACT

Radiometallo-labeled analogs of somatostatin have shown great benefit in the in vivo localization and targeted radiotherapy of human tumors. The progress and innovation in this clinical application came from the change in strategy, leaving the most widely used radiohalogens for a coordination chemistry approach. The use of chelators appended to the biologically active peptide which convey high thermodynamic and kinetic stability to the radiopeptides did not only improve the pharmacokinetics and pharmacodynamics of the molecules, but surprisingly the biological potency as well. The most urgent problem to be solved in the field is to improve the kidney clearance of the radiopeptides. The kidney turned out to be the dose limiting organ in this type of targeted radiotherapy. Coordination chemical strategies have already paved the way to a successful clinical application; it is most likely that chelator modification will further help to improve the renal handling of radiometallo-peptides.


Subject(s)
Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Radioisotopes/therapeutic use , Animals , Disease Models, Animal , Humans , Metals/pharmacokinetics , Metals/therapeutic use , Peptides/pharmacokinetics , Peptides/therapeutic use , Radioisotopes/pharmacokinetics , Radionuclide Imaging , Tissue Distribution
17.
Eur J Nucl Med Mol Imaging ; 30(10): 1338-47, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12937948

ABSTRACT

Earlier studies have shown that modification of the octapeptide octreotide in positions 3 and 8 may result in compounds with increased somatostatin receptor affinity that, if radiolabelled, display improved uptake in somatostatin receptor-positive tumours. The aim of a recent research study in our laboratory was to employ the parallel peptide synthesis approach by further exchanging the amino acid in position 3 of octreotide and coupling the macrocyclic chelator DOTA(1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) to these peptides for labelling with radiometals like gallium-67 or -68, indium-111, yttrium-90 and lutetium-177. The purpose was to find radiopeptides with an improved somatostatin receptor binding profile in order to extend the spectrum of targeted tumours. A first peptide, [111In,90Y-DOTA]-1-Nal3-octreotide (111In,90Y-DOTA-NOC), was isolated which showed an improved profile. InIII-DOTA-NOC exhibited the following IC50 values (nM) when studied in competition with [125I][Leu8, d-Trp22, Tyr25]somatostatin-28 (values for YIII-DOTA-NOC are shown in parentheses): sstr2, 2.9 +/- 0.1 (3.3 +/- 0.2); sstr3, 8 +/- 2 (26 +/- 1.9); sstr5, 11.2 +/- 3.5 (10.4 +/- 1.6). Affinity towards sstr1 and 4 was very low or absent. InIII-DOTA-NOC is superior to all somatostatin-based radiopeptides having this particular type of binding profile, including DOTA-lanreotide, and has three to four times higher binding affinity to sstr2 than InIII,YIII-DOTA-Tyr3-octreotide (InIII,YIII-DOTA-TOC). In addition, [111In]DOTA-NOC showed a specific and high rate of internalization into AR4-2J rat pancreatic tumour cells which, after 4 h, was about two times higher than that of [111In]DOTA-TOC and three times higher than that of [111In]DOTA-octreotide ([111In]DOTA-OC). The internalized radiopeptides were externalized intact upon 2 h of internalization followed by an acid wash. After 2-3 h of externalization a plateau is reached, indicating a steady-state situation explained by reactivation of the receptors followed by re-endocytosis. Biodistribution studies in CA 20948 tumour-bearing rats showed rapid clearance from all sstr-negative tissues except the kidneys. At 4 h the uptake of [111In]DOTA-NOC in the tumour and sstr-positive tissues, such as adrenals, stomach and pancreas, was three to four times higher than that of [111In]DOTA-TOC. Differential blocking studies indicate that this is at least partially due to the uptake mediated by sstr3 and sstr5. These very promising preclinical data justify the use of this new radiopeptide for imaging and potentially internal radiotherapy studies in patients.


Subject(s)
Biomarkers, Tumor/metabolism , Octreotide/analogs & derivatives , Organometallic Compounds/pharmacokinetics , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/metabolism , Receptors, Somatostatin/metabolism , Animals , Cell Line, Tumor , Drug Evaluation, Preclinical , Isotope Labeling/methods , Ligands , Male , Metabolic Clearance Rate , Metals/pharmacokinetics , Octreotide/pharmacokinetics , Organ Specificity , Protein Binding , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Inbred Lew , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL