Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Curr Pharm Des ; 19(25): 4647-72, 2013.
Article in English | MEDLINE | ID: mdl-23339739

ABSTRACT

Dysregulation of matrix metalloproteinase (MMP) activity can lead to a wide range of disease states such as atherosclerosis, inflammation or cancer. The ability to image MMP activity non-invasively in vivo, by radiolabelled synthetic inhibitors, would allow the characterization of atherosclerotic plaques, inflammatory lesions or tumors. Here we present an overview of radiolabelled MMP inhibitors (MMPIs) and MMP peptides for positron emission tomography (PET) and single photon emission computed tomography (SPECT) for the detection of proteolytic activity of MMPs. So far, most studies are at a preliminary stage; however, some hydroxamate-based tracers such as the peptidomimetics [¹¹¹In]-DTPA-RP782, [99mTc]-(HYNIC-RP805)(tricine)(TPPTS), or Marimastat-ArB[¹8F]F3 and the picolyl- benzenesulfonamide [¹²³I]I-HO-CGS 27023A identified specifically the enzymatic action of MMPs in animal models of various pathologies. The development of new compounds that may lead to novel tracers (e.g. modification of zinc-binding group, variation of substituents attached to the S1', S2' and S3' pockets of the MMP inhibitors) and the use of antibodies and cell penetrating peptides are also discussed. In general, preclinical studies with atherosclerosis models proved to be more successful than those with oncological models.


Subject(s)
Matrix Metalloproteinases/metabolism , Molecular Probes , Positron-Emission Tomography , Tomography, Emission-Computed, Single-Photon , Animals , Humans , Molecular Imaging
2.
Eur J Nucl Med Mol Imaging ; 38(12): 2117-24, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21792571

ABSTRACT

PURPOSE: We report on our experience in terms of eligibility, safety, response and survival for treatment of hepatocellular carcinoma (HCC) with (90)Y microspheres. Secondly, we investigated the urinary excretion of (90)Y following treatment. METHODS: We retrospectively reviewed all HCC patients referred to our department for (90)Y microsphere treatment. We recorded reasons for not proceeding to actual treatment. In case treatment was performed, we assessed the tolerance (Common Terminology Criteria for Adverse Events v3.0, CTCAE v3.0), the response [modified Response Evaluation Criteria in Solid Tumors (mRECIST) criteria] and long-term survival (Kaplan-Meier). The urinary excretion was estimated by 12-h urine collections post-injection for analysis in a gamma counter. RESULTS: Forty-three HCC patients were referred for radioembolization. Fourteen patients were excluded, mainly due to unfavourable (99m)Tc-macroaggregated albumin (MAA) distribution. Twenty-nine patients were treated with (90)Y microspheres (TheraSphere, mean activity 2.17 GBq). In four patients severe clinical adverse events were encountered, however only in one case clearly related to the therapy. Twenty patients were assessable by mRECIST: complete response in 15%, partial response in 35%, stable disease in 30% and progression in 20% were observed. A median survival of 12.3 months (95% confidence interval 9.4-15.2) was estimated. Concerning the substudy on urinary excretion, only 0.0025% of the administered activity was excreted in the urine within the first 12 h following TheraSphere. CONCLUSION: Following a strict workup before admitting patients to radioembolization with TheraSphere, we found good clinical tolerance in the vast majority of patients. Radiological response assessment yielded an overall response rate of 50%, when evaluated early following treatment. Urine analysis showed consistently only low activities of (90)Y excreted in the urine.


Subject(s)
Carcinoma, Hepatocellular/radiotherapy , Liver Neoplasms/radiotherapy , Yttrium Radioisotopes/administration & dosage , Yttrium Radioisotopes/urine , Aged , Aged, 80 and over , Belgium , Female , Humans , Injections, Intra-Arterial , Liver Neoplasms/complications , Longitudinal Studies , Male , Microspheres , Middle Aged , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/urine , Treatment Outcome
3.
Mol Imaging ; 10(5): 340-58, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21521554

ABSTRACT

In this review, data on noninvasive imaging of apoptosis in oncology are reviewed. Imaging data available are presented in order of occurrence in time of enzymatic and morphologic events occurring during apoptosis. Available studies suggest that various radiopharmaceutical probes bear great potential for apoptosis imaging by means of positron emission tomography and single-photon emission computed tomography (SPECT). However, for several of these probes, thorough toxicologic studies are required before they can be applied in clinical studies. Both preclinical and clinical studies support the notion that 99mTc-hydrazinonicotinamide-annexin A5 and SPECT allow for noninvasive, repetitive, quantitative apoptosis imaging and for assessing tumor response as early as 24 hours following treatment instigation. Bioluminescence imaging and near-infrared fluorescence imaging have shown great potential in small-animal imaging, but their usefulness for in vivo imaging in humans is limited to structures superficially located in the human body. Although preclinical tumor-based data using high-frequency-ultrasonography (US) are promising, whether or not US will become a routinely clinically useful tool in the assessment of therapy response in oncology remains to be proven. The potential of magnetic resonance imaging (MRI) and magnetic resonance spectroscopy (MRS) for imaging late apoptotic processes is currently unclear. Neither 31P MRS nor 1H MRS signals seems to be a unique identifier for apoptosis. Although MRI-measured apparent diffusion coefficients are altered in response to therapies that induce apoptosis, they are also altered by nonapoptotic cell death, including necrosis and mitotic catastrophe. In the future, rapid progress in the field of apoptosis imaging in oncology is expected.


Subject(s)
Apoptosis/physiology , Diagnostic Imaging , Neoplasms/pathology , Animals , Female , Humans , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Neoplasms/chemistry , Tomography, Emission-Computed
4.
Nucl Med Biol ; 37(8): 965-75, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21055628

ABSTRACT

INTRODUCTION: Apoptosis is one of the mechanisms behind successful chemotherapy and radiation treatment. Radiolabeled annexin A5 has been demonstrated to be a successful tool in the detection of apoptosis following chemotherapy in vivo. METHODS: His-tagged annexin A5 was labeled with [(99m)Tc]-tricarbonyl and evaluated as apoptosis imaging radiotracer in vitro and in vivo. The binding of the radiotracer was evaluated in Colo205 cells stimulated with 5-FU (1 mM) for 4 and 24 h, and confirmed by flow cytometry. Biodistribution and dosimetric studies were performed in healthy nude mice (n=5) via planar scintigraphy. [(99m)Tc]-(CO)(3) His-annexin A5 was also evaluated for in vivo imaging of spontaneous apoptosis in Colo205-bearing mice (n=12). RESULTS: The labeling procedure yielded a compound with 95-99% radiochemical purity and good in vitro stability. In vitro binding experiments indicated that the radiotracer retained its PS-binding activity. [(99m)Tc]-(CO)(3) His-annexin A5 rapidly cleared from the blood and predominantly accumulated in the kidneys. Absorbed dose (per organ) was found to be 116 ± 64 µGy/MBq for the kidneys and 10.38 ± 0.50 µGy/MBq for the liver. The effective dose was 7.00 ± 0.28 µSv/MBq. Spontaneous apoptosis in Colo205-bearing mice was visualised by [(99m)Tc]-(CO)(3) His-annexin A5 SPECT and correlated well with caspase-3 immunostaining (R=0.867, P<.01). CONCLUSION: [(99m)Tc]-(CO)(3) His-annexin A5 may be a useful novel radioligand for the in vivo detection of cell death associated with PS expression. A simple, noninvasive way of detecting apoptosis in vivo could have many applications including a better understanding of the extent and timing of apoptosis in response to cancer therapies and assessment of early tumor response.


Subject(s)
Annexin A5/chemistry , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Histidine/chemistry , Organotechnetium Compounds , Phosphatidylserines/metabolism , Animals , Annexin A5/metabolism , Annexin A5/pharmacokinetics , Apoptosis , Cell Line, Tumor , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Humans , Mice , Radiometry , Tomography, Emission-Computed, Single-Photon
5.
Cancer Biother Radiopharm ; 25(5): 511-20, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20854210

ABSTRACT

Matrix metalloproteinases (MMPs) are principal participants in tumor development. In addition to serve as a useful biochemical marker, MMP expression may also provide a target for the diagnostic in vivo imaging of tumors, using a radiolabeled inhibitor. This study investigates the use of membrane type 1 (MT1)-MMP as target for in vivo tumor diagnosis. Specific binding of the endogenous tissue inhibitor of metalloproteinase-2 (TIMP-2) to MT1-MMP has been previously described. In this study, biodistribution and imaging experiments were performed on MT1-MMP-overexpressing (S.1.5) and control (C.IV.3) tumor-inoculated mice using [(123)I]-recombinant human TIMP-2 (rhTIMP-2) as radioligand and [(123)I]-rhTIMP-1 as control. The expression profile was controlled in vitro and on tumor extracts. rhTIMP-2 as well as rhTIMP-1 were labeled using the Iodogen method and characterized. Biodistribution of [(123)I]-rhTIMP-2 showed a tumor uptake of 2.87% ± 1.58% ID/g at 3 hours postinjection in S.1.5. Tumor values of [(123)I]-rhTIMP-1 and [(123)I]-rhTIMP-2 evaluated in S.1.5 and C.IV.3, respectively, were significantly lower. Planar imaging revealed significant uptake of [(123)I]-rhTIMP-2 in S.1.5 compared with contralateral background areas. This could not be observed in C.IV.3 and with [(123)I]-rhTIMP-1 in S.1.5. All tumors were well established (200-800 mg). These results suggest that rhTIMP-2 holds potential for development of radiotracers for in vivo imaging in overexpressing MT1-MMP but not in similar tumors that do not express this protease.


Subject(s)
Iodine Radioisotopes , Matrix Metalloproteinase 1/analysis , Melanoma/pathology , Molecular Imaging/methods , Neoplasms/pathology , Radionuclide Imaging/methods , Tissue Inhibitor of Metalloproteinase-2 , Animals , Cell Line, Tumor , Humans , Iodine Radioisotopes/pharmacokinetics , Male , Matrix Metalloproteinase 1/metabolism , Melanoma/metabolism , Mice , Neoplasms/metabolism , Tissue Inhibitor of Metalloproteinase-2/pharmacokinetics
6.
J Mol Biol ; 371(4): 989-1006, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17599356

ABSTRACT

Human matrix metalloproteinase 9 (MMP-9), also called gelatinase B, is particularly involved in inflammatory processes, bone remodelling and wound healing, but is also implicated in pathological processes such as rheumatoid arthritis, atherosclerosis, tumour growth, and metastasis. We have prepared the inactive E402Q mutant of the truncated catalytic domain of human MMP-9 and co-crystallized it with active site-directed synthetic inhibitors of different binding types. Here, we present the X-ray structures of five MMP-9 complexes with gelatinase-specific, tight binding inhibitors: a phosphinic acid (AM-409), a pyrimidine-2,4,6-trione (RO-206-0222), two carboxylate (An-1 and MJ-24), and a trifluoromethyl hydroxamic acid inhibitor (MS-560). These compounds bind by making a compromise between optimal coordination of the catalytic zinc, favourable hydrogen bond formation in the active-site cleft, and accommodation of their large hydrophobic P1' groups in the slightly flexible S1' cavity, which exhibits distinct rotational conformations of the Pro421 carbonyl group in each complex. In all these structures, the side-chain of Arg424 located at the bottom of the S1' cavity is not defined in the electron density beyond C(gamma), indicating its mobility. However, we suggest that the mobile Arg424 side-chain partially blocks the S1' cavity, which might explain the weaker binding of most inhibitors with a long P1' side-chain for MMP-9 compared with the closely related MMP-2 (gelatinase A), which exhibits a short threonine side-chain at the equivalent position. These novel structural details should facilitate the design of more selective MMP-9 inhibitors.


Subject(s)
Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/metabolism , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Amino Acid Sequence , Arginine/genetics , Arginine/metabolism , Barbiturates/chemistry , Barbiturates/pharmacology , Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , Conserved Sequence , Crystallography, X-Ray , Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 1-Ring/pharmacology , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Matrix Metalloproteinase 9/genetics , Models, Molecular , Molecular Sequence Data , Phosphorous Acids/chemistry , Phosphorous Acids/pharmacology , Protein Binding , Protein Folding , Protein Structure, Tertiary , Sequence Alignment , Sulfones/chemistry , Sulfones/pharmacology
7.
Cancer Biother Radiopharm ; 21(5): 409-17, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17105415

ABSTRACT

During the past few years, several imaging probes targeting matrix metalloproteinases (MMPs) have been developed. Most of these probes have been validated in animal models. Overall, results derived from most of these studies have been disappointing. Whether or not this relates to shortcomings of the imaging probes used or to the set-up of the reported studies is currently unclear. Firstly, MMPs targeted in these studies, MMP-1, -2 and -9, are cell secreted and their expression is known to vary extensively within one tumor type, depending on the stage of development of the tumor and on the presence of naturally occurring TIMPs. Given the lack of data on the levels of MMP expression by incoculated tumor tissue at the time of imaging in most studies reported, it cannot be excluded that the negative results reported are, in fact, false-negative imaging results. Secondly, given that most of the agents used for imaging are intrinsically broad-spectrum agents, their higher affinity for specific subsets of MMPs does not necessarily imply that a positive imaging result also corresponds to overexpression of specific subsets of MMPs, as suggested in some papers published. Accordingly, well-characterized tumor models need to be developed for the purpose of validating currently available, as well as future, MMP-imaging probes. So far, only 111In-DTPA-N-TIMP-2 has been injected in patients, respectively suffering from Kaposi Sarcoma. Imaging results obtained with this agent proved disappointing. Imaging results obtained with other MMP-targeting probes in patients are awaited.


Subject(s)
Diagnostic Imaging/methods , Matrix Metalloproteinases/analysis , Peptides/chemistry , Radiopharmaceuticals/chemistry , Animals , Humans , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/biosynthesis , Matrix Metalloproteinases/metabolism , Neoplasms/diagnostic imaging , Neoplasms/enzymology , Positron-Emission Tomography/methods , Tissue Inhibitor of Metalloproteinases/analysis , Tissue Inhibitor of Metalloproteinases/biosynthesis , Tomography, Emission-Computed, Single-Photon/methods
8.
Appl Radiat Isot ; 64(6): 677-85, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16546398

ABSTRACT

Among matrix metalloproteinases (MMPs), the subfamily of gelatinases (MMP-2, MMP-9) is of particular interest due to their ability to degrade type IV collagen and other non-fibrillar collagen domains and proteins such as fibronectin and laminin. Whilst malignant cells often over-express various MMPs, the gelatinases have been most consistently detected in malignant tissues and associated with tumor growth, metastatic potential and angiogenesis. Radiosynthesis of carboxylic (1') and hydroxamic (2') MMPIs resulted in radiochemical yields of 70 +/- 5% (n = 6) and 60 +/- 5% (n = 4), respectively. Evaluation in A549-inoculated athymic mice showed a tumor uptake of 2. 0+/- 0.7%ID/g (3 h p.i.), a tumor/blood ratio of 0.5 and a tumor/muscle ratio of 4.6 at 48 h p.i. for 1'. For compound 2' a tumor uptake of 0.7 +/- 0.2%ID/g (3 h p.i.), a tumor/blood ratio of 1.2 and a tumor/muscle ratio of 1.8 at 24 h p.i. were observed. HPLC analysis of the blood (plasma) showed no dehalogenation or other metabolites of 1' 2 h p.i. For compound 2', 65.4% of intact compound was found in the blood (plasma) and one polar metabolite (31%) was detected whereas in the tumor 91.8% of the accumulated activity was caused by intact compound and only 8.1% by the metabolite. Planar imaging, using a Toshiba GCA-9300A/hg SPECT camera, showed that tumor tissue could be visualized and that image quality improved by decreasing specific activity resulting in lower liver uptake, indicating some degree of saturable binding in the liver. In vivo evaluation of these radioiodinated carboxylic and hydroxamic MMP inhibitor tracers revealed that MMP inhibitors could have potential as tumor imaging agents, but that further research is necessary.


Subject(s)
Butyrates , Matrix Metalloproteinase Inhibitors , Neoplasms, Experimental/diagnostic imaging , Radiopharmaceuticals , Sulfonamides , Valine/chemistry , Animals , Biphenyl Compounds , Butyrates/chemical synthesis , Butyrates/pharmacology , Iodine Radioisotopes , Male , Mice , Mice, Nude , Neoplasms, Experimental/metabolism , Organ Specificity , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Serum Albumin , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology , Tomography, Emission-Computed, Single-Photon
9.
Nucl Med Biol ; 32(5): 431-6, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15982572

ABSTRACT

One of the research challenges in oncology is to develop new biochemical methods for noninvasive tumor therapy evaluation to determine whether the chemotherapeutics is effective. Vascular endothelial growth factor (VEGF) was labeled with radioiodine and evaluated in vitro as well as in vivo, using A2058, a melanoma cell line overexpressing VEGFR-1 and -2. Saturation binding analysis with [(125)I]-VEGF resulted in a K(d) of 0.1 nM. Internalization assays indicate the preserved ligand induced internalization and metabolization of the tracer. Biodistribution studies with [(123)I]-VEGF in wild type and A2058 tumor-bearing athymic mice showed low background activity and a tumor to reference tissue ratio of maximum 6.12. These results suggest that [(123)I]-VEGF is a potentially suitable tracer for tumor therapy evaluation.


Subject(s)
Iodine Radioisotopes , Melanoma/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line, Tumor , Humans , Male , Melanoma/drug therapy , Mice , Receptors, Vascular Endothelial Growth Factor/analysis , Tissue Distribution
10.
Appl Radiat Isot ; 62(6): 903-13, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15799868

ABSTRACT

Excess matrix degradation is one of the hallmarks of cancer and is an important factor in the process of tumor progression. It is implicated in invasion, metastasis, growth, angiogenesis and migration. Many characteristics of matrix metalloproteinases (MMPs) make them attractive therapeutic and diagnostic targets. MMP expression is upregulated at the tumor site, with localization of activity in the tumor or the surrounding stroma, providing a target for medical imaging techniques. Radioiodinated carboxylic and hydroxamic MMP inhibitors 2-(4'-[123I] iodo-biphenyl-4-sulfonylamino)-3-methyl-butyric acid (9) and 2-(4'-[123I] iodo-biphenyl-4-sulfonylamino)-3-methyl-butyramide (11), their unlabelled standards and precursors were synthesized. Radioiodination was conducted by electrophilic aromatic substitution of the tributylstannyl precursors and resulted in radiochemical yields of 70+/-5% (n=6) and 60+/-5% (n=4), respectively. In vitro zymography and enzyme assays showed for both hydroxamic acid and carboxylic acid compounds a good inhibition activity and a high selectivity for MMP-2. In vivo biodistribution in NMRI mice showed no long-term accumulation in organs and the possibility to accumulate in the tumor in a later phase of this study.


Subject(s)
Carboxylic Acids/chemical synthesis , Hydroxamic Acids/chemical synthesis , Matrix Metalloproteinase Inhibitors , Protease Inhibitors/chemical synthesis , Animals , Carboxylic Acids/pharmacokinetics , Carboxylic Acids/pharmacology , Drug Evaluation , Female , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/pharmacology , Magnetic Resonance Spectroscopy , Male , Mice , Protease Inhibitors/pharmacokinetics , Spectrometry, Mass, Electrospray Ionization
11.
Nucl Med Biol ; 32(3): 293-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15820765

ABSTRACT

[(123)I]-3-(4-iodobenzyl)-1,2,3,4-tetrahydro-8-hydroxychromeno[3,4-c]pyridin-5-one ([(123)I]-ITCP), a presumed radioligand for visualization of the dopamine D4 receptor by single photon emission computed tomography, was evaluated in vivo in mice and rabbits. This new radioiodinated tracer exhibited high brain uptake (3.64% injected dose per gram of tissue at 10 min p.i.) in mice. No significant amounts (less than 5%) of labeled metabolites were present in the brain, as demonstrated by a metabolite study. Regional brain distribution in rabbits showed atypical CNS uptake with consistently low values in the cortex and high values in other brain parts including cerebellum. Saturable binding was confirmed by a competition experiment with unlabeled product. Selectivity was assessed by competition experiments with a known dopamine D4 ligand and later with a sigma receptor ligand. Both experiments showed no observable competition. In conclusion, our findings indicate that [(123)I]-ITCP is neither a dopamine D4 receptor ligand nor a sigma receptor ligand. The exact nature of [(123)I]-ITCP binding in the brain remains to be elucidated.


Subject(s)
Benzopyrans/pharmacokinetics , Brain/diagnostic imaging , Brain/metabolism , Pyridones/pharmacokinetics , Receptors, Dopamine D2/metabolism , Tomography, Emission-Computed, Single-Photon/methods , Animals , Drug Evaluation, Preclinical , Male , Metabolic Clearance Rate , Mice , Organ Specificity , Rabbits , Radiopharmaceuticals/pharmacokinetics , Receptors, Dopamine D4 , Tissue Distribution
12.
Nucl Med Biol ; 32(2): 193-200, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15721765

ABSTRACT

In this study, in vivo evaluation in mice and rabbits of [123I]-4-iodo-N-(4-(4-(2-methoxyphenyl)-piperazin-1-yl)butyl)-benzamide ([123I]-BPB), a potential radioligand for visualisation of the sigma receptor by single photon emission computed tomography (SPECT), is reported. The compound possesses appropriate lipophilicity (log P=2.2) and binds sigma-1 and sigma-2 receptors (pKi=6.51 and 6.79, respectively). In mice, this new radioiodinated tracer exhibited high brain uptake (4.99% ID/g tissue at 10 min postinjection) and saturable binding (3.06% ID/g tissue at 10 min postinjection) as determined by pretreatment with unlabeled [123I]-BPB. A metabolite study demonstrated no (less than 5%) labeled metabolites in the brain. In rabbits, regional brain distribution was investigated and the tracer displayed high, homogeneous central nervous system uptake. Selectivity was assessed by competition experiments with known sigma ligands. Metabolite analysis showed no (less than 8%) labeled metabolites in the rabbit brain. In conclusion, our findings indicate that [123I]-BPB is not a suitable tracer for visualisation of D3 receptors while its potential for sigma receptor imaging is severely hampered by its affinity for dopamine receptors.


Subject(s)
Benzamides , Brain/diagnostic imaging , Brain/metabolism , Piperazines , Receptors, sigma/metabolism , Tomography, Emission-Computed, Single-Photon/methods , Animals , Benzamides/pharmacokinetics , Feasibility Studies , Male , Metabolic Clearance Rate , Mice , Organ Specificity , Piperazines/pharmacokinetics , Rabbits , Radiopharmaceuticals/pharmacokinetics , Species Specificity , Tissue Distribution
13.
Nucl Med Commun ; 26(2): 147-53, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15657509

ABSTRACT

OBJECTIVE: The goal of this study was to develop a 99mTc labelled human epidermal growth factor (hEGF) for the in-vivo prediction of cancer cell response to farnesyltransferase inhibitor (FTI) therapy. This is based on the observation that internalization of EGF receptors is inhibited by FTIs. METHODS: We describe the radiolabelling of 99mTc-hEGF using the hydrazinonicotinamide (HYNIC) linker. Binding characteristics of 99mTc-HYNIC-hEGF to the EGF receptor are explored using an in-vitro binding assay. Biodistribution data of the compound in mice and tumour uptake in LoVo tumour bearing athymic mice before and after farnesyltransferase inhibitor therapy are presented. RESULTS: No colloid formation was observed. Binding parameters and LoVo tumour uptake of 99mTc-HYNIC-hEGF did not differ significantly from directly labelled 123I-hEGF values. However, the biodistribution data of the 99mTc-HYNIC-hEGF showed higher uptake in liver and intestines and decreased stomach uptake compared to its 123I analogue. Eight hours after farnesyltransferase inhibitor therapy with R115777, LoVo tumour uptake of 99mTc-HYNIC-hEGF decreased significantly, as shown using planar gamma scintigraphy (the ratio tumour vs. thigh dropped from 2.54+/-0.83 to 0.99+/-0.18). These data confirm the results obtained using 123I-hEGF. CONCLUSION: These data suggest that 99mTc-HYNIC-hEGF is a promising and selective new radiotracer for in-vivo monitoring of the EGF receptor with SPECT. Moreover, 99mTc-HYNIC-hEGF is a possible tool for early therapy response prediction of farnesyltransferase inhibitors.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Epidermal Growth Factor/pharmacokinetics , ErbB Receptors/metabolism , Organotechnetium Compounds/pharmacokinetics , Quinolones/administration & dosage , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Colonic Neoplasms/diagnostic imaging , Epidermal Growth Factor/chemistry , Farnesyltranstransferase , Humans , Male , Metabolic Clearance Rate , Mice , Organ Specificity , Organotechnetium Compounds/chemistry , Radionuclide Imaging , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Treatment Outcome
14.
Cancer Biother Radiopharm ; 20(6): 639-47, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16398616

ABSTRACT

AIM: As a part of our efforts to use small organic matrix metalloproteinase (MMP) inhibitors with improved characteristics for the diagnosis and treatment of different kinds of tumor tissues, biphenylsulfonamide analogues were synthesized. This study reports on the in vivo biodistribution of iodine-123-labeled biphenylsulfonide and analogues in A549 lung carcinoma inoculated into athymic mice and the evaluation of their suitability as imaging agents using a single photon emission computed tomography (SPECT) camera. METHODS: The radioiodinated carboxylic and hydroxamic MMP inhibitors 2-(4'- [(123)I]iodobiphenyl-4-sulfonylamino)-3-(1H-indol-3-yl)-propionic acid (1') and 2-(4'-[(123)I]iodobiphenyl-4- sulfonylamino)-3-(1H-indol-3-yl)-propionamide (2') were synthesized by electrophilic aromatic substitution of the tributylstannyl derivatives. Planar gamma camera imaging was performed in nu/nu athymic mice bearing an A549 tumor using a Toshiba GCA-9300A/hg SPECT camera in planar mode equipped with a high-resolution, parallel-hole collimator. RESULTS: Radiosynthesis of (1') and (2') resulted in radiochemical yields of 60 +/- 5% (n +/- 3) and 70 +/- 5% (n = 6), respectively. Evaluation of tumors induced in athymic mice by the inoculation of non-small cell lung A549 carcinoma cells, showed a tumor uptake of 0.27-0.01 percent injected dose per gram (%ID/g) (3 hours-48 hours p.i.), a tumor-blood ratio of 0.7, a tumor-muscle ratio of 1.6, and a tumor-fat ratio of 0.5 at 24 hours (p.i.) for compound 1'. For compound 2' a tumor uptake of 0.7-0.04 %ID/g (3 hours-48 hours p.i.), a postinjection tumor-blood ratio of 1.2, a tumor-muscle ratio of 3.2, and a tumor-fat ratio of 2.4 at 48 hours p.i. was observed. SPECT evaluation confirmed the results obtained from biodistribution. CONCLUSION: In vivo evaluation of these radioiodinated carboxylic and hydroxamic MMP inhibitor tracers revealed that they do not appear suitable as tumor-imaging agents.


Subject(s)
Biphenyl Compounds , Matrix Metalloproteinase Inhibitors , Neoplasms/diagnostic imaging , Protease Inhibitors/pharmacology , Sulfonamides , Animals , Biphenyl Compounds/pharmacokinetics , Breast Neoplasms/diagnostic imaging , Cell Line, Tumor , Humans , Iodine Radioisotopes/pharmacokinetics , Mice , Mice, Nude , Protease Inhibitors/pharmacokinetics , Sulfonamides/pharmacokinetics , Tissue Distribution , Tomography, Emission-Computed, Single-Photon , Tryptophan
15.
Nucl Med Biol ; 31(5): 649-54, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15219284

ABSTRACT

Acetylhomotaurine was labeled with (11)C via N-acetylation with [(11)C]acetyl chloride. The synthesis yielded 48.2+/-3.8%, decay corrected to end of bombardment. The specific activity of the (radio)chemically pure product was 20.8+/-2.0 GBq/micromol at EOS. In vivo studies revealed a very fast clearance of the tracer from the blood and a uniform distribution in the different brain regions. Unfortunately, the poor passage through the blood brain barrier makes the tracer not suitable for PET studies.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Positron-Emission Tomography/methods , Taurine/analogs & derivatives , Taurine/pharmacology , Taurine/pharmacokinetics , Acamprosate , Alcohol Deterrents/pharmacology , Alcoholism/drug therapy , Animals , Brain/drug effects , Carbon Radioisotopes/chemistry , Carbon Radioisotopes/pharmacokinetics , Humans , Isotope Labeling/methods , Male , Metabolic Clearance Rate , Mice , Rabbits , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Taurine/chemical synthesis , Tissue Distribution , Treatment Outcome
16.
Nucl Med Biol ; 31(4): 459-68, 2004 May.
Article in English | MEDLINE | ID: mdl-15093816

ABSTRACT

Several studies have demonstrated a positive correlation between tumor progression and expression of extracellular proteinases such as matrix metalloproteinases (MMPs). MMP-2 and MMP-9 have become attractive targets for cancer research because of their increased expression in human malignant tumor tissues of various organs, providing a target for medical imaging techniques. Radioiodinated carboxylic and hydroxamic MMP inhibitors 2-(4'-[(123)I]iodo-biphenyl-4-sulfonylamino)-3-(1H-indol-3-yl)-propionic acid (9) and 2-(4'-[(123)I]iodo-biphenyl-4-sulfonylamino)-3-(1H-indol-3-yl)-propionamide (11) were synthesized by electrophilic aromatic substitution of the tributylstannyl derivatives and resulted in radiochemical yields of 60% +/- 5% (n = 3) and 70% +/- 5% (n = 6), respectively. In vitro zymography and enzyme assays showed high inhibition capacities of the inhibitors on gelatinases. In vivo biodistribution showed no long-term accumulation in organs and the possibility to accumulate in the tumor. These results warrant further studies of radioiodinated carboxylic and hydroxamic MMP inhibitor tracers as potential SPECT tumor imaging agents.


Subject(s)
Amides/pharmacokinetics , Iodine Radioisotopes/pharmacokinetics , Matrix Metalloproteinases/metabolism , Neoplasms/metabolism , Propionates/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Amides/chemistry , Animals , Biomarkers, Tumor/metabolism , Feasibility Studies , Iodine Radioisotopes/chemistry , Isotope Labeling/methods , Matrix Metalloproteinase Inhibitors , Metabolic Clearance Rate , Mice , Neoplasms/diagnostic imaging , Organ Specificity , Propionates/chemistry , Radionuclide Imaging , Radiopharmaceuticals/chemical synthesis , Tissue Distribution
17.
Eur J Nucl Med Mol Imaging ; 29(5): 699-709, 2002 May.
Article in English | MEDLINE | ID: mdl-11976811

ABSTRACT

Tumour angiogenesis is essential for growth, invasion and metastasis. Retrospective studies suggest that it is an independent prognostic factor that merits prospective validation. Furthermore, as tumour blood vessels show many differences from normal vessels and are not genetically unstable, they form a key area for therapy development. However, as anti-angiogenic therapy is primarily cytostatic and not cytotoxic, novel tailor-made specific end-points for treatment monitoring are required. In this regard, suitable molecular parameters for imaging tumour angiogenesis by means of nuclear medicine are being explored. Here we review current knowledge on the multiple pathways controlling tumour angiogenesis and try to assess which are the most clinically relevant for nuclear medicine imaging. Parameters that may influence the imaging potential of radiopharmaceuticals for angiogenesis imaging such as molecular weight and structure, their targeted location within the tumour and their usefulness in terms of specificity and constancy of the targeted molecular pathway are discussed.


Subject(s)
Neoplasms/blood supply , Neoplasms/diagnostic imaging , Neovascularization, Pathologic/diagnostic imaging , Radiopharmaceuticals , Angiogenesis Inhibitors/pharmacology , Humans , Neovascularization, Pathologic/physiopathology , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL