Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 147
Filter
1.
Eur J Nucl Med Mol Imaging ; 47(11): 2647-2655, 2020 10.
Article in English | MEDLINE | ID: mdl-32198613

ABSTRACT

PURPOSE: Angiogenesis plays an important role in the growth and metastatic spread of solid tumours and is characterised by the expression of integrins on the cell surface of endothelial cells. Radiolabelled RGD peptides specifically target angiogenesis-related αvß3 integrins, expressed on the activated endothelial cells of sprouting blood vessels. Here, we validated the feasibility of 68Ga[Ga]-DOTA-E-[c(RGDfK)]2 (68Ga-RGD) PET/CT to visualise angiogenesis in patients with oral squamous cell carcinoma (OSCC). METHODS: Ten patients with OSCC and scheduled for surgical resection including elective neck dissection received an intravenously administration of 68Ga-RGD (42 ± 8 µg; 214 ± 9 MBq). All patients subsequently underwent dynamic (n = 5) or static PET/CT imaging (n = 5) for 60 min or for 4 min/bed position at 30, 60 and 90 min after injection, respectively. Quantitative tracer uptake in tumour lesions was expressed as standardised uptake values (SUV). Additionally, tumour tissue was immunohistochemically stained for αvß3 integrin to assess the expression pattern. RESULTS: 68Ga-RGD tumour accumulation was observed in all patients. At 60 min post injection, tumour SUVmax ranged between 4.0 and 12.7. Tracer accumulation in tumour tissue plateaued at 10 min after injection. Uptake in background tissue did not change over time, resulting in tumour-to-muscle tissue of 6.4 ± 0.7 at 60 min post injection. CONCLUSIONS: 68Ga-RGD PET/CT of αvß3 integrin expression in OSCC patients is feasible with adequate tumour-to-background ratios. It will provide more insight in angiogenesis as a hallmark of the head and neck squamous cell carcinomas' tumour microenvironment. TRIAL REGISTRATION: https://eudract.ema.europa.eu no. 2015-000917-31.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Carcinoma, Squamous Cell/diagnostic imaging , Endothelial Cells , Gallium Radioisotopes , Head and Neck Neoplasms/diagnostic imaging , Humans , Integrin alphaVbeta3 , Mouth Neoplasms/diagnostic imaging , Positron Emission Tomography Computed Tomography , Squamous Cell Carcinoma of Head and Neck , Tumor Microenvironment
2.
Mol Pharm ; 11(11): 3930-7, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-24992368

ABSTRACT

Cholecystokinin-2 (CCK-2) receptors, overexpressed in cancer types such as small cell lung cancers (SCLC) and medullary thyroid carcinomas (MTC), may serve as targets for peptide receptor radionuclide imaging. A variety of CCK and gastrin analogues has been developed, but a major drawback is metabolic instability or high kidney uptake. The minigastrin analogue PP-F11 has previously been shown to be a promising peptide for imaging of CCK-2 receptor positive tumors and was therefore further evaluated. The peptide was conjugated with one of the macrocyclic chelators DOTA, NOTA, or NODAGA. The peptide conjugates were then radiolabeled with either (68)Ga, (64)Cu, or (111)In. All (radio)labeled compounds were evaluated in vitro (IC50) and in vivo (biodistribution and PET/CT and SPECT/CT imaging). IC50 values were in the low nanomolar range for all compounds (0.79-1.51 nM). In the biodistribution studies, (68)Ga- and (111)In-labeled peptides showed higher tumor-to-background ratios than the (64)Cu-labeled compounds. All tested radiolabeled compounds clearly visualized the CCK2 receptor positive tumor in PET or SPECT imaging. The chelator did not seem to affect in vivo behavior of the peptide for (111)In- and (68)Ga-labeled peptides. In contrast, the biodistribution of the (64)Cu-labeled peptides showed high uptake in the liver and in other organs, most likely caused by high blood levels, probably due to dissociation of (64)Cu from the chelator and subsequent transchelation to proteins. Based on the present study, (68)Ga-DOTA-PP-F11 might be a promising radiopharmaceutical for PET/CT imaging of CCK2 receptor expressing tumors such as MTC and SCLC. Clinical studies are warranted to investigate the potential of this tracer.


Subject(s)
Acetates/pharmacology , Copper Radioisotopes/chemistry , Gallium Radioisotopes/chemistry , Gastrins/chemistry , Heterocyclic Compounds, 1-Ring/pharmacology , Heterocyclic Compounds/pharmacology , Indium Radioisotopes/chemistry , Animals , Cell Line, Tumor , Chelating Agents/chemistry , Female , Humans , Inhibitory Concentration 50 , Mice , Mice, SCID , Multimodal Imaging , Neoplasm Transplantation , Peptides/chemistry , Positron-Emission Tomography , Radiopharmaceuticals/chemistry , Receptor, Cholecystokinin B/metabolism , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed
3.
Mol Imaging Biol ; 16(6): 747-55, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24849133

ABSTRACT

In oncology, sensitive and reliable detection tumor tissue is crucial to prevent recurrences and to improve surgical outcome. Currently, extensive research is focused on the use of radionuclides as well as fluorophores to provide real-time guidance during surgery to aid the surgeon in the identification of malignant tissue. Particularly, dual-modality approaches combining radionuclide and near-infrared fluorescence (NIRF) imaging have shown promising results in preclinical studies. Radionuclide imaging allows sensitive intra-operative localization of tumor lesions using a gamma probe, whereas NIRF imaging allows more accurate real-time tumor delineation. Consequently, both radionuclide and NIRF imaging might complement each other, and dual-modality image-guided surgery may overcome limitations of the currently used single-modality imaging techniques. In this review, a comprehensive overview on recent preclinical advances in tumor-targeted radionuclide and fluorescence dual-modality imaging is provided. Subsequently, the clinical applicability of dual-modality image-guided surgery is discussed.


Subject(s)
Fluorescent Dyes , Multimodal Imaging/methods , Neoplasms/diagnosis , Radioisotopes , Animals , Fluorescent Dyes/chemistry , Humans , Neoplasms/diagnostic imaging , Neoplasms/pathology , Positron-Emission Tomography/methods , Radioisotopes/chemistry , Spectroscopy, Near-Infrared , Tomography, Emission-Computed, Single-Photon/methods
4.
Contrast Media Mol Imaging ; 9(3): 237-45, 2014.
Article in English | MEDLINE | ID: mdl-24700751

ABSTRACT

The purpose of this study was to assess the effect of bevacizumab on vasculature and hypoxia in a colorectal tumor model. Nude mice with subcutaneous LS174T tumors were treated with bevacizumab or saline. To assess tumor properties, separate groups of mice were imaged using (18) F-Fluoromisonidazole (FMISO) and (18) F-Fluorodeoxyglucose (FDG) positron emission tomography or magnetic resonance imaging before and 2, 6 and 10 days after the start of treatment. Tumors were harvested after imaging to determine hypoxia and vascular density immunohistochemically. The T2 * time increased significantly less in the bevacizumab group. FMISO uptake increased more over time in the control group. Vessel density significantly decreased in the bevacizumab-treated group. The Carbonic anhydrase 9 (CAIX) and glucose uptake transporter 1 (GLUT1) fractions were higher in bevacizumab-treated tumors. However, the hypoxic fraction showed no significant difference. Bevacizumab led to shorter T2 * times and higher GLUT1 and CAIX expression, suggesting an increase in hypoxia and a higher glycolytic rate. This could be a mechanism of resistance to bevacizumab. The increase in hypoxia, however, could not be demonstrated by pimonidazole/FMISO, possibly because distribution of these tracers is hampered by bevacizumab-induced effects on vascular permeability and perfusion.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/pathology , Hypoxia/diagnosis , Neovascularization, Pathologic/diagnosis , Radiation-Sensitizing Agents , Angiogenesis Inhibitors/blood , Animals , Antibodies, Monoclonal, Humanized/blood , Bevacizumab , Colorectal Neoplasms/drug therapy , Enzyme-Linked Immunosorbent Assay , Female , Fluorodeoxyglucose F18 , Hypoxia/drug therapy , Hypoxia/metabolism , Immunoenzyme Techniques , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Mice, Nude , Misonidazole/analogs & derivatives , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Nitroimidazoles , Positron-Emission Tomography , Radiopharmaceuticals
5.
Bioconjug Chem ; 25(2): 335-41, 2014 Feb 19.
Article in English | MEDLINE | ID: mdl-24382090

ABSTRACT

A facile and rapid method to label peptides with (18)F based on chelation of [(18)F]AlF has been developed recently. Since this method requires heating to 100 °C, it cannot be used to label heat-sensitive proteins. Here, we used a two-step procedure to prepare (18)F-labeled heat-labile proteins using the [(18)F]AlF method based on hot maleimide conjugation. 1,4,7-Triazacyclononae-1,4-diacetate (NODA) containing a methyl phenylacetic acid group (MPA) functionalized with N-(2-aminoethyl)maleimide (EM) was used as a ligand which was labeled with [(18)F]AlF and then conjugated to the humanized anti-CEA antibody derivatives hMN-14-Fab', hMN-14-(scFv)2 (diabody), and a Dock-and-Lock engineered dimeric fragment hMN-14 Fab-AD2 at room temperature. The in vivo tumor targeting characteristics of the (18)F-labeled antibody derivatives were determined by PET imaging of mice with s.c. xenografts. NODA-MPAEM was radiolabeled with [(18)F]AlF at a specific activity of 29-39 MBq/nmol and a labeling efficiency of 94 ± 2%. The labeling efficiencies of the maleimide conjugation ranged from 70% to 77%, resulting in [(18)F]AlF-labeled hMN14-Fab', hMN14-Fab-AD2, or hMN14-diabody with a specific activity of 15-17 MBq/nmol. The radiolabeled conjugates were purified by gel filtration. For biodistribution and microPET imaging, antibody fragments were injected intravenously into BALB/c nude mice with s.c. CEA-expressing LS174T xenografts (right flank) and CEA-negative SK-RC-52 xenografts (left flank). All [(18)F]AlF-labeled conjugates showed specific uptake in the LS174T xenografts with a maximal tumor uptake of 4.73% ID/g at 4 h after injection. Uptake in CEA-negative SK-RC-52 xenografts was significantly lower. Tumors were clearly visualized on microPET images. Using a [(18)F]AlF-labeled maleimide functionalized chelator, antibody fragments could be radiofluorinated within 4 h at high specific activity. Here, we translated this method to preclinical PET imaging studies and showed feasibility of [(18)F]AlF-fluorinated hMN-14-Fab', [(18)F]AlF-hMN-14-Fab-AD2, and [(18)F]AlF-hMN-14-diabody for microPET imaging of CEA-expressing colonic cancer.


Subject(s)
Aluminum Compounds , Carcinoembryonic Antigen/chemistry , Fluorides , Fluorine Radioisotopes , Immunoglobulin Fragments , Neoplasms/diagnostic imaging , Positron-Emission Tomography , Animals , Cell Line, Tumor , Female , Heterografts , Humans , Immunoglobulin Fragments/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude
6.
J Tissue Eng Regen Med ; 8(6): 473-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-22777771

ABSTRACT

The present study aimed to provide information on material degradation and subsequent alveolar bone formation, using composites consisting of calcium phosphate cement (CPC) and poly(lactic-co-glycolic) acid (PLGA) with different microsphere morphology (hollow vs dense). In addition to the plain CPC-PLGA composites, loading the microspheres with the growth factors platelet-derived growth factor (PDGF) and insulin-like growth factor (IGF) was investigated. A total of four different CPC composites were applied into one-wall mandible bone defects in beagle dogs in order to evaluate them as candidates for alveolar bone regeneration. These composites consisted of CPC and hollow or dense PLGA microspheres, with or without the addition of PDGF-IGF growth factor combination (CPC-hPLGA, CPC-dPLGA, CPC-hPLGAGF , CPC-dPLGAGF ). Histological evaluation revealed significantly more bone formation in CPC-dPLGA than in CPC-hPLGA composites. The combination PDGF-IGF enhanced bone formation in CPC-hPLGA materials, but significantly more bone formation occurred when CPC-dPLGA was used, with or without the addition of growth factors. The findings demonstrated that CPC-dPLGA composite was the biologically superior material for use as an off-the-shelf material, due to its good biocompatibility, enhanced degradability and superior bone formation.


Subject(s)
Alveolar Process/physiology , Bone Cements/pharmacology , Bone Regeneration/drug effects , Calcium Phosphates/pharmacology , Alveolar Process/diagnostic imaging , Alveolar Process/drug effects , Alveolar Process/surgery , Animals , Dogs , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lactic Acid/pharmacology , Microspheres , Osteogenesis/drug effects , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Porosity , Radiography
7.
Curr Mol Med ; 13(10): 1506-22, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24206139

ABSTRACT

Breast cancer is a heterogeneous disease that can be subdivided into different groups, based on gene expression profiles or clinicopathological characteristics such as estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) expression. The expression of these receptors has both prognostic and predictive value. To improve breast cancer treatment, accurate methods for patient selection and response monitoring are required. One way to achieve this is by using molecular imaging, which can be used to measure the expression and accessibility of tumor-associated antigens in vivo, without the need of invasive biopsies. This review will focus on tumor-receptor imaging for currently approved targeted therapies and discuss the potential role of molecular imaging in the development of new therapeutic agents in breast cancer. Progress has been made in radionuclide imaging of ER, PR, HER2 and epidermal growth factor receptor (EGFR) expression, which can be used for treatment selection and response prediction to endocrine and other targeted therapy. Moreover, clinical studies have shown the feasibility for molecular imaging of the angiogenic pathway exploiting the expression of antigens closely associated with angiogenesis, such as αvß3 and VEGF. As proof of concept has been established, further research should be directed towards validation of the imaging methods and the impact on patient management.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Breast Neoplasms/diagnosis , Diagnostic Imaging , Drug Monitoring , Patient Selection , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Female , Humans , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism
8.
Br J Cancer ; 109(4): 934-42, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23860529

ABSTRACT

BACKGROUND: Radiolabelled antibody targeting of cancer is limited by slow blood clearance. Pretargeting with a non-radiolabelled bispecific monoclonal antibody (bsMAb) followed by a rapidly clearing radiolabelled hapten peptide improves tumour localisation. The primary goals of this first pretargeting study in patients with the anti-CEACAM5 × anti-hapten (HSG) bsMAb, TF2, and the radiolabelled hapten-peptide, IMP288, were to assess optimal pretargeting conditions and safety in patients with metastatic colorectal cancer (CRC). METHODS: Different dose schedules were studied in four cohorts of five patients: (1) shortening the interval between the bsMAb and peptide administration (5 days vs 1 day), (2) escalating the TF2 dose (from 75 to 150 mg), and (3) reducing the peptide dose (from 100 to 25 µg). After confirmation of tumour targeting by (111)In-IMP288, patients were treated with a bsMAb/(177)Lu-IMP288 cycle. RESULTS: Rapid and selective tumour targeting of the radiolabelled peptide was visualised within 1 h, with high tumour-to-tissue ratios (>20 at 24 h). Improved tumour targeting was achieved with a 1-day interval between the administration of the bsMAb and the peptide and with the 25-µg peptide dose. High (177)Lu-IMP288 doses (2.5-7.4 GBq) were well tolerated, with some manageable TF2 infusion reactions, and transient grades 3-4 thrombocytopaenia in 10% of the patients who received (177)Lu-IMP288. CONCLUSION: This phase I study demonstrates for the first time that pretargeting with bsMAb TF2 and radiolabelled IMP288 in patients with CEA-expressing CRC is feasible and safe. With this pretargeting method, tumours are specifically and rapidly targeted.


Subject(s)
Antibodies, Bispecific/therapeutic use , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/radiotherapy , Haptens/immunology , Heterocyclic Compounds, 1-Ring/therapeutic use , Oligopeptides/therapeutic use , Radioimmunotherapy/methods , Adult , Aged , Cohort Studies , Colorectal Neoplasms/pathology , Female , GPI-Linked Proteins/immunology , Humans , Male , Middle Aged
9.
Diabetologia ; 55(5): 1247-57, 2012 May.
Article in English | MEDLINE | ID: mdl-22358499

ABSTRACT

For more than a decade, researchers have been trying to develop non-invasive imaging techniques for the in vivo measurement of viable pancreatic beta cells. However, in spite of intense research efforts, only one tracer for positron emission tomography (PET) imaging is currently under clinical evaluation. To many diabetologists it may remain unclear why the imaging world struggles to develop an effective method for non-invasive beta cell imaging (BCI), which could be useful for both research and clinical purposes. Here, we provide a concise overview of the obstacles and challenges encountered on the way to such BCI, in both native and transplanted islets. We discuss the major difficulties posed by the anatomical and cell biological features of pancreatic islets, as well as the chemical and physical limits of the main imaging modalities, with special focus on PET, SPECT and MRI. We conclude by indicating new avenues for future research in the field, based on several remarkable recent results.


Subject(s)
Insulin-Secreting Cells/diagnostic imaging , Molecular Imaging/methods , Animals , Humans , Insulin-Secreting Cells/transplantation , Islets of Langerhans Transplantation/diagnostic imaging , Magnetic Resonance Imaging/methods , Mice , Positron-Emission Tomography/methods , Rats , Tomography, Emission-Computed, Single-Photon/methods
10.
J Control Release ; 159(1): 69-77, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-22245686

ABSTRACT

Bone morphogenic protein-2 (BMP-2) is a well-known growth factor that can improve the biological performance of bone substitute materials. BMP-2 produced via bacterial expression systems are non-glycosylated (ng) whereas native and recombinant equivalents produced in mammalian cell expression systems are glycosylated (g) proteins. ngBMP-2 is less soluble, resulting in lower BMP-2 release from carriers as used as bone substitute materials. This seems promising for reducing the amount of included growth factor in bone substitute materials. Hence, it was hypothesized that ngBMP-2 would induce formation of the same amount of bone at an ectopic site at lower dosage as gBMP-2. To that end, gBMP-2 and ngBMP-2 were firstly in vitro comparatively evaluated for biological activity and release from a calcium phosphate (CaP) based bone substitute material. Thereafter, an ectopic implantation model in rats was used, in which gBMP-2 and ngBMP2 were loaded in various dosages (2-20 µg/implant) on the CaP-based bone substitute material and implanted for 4 and 12 weeks. The results revealed that both the in vitro biological activity of and the in vitro release of ngBMP-2 are lower compared to gBMP2. Upon ectopic implantation, however, ngBMP-2 loaded implants induced more bone formation at lower concentrations from 4-weeks onward compared to gBMP-2 equivalents, indicating the value of ngBMP-2 as a potential alternative for mammalian produced recombinant BMP-2 for bone regenerative therapies.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Osteogenesis/drug effects , Alkaline Phosphatase/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Substitutes , Calcium Phosphates , Cell Line , Glycosylation , Male , Mice , Prostheses and Implants , Rats , Rats, Wistar
11.
Phys Med Biol ; 56(18): N183-93, 2011 Sep 21.
Article in English | MEDLINE | ID: mdl-21865622

ABSTRACT

Attenuation of photon flux on trajectories between the source and pinhole apertures affects the quantitative accuracy of reconstructed single-photon emission computed tomography (SPECT) images. We propose a Chang-based non-uniform attenuation correction (NUA-CT) for small-animal SPECT/CT with focusing pinhole collimation, and compare the quantitative accuracy with uniform Chang correction based on (i) body outlines extracted from x-ray CT (UA-CT) and (ii) on hand drawn body contours on the images obtained with three integrated optical cameras (UA-BC). Measurements in phantoms and rats containing known activities of isotopes were conducted for evaluation. In (125)I, (201)Tl, (99m)Tc and (111)In phantom experiments, average relative errors comparing to the gold standards measured in a dose calibrator were reduced to 5.5%, 6.8%, 4.9% and 2.8%, respectively, with NUA-CT. In animal studies, these errors were 2.1%, 3.3%, 2.0% and 2.0%, respectively. Differences in accuracy on average between results of NUA-CT, UA-CT and UA-BC were less than 2.3% in phantom studies and 3.1% in animal studies except for (125)I (3.6% and 5.1%, respectively). All methods tested provide reasonable attenuation correction and result in high quantitative accuracy. NUA-CT shows superior accuracy except for (125)I, where other factors may have more impact on the quantitative accuracy than the selected attenuation correction.


Subject(s)
Models, Animal , Phantoms, Imaging , Tomography, Emission-Computed, Single-Photon/methods , Animals , Iodine Radioisotopes , Photons , Rats , Reproducibility of Results , Scattering, Radiation , Sensitivity and Specificity , Technetium , Thallium Radioisotopes , Tomography, Emission-Computed, Single-Photon/instrumentation , Tomography, X-Ray Computed/methods
12.
Br J Surg ; 98(3): 436-41, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21254023

ABSTRACT

BACKGROUND: Radioimmunotherapy (RIT) has been shown to reduce the incidence of local recurrence of colorectal cancer in an experimental model. The aim of the present study was to investigate the survival benefit of RIT compared with chemotherapy. METHODS: An anastomosis was constructed in male Wag/Rij rats after intraluminal injection of CC531 tumour cells. The therapeutic efficacy of (177) Lu-labelled MG1 (single intravenous dose of 300 MBq/kg, n = 20) was compared with that of 5-fluorouracil-based chemotherapy (6 weekly cycles administered intraperitoneally, n = 20) and no treatment (n = 20). The primary endpoint was survival. Toxicity was monitored by bodyweight measurement. RESULTS: Both chemotherapy and RIT affected bodyweight, but the weight of animals in the RIT group remained significantly higher than in the chemotherapy group (median slope of bodyweight plot 0·48 versus 0·30 g/day; P < 0·001). Kaplan-Meier analysis showed that overall survival in the RIT and chemotherapy groups was significantly better than that in the control group (50 and 46 per cent versus 25 per cent respectively after 170 days; P = 0·024 and P = 0·029). Survival after treatment with RIT did not differ from that after chemotherapy (P = 0·911). CONCLUSION: RIT is as effective as chemotherapy in experimental colorectal cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/radiotherapy , Lutetium/therapeutic use , Radioimmunotherapy/methods , Radioisotopes/therapeutic use , Animals , Antibodies, Monoclonal , Male , Neoplasm Transplantation , Pentetic Acid/therapeutic use , Rats , Tumor Cells, Cultured
13.
Methods ; 48(2): 188-92, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19318127

ABSTRACT

Angiogenesis, the formation of new blood vessels, is a multi-step process regulated by pro- and anti-angiogenic factors. In order to grow and metastasize, tumors need a constant supply of oxygen and nutrients. For their growth beyond the size of 1-2 mm tumors are dependent on angiogenesis. Recently, various new anti-cancer agents (e.g. bevacizumab, sorafenib and sunitinib) have become available that specifically inhibit angiogenesis in tumors. To evaluate the effects of these new anti-angiogenic agents it would be of interest to scintigraphically image the process of angiogenesis in tumors. Several markers have been described that are preferentially expressed on newly formed blood vessels in tumors (alpha(v)beta(3) integrin, vascular endothelial growth factor and its receptor, prostate-specific membrane antigen) and in the extracellular matrix surrounding newly formed blood vessels (extra-domain B of fibronectin, Tenascin-C, matrix metalloproteinases, Robo-4). Several ligands targeting these markers have been tested as a radiotracer for imaging angiogenesis in tumors. The potential of some of these tracers such as radiolabeled cyclic RGD peptides and radiolabeled anti-PSMA antibodies has already been tested cancer patients, while for markers such as Robo-4 the ligand has not yet been identified. Here the preclinical and clinical studies with these new tracers to image angiogenesis in tumors are reviewed.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/diagnosis , Neovascularization, Pathologic/diagnosis , Animals , Antigens, Surface , Biomarkers, Tumor/metabolism , Extracellular Matrix Proteins , Glutamate Carboxypeptidase II , Humans , Integrin alphaVbeta3/metabolism , Matrix Metalloproteinases , Neoplasms/drug therapy , Neoplasms/physiopathology , Neovascularization, Pathologic/drug therapy , Oligopeptides/metabolism , Radiopharmaceuticals , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A
14.
Br J Surg ; 96(3): 314-21, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19224516

ABSTRACT

BACKGROUND: Radioimmunotherapy (RIT) is suitable for the treatment of microscopic residual disease and might therefore have an adjuvant role after colonic cancer surgery. METHODS: An anastomosis was constructed in male Wag/Rij rats after intraluminal injection of 2 x 10(6) CC531 tumour cells. The biodistribution of (111)In-labelled MG1 monoclonal antibody was assessed after intraperitoneal administration. The therapeutic efficacy of (177)Lu-labelled MG1 (74 MBq per rat), administered on the day of surgery (D0, n = 13) or 5 days later (D5, n = 13), was compared with that of carrier only (n = 13). The primary endpoint was perianastomotic tumour growth 28 days after surgery. RESULTS: (111)In-labelled MG1 preferentially accumulated in perianastomotic CC531 tumours. RIT resulted in a transient reduction in bodyweight in both treatment groups compared with controls, but there were no other signs of clinical discomfort. No macroscopic or microscopic perianastomotic tumour growth was found in eight of 11 animals in the D0 group and 11 of 13 in the D5 group, whereas 11 of 13 controls had macroscopic tumour (P = 0.011 and P = 0.001 respectively). CONCLUSION: This study suggests that RIT may be an effective adjuvant treatment for preventing local recurrence after resection of colonic cancer.


Subject(s)
Colonic Neoplasms/prevention & control , Neoplasm Recurrence, Local/prevention & control , Radioimmunotherapy , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Body Weight , Cell Line, Tumor , Colonic Neoplasms/pathology , Lutetium/pharmacokinetics , Lutetium/therapeutic use , Male , Neoplasm Recurrence, Local/pathology , Neoplasm Transplantation , Pentetic Acid/pharmacokinetics , Pentetic Acid/therapeutic use , Radioisotopes/pharmacokinetics , Radioisotopes/therapeutic use , Rats , Tumor Burden
15.
J Biomed Mater Res A ; 91(2): 614-22, 2009 Nov.
Article in English | MEDLINE | ID: mdl-18985784

ABSTRACT

To improve the in vivo resorption of an injectable calcium phosphate cement (CPC) for bone tissue engineering purposes, in previous experiments macroporosity was introduced by the in situ degradation of incorporated gelatin microspheres. Gelatin microspheres are also suitable carriers for osteoinductive drugs/growth factors, where release occurs concomitantly with degradation of the hydrogel. Introduction of these microspheres into CPC can alter the release pattern of the cement, which usually shows a marginal release of incorporated drugs. The goal of this study was to determine the in vitro release characteristics of gelatin microsphere CPC. For this, recombinant human TGF-beta1, bFGF, and BMP-2 were labeled with (125)I and loaded onto gelatin type A (porcine, pI = 7.0-9.0)/type B (bovine, pI = 4.5-5.0) microspheres for a short (instant) and longer (prolonged) time before mixing them with the cement. Radioactivity of the resulting 5 or 10 wt % gelatin microsphere CPC composites was monitored for 6 weeks when subjected to proteolytic medium. Drug-loaded CPC was used as control. Results showed that release pattern/efficiency of gelatin microsphere CPCs and CPC controls was highly dependent on the type of growth factor but unaffected by the amount of growth factor. With gelatin microsphere CPC, release was also dependent on the type of gelatin, total volume of incorporated microspheres, and loading method.


Subject(s)
Calcium Phosphates/chemistry , Gelatin/chemistry , Intercellular Signaling Peptides and Proteins/administration & dosage , Microspheres , Tissue Scaffolds/chemistry , Animals , Cattle , Humans , Porosity , Swine
16.
Curr Pharm Des ; 14(31): 3316-25, 2008.
Article in English | MEDLINE | ID: mdl-19075710

ABSTRACT

The current gold standard for imaging infection is radiolabeled white blood cells. For reasons of safety, simplicity and cost, it would be desirable to have a receptor-specific ligand that could be used for imaging infection and that would allow a differential diagnosis between sterile and septic inflammatory processes. Ligands tested for this purpose include labeled peptides ((99m)Tc-labeled f-Met-Leu-Phe, (123)I-IL-1ra, (99m)Tc-IL-8, (99m)Tc-P483H, (99m)Tc-P1827DS, (99m)Tc-C5a-des-Arg, (99m)Tc-RP517, (11)In-DPC11870-11), human polyclonal antibodies, monoclonal antibodies, antibody fragments, antimicrobial agents (ciprofloxacin, sparfloxacin, ceftizoxime, isoniazid, ethambutol, fluconazole, all labeled with (99m)Tc), antimicrobial peptides and bacteriophages. Radiolabeled antibodies represent a valid alternative to labeled white blood cells under specific conditions and indications. Radiolabeled antibiotics and antimicrobial peptides are promising candidates for an infection-specific radiopharmaceutical. However, at present we still need to investigate many basic aspects to better understand the mechanisms of binding and accumulation of this class of radiopharmaceuticals to bacteria.


Subject(s)
Infections/diagnostic imaging , Leukocytes/diagnostic imaging , Radiopharmaceuticals , Receptors, Cell Surface/metabolism , Animals , Humans , Ligands , Protein Binding , Radioligand Assay , Radionuclide Imaging
17.
Ann Surg Oncol ; 15(11): 3299-307, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18712445

ABSTRACT

BACKGROUND: Cytoreductive surgery (CS) followed by hyperthermic intraperitoneal chemotherapy (HIPEC) results in limited survival benefit and high morbidity and mortality rates in patients with peritoneal carcinomatosis (PC). Radioimmunotherapy (RIT) after CS of experimental PC has been shown to increase survival and compare favorably to HIPEC. The effects of RIT and HIPEC on wound healing after CS need to be determined. METHODS: PC was induced by intraperitoneal inoculation of CC-531 colon carcinoma cells in Wag/Rij rats. Animals were subjected to CS and anastomotic construction only or followed by RIT or HIPEC. RIT consisted of 74 MBq (177)lutetium-labeled anti-CC531 antibody MG1. HIPEC was performed by a closed abdominal perfusion technique using mitomycin-C during 60 minutes. Anastomotic and abdominal wall strength measurements were performed 3 and 5 days after surgery. RESULTS: At day 5, bursting pressure in ileum and colon anastomoses in the CS + HIPEC group, but not in the CS + RIT group, was lower (P < .01) than in the CS group. In the CS group, the colonic bursting site was more often outside the true anastomotic area (8 of 12 animals) than in the CS + HIPEC (1 of 12) and CS + RIT (5 of 12) groups. Abdominal wall strength in the CS + HIPEC group was significantly (P < .01) lower, at both measuring points, than that in both the CS group and the CS + RIT group. There was no difference between the latter. CONCLUSION: As adjuvant to CS, HIPEC showed a decrease in anastomotic and abdominal wall wound strength in a model of PC of CRC, whereas RIT did not.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Colonic Neoplasms/therapy , Hyperthermia, Induced , Mitomycin/therapeutic use , Peritoneal Neoplasms/therapy , Radioimmunotherapy , Wound Healing , Abdominal Wall/physiology , Abdominal Wall/surgery , Anastomosis, Surgical , Animals , Chemotherapy, Cancer, Regional Perfusion , Colonic Neoplasms/pathology , Colonic Neoplasms/surgery , Combined Modality Therapy , Disease Models, Animal , Gelatinases/metabolism , Hydroxyproline/metabolism , Injections, Intraperitoneal , Intestines/drug effects , Intestines/surgery , Lutetium/therapeutic use , Male , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/surgery , Rats , Rats, Inbred Strains , Survival Rate , Treatment Outcome
18.
Nuklearmedizin ; 47(3): 110-5, 2008.
Article in English | MEDLINE | ID: mdl-18493690

ABSTRACT

AIM: In peptide receptor radionuclide therapy (PRRT) using radiolabelled somatostatin analogues, kidney uptake of radiolabelled compound is the major dose-limiting factor. We studied the effects of Gelofusine (20 mg) and lysine (100 mg) and the combination of both after injection of therapeutic doses of radiolabelled [DOTA0,Tyr3]octreotate (60 MBq 111In or 555 MBq 177Lu labelled to 15 microg peptide) in male Lewis rats. METHODS: Kidney uptake was measured by single photon emission computed tomography (SPECT) scans with a four-headed multi-pinhole camera (NanoSPECT) at 24 h, 5 and 7 days p. i. and was quantified by volume of interest analysis. For validation the activity concentration in the dissected kidneys was also determined ex vivo using a gamma counter and a dose calibrator. RESULTS: Gelofusine and lysine both reduced kidney uptake of [177Lu-DOTA0,Tyr3]octreotate significantly by about 40% at all time points. The combination of Gelofusine and lysine resulted in a 62% inhibition of kidney uptake (p < 0.01 vs. lysine alone). A weak but significant dose-response relationship for Gelofusine, but not for lysine, was found. In a study with [111In-DOTA0,Tyr3]octreotate, conclusions drawn from NanoSPECT data were confirmed by biodistribution data. CONCLUSIONS: We conclude that rat kidney uptake of radiolabelled somatostatin analogues can be monitored for a longer period in the same animal using animal SPECT. Gelofusine and lysine had equal potential to reduce kidney uptake of therapeutic doses of [177Lu-DOTA0,Tyr3]octreotate. The combination of these compounds caused a significantly larger reduction than lysine or Gelofusine alone and may therefore offer new possibilities in PRRT. The NanoSPECT data were validated by standard biodistribution experiments.


Subject(s)
Kidney/diagnostic imaging , Kidney/metabolism , Lutetium , Lysine/pharmacology , Octreotide/analogs & derivatives , Organometallic Compounds , Polygeline/pharmacology , Animals , Biological Transport/drug effects , Drug Hypersensitivity , Humans , Kidney/drug effects , Lutetium/pharmacokinetics , Octreotide/pharmacokinetics , Organometallic Compounds/pharmacokinetics , Polygeline/adverse effects , Radioisotopes/pharmacokinetics , Rats , Tomography, Emission-Computed, Single-Photon/methods
19.
Int J Biol Markers ; 22(3): 167-71, 2007.
Article in English | MEDLINE | ID: mdl-17922458

ABSTRACT

The aim of this study was to develop and validate ELISAs for quantification of HAMA-IgM and HAMA-IgG in serum of patients with ovarian cancer who enrolled in a large international randomized phase III trial of intraperitoneal Yttrium-90-labeled HMFG1 murine monoclonal antibody therapy. The capture antibody of these 2 assays was the murine antibody HMFG1, while mouse anti-human IgM-HRP or mouse anti-human IgG(Fc)-HRP served as tracer antibodies. A pool of HAMA-positive serum samples was used to prepare a series of assay standards and another pool served as reference preparation. The analytical sensitivity of the HAMA-IgM assay was 2.5 arbitrary units per mL (AU/mL) and 4.7 AU/mL for the HAMA-IgG ELISA. Diluted serum samples showed good parallelism with the HAMA-IgM and HAMA-IgG standard dose-response curves. Within-assay coefficient of variation was 7.5% for HAMA-IgM and 6.5% for HAMA-IgG. Between-assay variation was 14.2% for HAMA-IgM and 15.3% for HAMA-IgG. The developed HAMA-IgM and HAMA-IgG ELISAs show satisfactory reliability criteria (sensitivity, parallelism and precision) and are suitable for monitoring of HAMA-IgM and HAMA-IgG responses in ovarian cancer patients. These ELISAs will be used to monitor the development of HAMAs in patients who received radioimmunotherapy with murine HMFG1.


Subject(s)
Antibodies, Monoclonal/immunology , Biomarkers, Tumor/blood , Enzyme-Linked Immunosorbent Assay/methods , Immunoglobulin G/blood , Immunoglobulin M/blood , Immunotoxins/therapeutic use , Ovarian Neoplasms/immunology , Animals , Antibodies, Anti-Idiotypic/immunology , Female , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Immunotoxins/immunology , Injections, Intraperitoneal , Mice , Ovarian Neoplasms/surgery , Ovarian Neoplasms/therapy , Radioimmunotherapy/methods , Sensitivity and Specificity , Yttrium Radioisotopes/therapeutic use
20.
Ann Surg Oncol ; 14(11): 3274-82, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17653591

ABSTRACT

BACKGROUND: Cytoreductive surgery (CS) followed by heated intraperitoneal chemotherapy (HIPEC) is considered the standard of care for the treatment of patients with peritoneal carcinomatosis (PC) of colorectal cancer (CRC). These surgical procedures result in a median survival of 2 years at the cost of considerable morbidity and mortality. In preclinical studies, radioimmunotherapy (RIT) improved survival after CS in a model of induced PC of colonic origin. In the present studies we aimed to compare the efficacy and toxicity of CS followed by adjuvant RIT in experimental PC to the standard of care, HIPEC. METHODS: PC was induced by intraperitoneal inoculation of CC-531 colon carcinoma cells in three groups of Wag/Rij rats. Treatment comprised CS only, CS + RIT or CS + HIPEC, immediately after surgery. RIT consisted of intraperitoneal administration of 74 MBq Lutetium-177 labeled MG1. HIPEC was performed by a closed abdomen perfusion technique using mitomycin C (16 mg/L during 60 minutes). The primary endpoint was survival. RESULTS: CS only or combined with RIT was well tolerated. Rats receiving CS + HIPEC were lethargic, suffered from diarrhea, and lost significantly more weight in the first postoperative week. Median survival of rats treated with CS + RIT was significantly longer than after CS alone (97 and 57 days, respectively, P < .004), whereas survival after CS + HIPEC or CS alone were not significantly different (76 and 57 days, respectively, P = .17). CONCLUSION: Survival after CS was significantly improved by RIT with Lutetium-177-MG1 in rats with PC of colorectal origin. Adjuvant HIPEC did not improve survival and was more toxic than adjuvant RIT.


Subject(s)
Colonic Neoplasms/pathology , Hyperthermia, Induced , Mitomycin/therapeutic use , Neoplasms, Experimental/therapy , Peritoneal Neoplasms/therapy , Radioimmunotherapy , Animals , Antibiotics, Antineoplastic/therapeutic use , Antibodies, Monoclonal/therapeutic use , Body Weight , Combined Modality Therapy , Disease Models, Animal , Infusions, Parenteral , Lutetium/therapeutic use , Neoplasms, Experimental/secondary , Neoplasms, Experimental/surgery , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/surgery , Rats , Rats, Inbred Strains , Survival Rate , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...