Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nucl Med Biol ; 102-103: 97-105, 2021.
Article in English | MEDLINE | ID: mdl-34743064

ABSTRACT

INTRODUCTION: Parathyroid hyperplasia is a disease characterized by overactive parathyroid glands secreting increased levels of parathyroid hormone. Surgical removal of the parathyroid glands is the standard treatment but requires precise pre-operative localization of the glands. However, currently available imaging modalities show limited sensitivity. Since positron emission tomography (PET) is a molecular imaging technique with high accuracy and sensitivity, our aim was to develop a new PET tracer for overactive parathyroid glands imaging by radiolabelling cinacalcet, a drug binding to the calcium-sensing receptor of the parathyroid glands. METHODS: [18F]Cinacalcet was synthesized by copper-catalysed [18F]trifluoromethylation of a boronic acid precursor using high molar activity [18F]fluoroform. Ex vivo biodistribution and metabolism were evaluated in 12 healthy male Wistar rats at 5, 15, 45 and 90 min. PET scans were performed at baseline and after blocking with NPS R-568. RESULTS: [18F]Cinacalcet was obtained in an overall radiosynthesis time of 1 h with a radiochemical purity of 98 ± 1%, a radiochemical yield of 8 ± 4% (overall, n = 7, corrected for decay) and a molar activity of 40 ± 11 GBq/µmol (n = 7, at EOS). The ex vivo biodistribution showed uptake in the thyroid and parathyroid glands as well as in other glands such as adrenals, salivary glands and pancreas. The tracer was rapidly cleared from the blood via liver and kidneys and showed fast metabolism. PET images confirmed uptake in the target organ. However, in a blocking study with NPS R-568 specific binding of [18F]cinacalcet to the CaSR could not be confirmed. CONCLUSIONS: [18F]Cinacalcet was successfully synthesized. First in vivo experiments in healthy rats showed uptake of the tracer in the target organ and fast metabolism, encouraging further in vivo evaluation of this tracer.


Subject(s)
Cinacalcet
2.
J Labelled Comp Radiopharm ; 64(12): 466-476, 2021 10.
Article in English | MEDLINE | ID: mdl-34382259

ABSTRACT

The trifluoromethyl group is a prominent motif in biologically active compounds and therefore of great interest for the labeling with the positron emitter fluorine-18 for positron emission tomography (PET) imaging. Multiple labeling strategies have been explored in the past; however, most of them suffer from low molar activity due to precursor degradation. In this study, the potential of 1-(difluoromethyl)-3-methyl-4-phenyl-1H-1,2,3-triazol-3-ium triflate as precursor for the synthesis of the [18 F]trifluoromethylation building block [18 F]fluoroform with high molar activity was investigated. The triazolium precursor was reacted under various conditions with [18 F]fluoride, providing [18 F]fluoroform with radiochemical yields (RCY) and molar activities (Am ) comparable and even superior with already existing methods. Highest molar activities (Am = 153 ± 14 GBq/µmol, dc, EOS) were observed for the automated procedure on the Neptis® perform module. Due to its easy handling and good RCY and Am in the [18 F]fluoroform synthesis, the triazolium precursor is a valuable alternative to already known precursors.


Subject(s)
Radiopharmaceuticals
3.
Chem Commun (Camb) ; 57(43): 5286-5289, 2021 May 27.
Article in English | MEDLINE | ID: mdl-33942818

ABSTRACT

This article describes the first synthesis and application of fluorine-18 labelled Ruppert-Prakash reagent [18F]Me3SiCF3. [18F]Me3SiCF3 was synthesized from [18F]fluoroform with radiochemical yields of 85-95% and radiochemical purities of >95% within 20 minutes. 18F-trifluoromethylated compounds were successfully prepared by reaction of [18F]Me3SiCF3 with benzaldehydes, acetophenones and benzophenones.

4.
J Nucl Med ; 60(5): 691-695, 2019 May.
Article in English | MEDLINE | ID: mdl-30530830

ABSTRACT

Dozens of monoclonal antibodies (mAbs) have been approved for clinical use, and hundreds more are under development. To support these developments and facilitate a personalized medicine approach, PET imaging and quantification of mAbs, after chelation with desferrioxamine B (DFO) and radiolabeling with 89Zr, has become attractive. Also, the use of 89Zr-mAbs in preclinical and clinical studies is expanding rapidly. Despite these rapid developments, 89Zr radiolabeling is still performed manually. Therefore, we aimed to develop a simple, fully automated, good-manufacturing-practice (GMP)-compliant production procedure for the 89Zr labeling of mAbs. Such procedures should increase the robustness and capacity of 89Zr-mAb production while minimizing the radiation dose to the operator. Here, the procedures for fully automated 89Zr-mAb production are described and applied to produce batches of 89Zr-DFO-N-suc-cetuximab and 89Zr-DFO-N-suc-rituximab suitable for clinical use. Both products had to meet the GMP-compliant quality standards with respect to yield, radiochemical purity, protein integrity, antigen binding, sterility, and endotoxin levels. Methods: Automated 89Zr labeling of mAbs was developed on a Scintomics GRP 2V module and comprised the following steps: reagent transfer to the 89Zr-containing reaction vial, mixing of the reagents followed by a 60-min reaction at room temperature to obtain optimal radiolabeling yields, and product purification using a PD-10 desalting column. Results: Radiochemical yields of 89Zr-DFO-N-suc-cetuximab and 89Zr-DFO-N-suc-rituximab were all more than 90% according to instant thin-layer chromatography. Isolated yields were 74.6% ± 2.0% and 62.6% ± 3.0% for 89Zr-DFO-N-suc-cetuximab and 89Zr-DFO-N-suc-rituximab, respectively, which are similar to isolated yields obtained using GMP protocols for manual 89Zr labeling of mAbs. To meet the GMP-compliant quality standards, only the radiochemically pure fractions were collected from PD-10, resulting in a lower isolated yield than the radiochemical yield according to instant thin-layer chromatography. The radiochemical purity and protein integrity were more than 95% for both products, and the antigen binding was 95.6% ± 0.6% and 87.1% ± 2.2% for 89Zr-DFO-N-suc-cetuximab and 89Zr-DFO-N-suc-rituximab, respectively. The products were sterile, and the endotoxin levels were within acceptable limits, allowing future clinical production using this procedure. Conclusion: Procedures for fully automated GMP-compliant production of 89Zr-mAbs were developed on a commercially available synthesis module, which also allows the GMP production of other radiolabeled mAbs.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , Isotope Labeling/methods , Radioisotopes/chemistry , Radiopharmaceuticals/chemistry , Zirconium/chemistry , Automation , Reproducibility of Results
5.
J Biol Chem ; 288(41): 29562-72, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-23979133

ABSTRACT

The chemokine receptor CXCR7, belonging to the membrane-bound G protein-coupled receptor superfamily, is expressed in several tumor types. Inhibition of CXCR7 with either small molecules or small interference (si)RNA has shown promising therapeutic benefits in several tumor models. With the increased interest and effectiveness of biologicals inhibiting membrane-bound receptors we made use of the "Nanobody platform" to target CXCR7. Previously we showed that Nanobodies, i.e. immunoglobulin single variable domains derived from naturally occurring heavy chain-only camelids antibodies, represent new biological tools to efficiently tackle difficult drug targets such as G protein-coupled receptors. In this study we developed and characterized highly selective and potent Nanobodies against CXCR7. Interestingly, the CXCR7-targeting Nanobodies displayed antagonistic properties in contrast with previously reported CXCR7-targeting agents. Several high affinity CXCR7-specific Nanobodies potently inhibited CXCL12-induced ß-arrestin2 recruitment in vitro. A wide variety of tumor biopsies was profiled, showing for the first time high expression of CXCR7 in head and neck cancer. Using a patient-derived CXCR7-expressing head and neck cancer xenograft model in nude mice, tumor growth was inhibited by CXCR7-targeting Nanobody therapy. Mechanistically, CXCR7-targeting Nanobodies did not inhibit cell cycle progression but instead reduced secretion of the angiogenic chemokine CXCL1 from head and neck cancer cells in vitro, thus acting here as inverse agonists, and subsequent angiogenesis in vivo. Hence, with this novel class of CXCR7 inhibitors, we further substantiate the therapeutic relevance of targeting CXCR7 in head and neck cancer.


Subject(s)
Head and Neck Neoplasms/immunology , Receptors, CXCR/immunology , Single-Domain Antibodies/immunology , Xenograft Model Antitumor Assays , Animals , Arrestins/immunology , Arrestins/metabolism , Binding, Competitive/immunology , Camelids, New World/immunology , Cell Line, Tumor , Chemokine CXCL12/pharmacology , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/prevention & control , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Radioligand Assay , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/immunology , Single-Domain Antibodies/pharmacology , Tumor Burden/drug effects , Tumor Burden/immunology , beta-Arrestins
6.
Mol Cancer Ther ; 11(4): 1017-25, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22319202

ABSTRACT

Hepatocyte growth factor (HGF) and its receptor c-Met are associated with increased aggressiveness of tumors and poor prognostic outcome of patients with cancer. Here, we report the development and characterization of therapeutic anti-HGF (αHGF)-Nanobodies and their potential for positron emission tomographic (PET) imaging to assess HGF expression in vivo. Two αHGF-Nanobodies designated 1E2 and 6E10 were identified, characterized, and molecularly fused to an albumin-binding Nanobody unit (Alb8) to obtain serum half-life extension. The resulting Nanobody formats were radiolabeled with the positron emitter zirconium-89 ((89)Zr, t(1/2;) = 78 hours), administered to nude mice bearing U87 MG glioblastoma xenografts, and their biodistribution was assessed. In addition, their therapeutic effect was evaluated in the same animal model at doses of 10, 30, or 100 µg per mouse. The (89)Zr-Nanobodies showed similar biodistribution with selective tumor targeting. For example, 1E2-Alb8 showed decreased blood levels of 12.6%ID/g ± 0.6%ID/g, 7.2%ID/g ± 1.0%ID/g, 3.4%ID/g ± 0.3%ID/g, and 0.3%ID/g ± 0.1%ID/g at 1, 2, 3, and 7 days after injection, whereas tumor uptake levels remained relatively stable at these time points: 7.8%ID/g ± 1.1%ID/g, 8.9%ID/g ± 1.0%ID/g, 8.7%ID/g ± 1.5%ID/g, and 7.2%ID/g ±1.6%ID/g. Uptake in normal tissues was lower than in tumor, except for kidneys. In a therapy study, all Nanobody-treated mice showed tumor growth delay compared with the control saline group. In the 100-µg group, four of six mice were cured after treatment with 1E2-Alb8 and 73 days follow-up, and three of six mice when treated with 6E10-Alb8. These results provide evidence that Nanobodies 1E2-Alb8 and 6E10-Alb8 have potential for therapy and PET imaging of HGF-expressing tumors.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Hepatocyte Growth Factor/metabolism , Radiopharmaceuticals/pharmacology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Drug Delivery Systems , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Nanoparticles/administration & dosage , Positron-Emission Tomography/methods , Radioisotopes/pharmacokinetics , Radioisotopes/pharmacology , Radiopharmaceuticals/pharmacokinetics , Xenograft Model Antitumor Assays , Zirconium/pharmacokinetics , Zirconium/pharmacology
7.
Eur J Nucl Med Mol Imaging ; 39(3): 512-20, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22218876

ABSTRACT

PURPOSE: Positron emission tomography (PET) with (89)Zr-ibritumomab tiuxetan can be used to monitor biodistribution of (90)Y-ibritumomab tiuxetan as shown in mice. The aim of this study was to assess biodistribution and radiation dosimetry of (90)Y-ibritumomab tiuxetan in humans on the basis of (89)Zr-ibritumomab tiuxetan imaging, to evaluate whether co-injection of a therapeutic amount of (90)Y-ibritumomab tiuxetan influences biodistribution of (89)Zr-ibritumomab tiuxetan and whether pre-therapy scout scans with (89)Zr-ibritumomab tiuxetan can be used to predict biodistribution of (90)Y-ibritumomab tiuxetan and the dose-limiting organ during therapy. METHODS: Seven patients with relapsed B-cell non-Hodgkin's lymphoma scheduled for autologous stem cell transplantation underwent PET scans at 1, 72 and 144 h after injection of ~70 MBq (89)Zr-ibritumomab tiuxetan and again 2 weeks later after co-injection of 15 MBq/kg or 30 MBq/kg (90)Y-ibritumomab tiuxetan. Volumes of interest were drawn over liver, kidneys, lungs, spleen and tumours. Ibritumomab tiuxetan organ absorbed doses were calculated using OLINDA. Red marrow dosimetry was based on blood samples. Absorbed doses to tumours were calculated using exponential fits to the measured data. RESULTS: The highest (90)Y absorbed dose was observed in liver (3.2 ± 1.8 mGy/MBq) and spleen (2.9 ± 0.7 mGy/MBq) followed by kidneys and lungs. The red marrow dose was 0.52 ± 0.04 mGy/MBq, and the effective dose was 0.87 ± 0.14 mSv/MBq. Tumour absorbed doses ranged from 8.6 to 28.6 mGy/MBq. Correlation between predicted pre-therapy and therapy organ absorbed doses as based on (89)Zr-ibritumomab tiuxetan images was high (Pearson correlation coefficient r = 0.97). No significant difference between pre-therapy and therapy tumour absorbed doses was found, but correlation was lower (r = 0.75). CONCLUSION: Biodistribution of (89)Zr-ibritumomab tiuxetan is not influenced by simultaneous therapy with (90)Y-ibritumomab tiuxetan, and (89)Zr-ibritumomab tiuxetan scout scans can thus be used to predict biodistribution and dose-limiting organ during therapy. Absorbed doses to spleen were lower than those previously estimated using (111)In-ibritumomab tiuxetan. The dose-limiting organ in patients undergoing stem cell transplantation is the liver.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/radiotherapy , Positron-Emission Tomography , Zirconium/pharmacokinetics , Zirconium/therapeutic use , Adult , Female , Humans , Lymphoma, B-Cell/diagnostic imaging , Lymphoma, B-Cell/surgery , Male , Middle Aged , Radioisotopes/pharmacokinetics , Radioisotopes/therapeutic use , Radiometry , Recurrence , Stem Cell Transplantation , Tissue Distribution
8.
Int J Cancer ; 129(8): 2013-24, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21520037

ABSTRACT

The epidermal growth factor receptor (EGFR) has been shown to be a valid cancer target for antibody-based therapy. At present, several anti-EGFR monoclonal antibodies have been successfully used, such as cetuximab and matuzumab. X-ray crystallography data show that these antibodies bind to different epitopes on the ecto-domain of EGFR, providing a rationale for the combined use of these two antibody specificities. We have previously reported on the successful isolation of antagonistic anti-EGFR nanobodies. In our study, we aimed to improve the efficacy of these molecules by combining nanobodies with specificities similar to both cetuximab and matuzumab into a single biparatopic molecule. Carefully designed phage nanobody selections resulted in two sets of nanobodies that specifically blocked the binding of either matuzumab or cetuximab to EGFR and that did not compete for each others' binding. A combination of nanobodies from both epitope groups into the biparatopic nanobody CONAN-1 was shown to block EGFR activation more efficiently than monovalent or bivalent (monospecific) nanobodies. In addition, this biparatopic nanobody potently inhibited EGF-dependent cell proliferation. Importantly, in an in vivo model of athymic mice bearing A431 xenografts, CONAN-1 inhibited tumour outgrowth with an almost similar potency as the whole mAb cetuximab, despite the fact that CONAN-1 is devoid of an Fc portion that could mediate immune effector functions. Compared to therapy using bivalent, monospecific nanobodies, CONAN-1 was clearly more potent in tumour growth inhibition. These results show that the rational design of biparatopic nanobody-based anticancer therapeutics may yield potent lead molecules for further development.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody Specificity , Carcinoma, Squamous Cell/therapy , Epitopes , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , Single-Chain Antibodies/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antibody Affinity , Cell Line, Tumor , Cetuximab , Humans , Mice , Mice, Nude , Xenograft Model Antitumor Assays
9.
Eur J Nucl Med Mol Imaging ; 38(4): 753-63, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21210114

ABSTRACT

PURPOSE: The ∼15 kDa variable domains of camelid heavy-chain-only antibodies (called Nanobodies®) have the flexibility to be formatted as monovalent, monospecific, multivalent or multispecific single chain proteins with either fast or slow pharmacokinetics. We report the evaluation of the fast kinetic anti-epidermal growth factor receptor (EGFR) Nanobody 7D12, labelled with (68)Ga via the novel bifunctional chelate (BFC) p-isothiocyanatobenzyl-desferrioxamine (Df-Bz-NCS). Df-Bz-NCS has recently been introduced as the chelate of choice for (89)Zr immuno-positron emission tomography (PET). METHODS: Nanobody 7D12 was premodified with Df-Bz-NCS at pH 9. Radiolabelling with purified (68)Ga was performed at pH 5.0-6.5 for 5 min at room temperature. For in vitro stability measurements in storage buffer (0.25 M NaOAc with 5 mg ml(-1) gentisic acid, pH 5.5) at 4°C or in human serum at 37°C, a mixture of (67)Ga and (68)Ga was used. Biodistribution and immuno-PET studies of (68)Ga-Df-Bz-NCS-7D12 were performed in nude mice bearing A431 xenografts using (89)Zr-Df-Bz-NCS-7D12 as the reference conjugate. RESULTS: The Df-Bz-NCS chelate was conjugated to Nanobody 7D12 with a chelate to Nanobody molar substitution ratio of 0.2:1. The overall (68)Ga radiochemical yield was 55-70% (not corrected for decay); specific activity was 100-500 MBq/mg. Radiochemical purity of the conjugate was >96%, while the integrity and immunoreactivity were preserved. (68/67)Ga-Df-Bz-NCS-7D12 was stable in storage buffer as well as in human serum during a 5-h incubation period (<2% radioactivity loss). In biodistribution studies the (68)Ga-labelled Nanobody 7D12 showed high uptake in A431 tumours (ranging from 6.1 ± 1.3 to 7.2 ± 1.5%ID/g at 1-3 h after injection) and high tumour to blood ratios, which increased from 8.2 to 14.4 and 25.7 at 1, 2 and 3 h after injection, respectively. High uptake was also observed in the kidneys. Biodistribution was similar to that of the reference conjugate (89)Zr-Df-Bz-NCS-7D12. Tumours were clearly visualized in a PET imaging study. CONCLUSION: Via a rapid procedure under mild conditions a (68)Ga-Nanobody was obtained that exhibited high tumour uptake and tumour to normal tissue ratios in nude mice bearing A431 xenografts. Fast kinetic (68)Ga-Nanobody conjugates can be promising tools for tumour detection and imaging of target expression.


Subject(s)
Antibodies/chemistry , Deferoxamine/analogs & derivatives , ErbB Receptors/immunology , Isothiocyanates/chemistry , Isotope Labeling/methods , Positron-Emission Tomography/methods , Radioimmunodetection/methods , Animals , Antibodies/blood , Antibodies/immunology , Buffers , Cell Line, Tumor , Chelating Agents/chemistry , Cross-Linking Reagents/chemistry , Deferoxamine/chemistry , Drug Stability , Gallium Radioisotopes , Humans , Mice , Temperature
10.
Cancer Biother Radiopharm ; 25(4): 375-85, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20707716

ABSTRACT

Summation Monoclonal antibodies (mAbs) have been approved for therapeutic use in a broad range of medical indications, especially in oncology, and are forming the most rapidly expanding category of pharmaceuticals. Although engineered mAb fragments and nontraditional antibody-like scaffolds are receiving increasingly more attention, most of the mAb candidates evaluated in past and ongoing clinical trials are full-length mAbs. Immuno-positron emission tomography (PET), the tracking and quantification of mAbs with PET in vivo at superior imaging quality, is an exciting novel option for better understanding the in vivo behavior and efficacy of mAbs in individual patients. This review focuses on immuno-PET with full-length mAbs, and the associated use of the long-lived positron emitters zirconium-89 ((89)Zr) and iodine-124 ((124)I). Very recently, crucial achievements have been obtained to allow broad-scale application of (89)Zr- and (124)I-immuno-PET in clinical mAb development and applications. (89)Zr and (124)I became commercially available worldwide for clinical use. A chelate for facile coupling of (89)Zr to mAbs became commercially available, and generic procedures for labeling of mAbs with (89)Zr and (124)I in a current good manufacturing practice compliant way were established. In this review, critical aspects for the translation of immuno-PET from preclinical investigations to clinical trials will be discussed, as well as the potential clinical applications of immuno-PET. An overview of the results of the first clinical immuno-PET studies will be provided.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/radiotherapy , Positron-Emission Tomography , Radioimmunotherapy , Animals , Clinical Trials as Topic , Humans , Xenograft Model Antitumor Assays
11.
Nat Protoc ; 5(4): 739-43, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20360768

ABSTRACT

The positron emitter zirconium-89 ((89)Zr) has very attractive properties for positron emission tomography (PET) imaging of intact monoclonal antibodies (mAbs) using immuno-PET. This protocol describes the step-by-step procedure for the facile radiolabeling of mAbs or other proteins with (89)Zr using p-isothiocyanatobenzyl-desferrioxamine (Df-Bz-NCS). First, Df-Bz-NCS is coupled to the lysine-NH(2) groups of a mAb at pH 9.0 (pre-modification), followed by purification using gel filtration. Next, the pre-modified mAb is labeled at room temperature by the addition of [(89)Zr]Zr-oxalic acid solution followed by purification using gel filtration. The entire process of pre-modification, radiolabeling and purification steps will take about 2.5 h.


Subject(s)
Antibodies, Monoclonal , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemical synthesis , Zirconium , Animals , Antibodies, Monoclonal/chemistry , Chelating Agents , Deferoxamine/analogs & derivatives , Head and Neck Neoplasms/diagnostic imaging , Humans , Isothiocyanates , Mice , Mice, Nude , Radioisotopes , Radiopharmaceuticals/chemistry , Transplantation, Heterologous
12.
Eur J Nucl Med Mol Imaging ; 37(2): 250-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19763566

ABSTRACT

PURPOSE: Immuno-PET is an emerging imaging tool for the selection of high potential antibodies (mAbs) for imaging and therapy. The positron emitter zirconium-89 ((89)Zr) has attractive characteristics for immuno-PET with intact mAbs. Previously, we have described a multi-step procedure for stable coupling of (89)Zr to mAbs via the bifunctional chelate (BFC) tetrafluorophenol-N-succinyldesferal (TFP-N-sucDf). To enable widespread use of (89)Zr-immuno-PET, we now introduce the novel BFC p-isothiocyanatobenzyl-desferrioxamine B (Df-Bz-NCS) and compare its performance in (89)Zr-immuno-PET with the reference BFC TFP-N-sucDf. METHODS: Three mAbs were premodified with Df-Bz-NCS and labeled with (89)Zr at different pHs to assess the reaction kinetics and robustness of the radiolabeling. Stability of both (89)Zr-Df-Bz-NCS- and (89)Zr-N-sucDf-conjugates was evaluated in different buffers and human serum. Comparative biodistribution and PET studies in tumor-bearing mice were undertaken. RESULTS: The selected conjugation conditions resulted in a chelate:mAb substitution ratio of about 1.5:1. Under optimal radiolabeling conditions (pH between 6.8-7.2), the radiochemical yield was >85% after 60 min incubation at room temperature, resulting in radioimmunoconjugates with preserved integrity and immunoreactivity. The new radioimmunoconjugate was very stable in serum for up to 7 days at 37 degrees C, with <5% (89)Zr release, and was equally stable compared to the reference conjugate when stored in the appropriate buffer at 4 degrees C. In biodistribution and imaging experiments, the novel and the reference radioimmunoconjugates showed high and similar accumulation in tumors in nude mice. CONCLUSIONS: The novel Df-Bz-NCS BFC allows efficient and easy preparation of optimally performing (89)Zr-labeled mAbs, facilitating further exploration of (89)Zr-immuno-PET as an imaging tool.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Carcinoma, Squamous Cell/metabolism , Chelating Agents/chemistry , Deferoxamine/analogs & derivatives , Isothiocyanates/chemistry , Positron-Emission Tomography/methods , Radioisotopes/pharmacokinetics , Zirconium/pharmacokinetics , Animals , Carcinoma, Squamous Cell/diagnostic imaging , Cell Line, Tumor , Deferoxamine/chemistry , Female , Humans , Isotope Labeling/methods , Metabolic Clearance Rate , Mice , Mice, Nude , Organ Specificity , Radioimmunodetection/methods , Radioisotopes/chemistry , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Zirconium/chemistry
13.
Eur J Nucl Med Mol Imaging ; 35(10): 1857-67, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18491091

ABSTRACT

PURPOSE: Targeting the c-Met receptor with monoclonal antibodies (MAbs) is an appealing approach for cancer diagnosis and treatment because this receptor plays a prominent role in tumour invasion and metastasis. Positron emission tomography (PET) might be a powerful tool for guidance of therapy with anti-Met MAbs like the recently described MAb DN30 because it allows accurate quantitative imaging of tumour targeting (immuno-PET). We considered the potential of PET with either (89)Zr-labelled (residualising radionuclide) or (124)I-labelled (non-residualising radionuclide) DN30 for imaging of Met-expressing tumours. MATERIALS AND METHODS: The biodistribution of co-injected (89)Zr-DN30 and iodine-labelled DN30 was compared in nude mice bearing either the human gastric cancer line GLT-16 (high Met expression) or the head-and-neck cancer line FaDu (low Met expression). PET images were acquired in both xenograft models up to 4 days post-injection (p.i.) and used for quantification of tumour uptake. RESULTS: Biodistribution studies in GTL-16-tumour-bearing mice revealed that (89)Zr-DN30 achieved much higher tumour uptake levels than iodine-labelled DN30 (e.g. 19.6%ID/g vs 5.3%ID/g, 5 days p.i.), while blood levels were similar, indicating internalisation of DN30. Therefore, (89)Zr-DN30 was selected for PET imaging of GLT-16-bearing mice. Tumours as small as 11 mg were readily visualised with immuno-PET. A distinctive lower (89)Zr uptake was observed in FaDu compared to GTL-16 xenografts (e.g. 7.8%ID/g vs 18.1%ID/g, 3 days p.i.). Nevertheless, FaDu xenografts were also clearly visualised with (89)Zr-DN30 immuno-PET. An excellent correlation was found between PET-image-derived (89)Zr tumour uptake and ex-vivo-assessed (89)Zr tumour uptake (R(2)=0.98). CONCLUSIONS: The long-lived positron emitter (89)Zr seems attractive for PET-guided development of therapeutic anti-c-Met MAbs.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Radioisotopes/pharmacokinetics , Receptors, Growth Factor/metabolism , Zirconium/pharmacokinetics , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Humans , Metabolic Clearance Rate , Mice , Mice, Nude , Organ Specificity , Positron-Emission Tomography/methods , Proto-Oncogene Proteins c-met , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
14.
Eur J Nucl Med Mol Imaging ; 33(11): 1337-45, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16832633

ABSTRACT

PURPOSE: To evaluate whether (89)Zr can be used as a PET surrogate label for quantification of (90)Y-ibritumomab tiuxetan ((90)Y-Zevalin) biodistribution and dosimetry before myeloablative radioimmunotherapy. METHODS: Zevalin was labelled with (89)Zr by introducing N-succinyldesferal (N-sucDf) as a second chelate. For comparison of the in vitro stability of (89)Zr-Zevalin and (88)Y-Zevalin (as a substitute for (90)Y), samples were incubated in human serum at 37 degrees C up to 6 days. Biodistribution of (89)Zr-Zevalin and (88)Y-Zevalin was assessed at 24, 48, 72 and 144 h p.i. by co-injection in nude mice bearing the non-Hodgkin's lymphoma (NHL) xenograft line Ramos. The clinical performance of (89)Zr-Zevalin-PET was evaluated via a pilot imaging study in a patient with NHL, who had undergone [(18)F]FDG-PET 2 weeks previously. RESULTS: Modification of Zevalin with N-sucDf resulted in an N-sucDf-to-antibody molar ratio of 0.83+/-0.04. After radiolabelling and purification, the radiochemical purity and immunoreactivity of (89)Zr-Zevalin always exceeded 95% and 80%, respectively. (89)Zr-Zevalin showed the same stability in serum as (88)Y-Zevalin, with a radiochemical purity >95% during a period of 6 days. The co-injected (89)Zr-Zevalin and (88)Y-Zevalin conjugates showed a very similar biodistribution, except for liver and bone accumulation at 72 and 144 h p.i., which was significantly higher for (89)Zr than for (88)Y. PET images obtained after injection of (89)Zr-Zevalin showed clear targeting of all known tumour lesions. CONCLUSION: (89)Zr-Zevalin and (88)Y-Zevalin showed a very similar biodistribution in mice, implying that (89)Zr-Zevalin-PET might be well suited for prediction of (90)Y-Zevalin biodistribution in a myeloablative setting.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Positron-Emission Tomography/methods , Whole-Body Counting/methods , Yttrium Radioisotopes/pharmacokinetics , Zirconium/pharmacokinetics , Animals , Antibodies, Monoclonal/therapeutic use , Female , Metabolic Clearance Rate , Mice , Mice, Nude , Organ Specificity , Radioisotopes/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Radiotherapy Planning, Computer-Assisted/methods , Tissue Distribution , Yttrium Radioisotopes/therapeutic use
15.
Clin Cancer Res ; 12(7 Pt 1): 2133-40, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16609026

ABSTRACT

PURPOSE: Immuno-positron emission tomography (PET), the combination of PET with monoclonal antibodies (mAb), is an attractive option to improve tumor detection and to guide mAb-based therapy. The long-lived positron emitter zirconium-89 ((89)Zr) has ideal physical characteristics for immuno-PET with intact mAbs but has never been used in a clinical setting. In the present feasibility study, we aimed to evaluate the diagnostic imaging performance of immuno-PET with (89)Zr-labeled-chimeric mAb (cmAb) U36 in patients with squamous cell carcinoma of the head and neck (HNSCC), who were at high risk of having neck lymph node metastases. EXPERIMENTAL DESIGN: Twenty HNSCC patients, scheduled to undergo neck dissection with or without resection of the primary tumor, received 75 MBq (89)Zr coupled to the anti-CD44v6 cmAb U36 (10 mg). All patients were examined by computed tomography (CT) and/or magnetic resonance imaging (MRI) and immuno-PET before surgery. Six patients also underwent PET with (18)F-fluoro-2-deoxy-d-glucose. Immuno-PET scans were acquired up to 144 hours after injection. Diagnostic findings were recorded per neck side (left or right) as well as per lymph node level (six levels per side), and compared with histopathologic outcome. For this purpose, the CT/MRI scores were combined and the best of both scores was used for analysis. RESULTS: Immuno-PET detected all primary tumors (n = 17) as well as lymph node metastases in 18 of 25 positive levels (sensitivity 72%) and in 11 of 15 positive sides (sensitivity 73%). Interpretation of immuno-PET was correct in 112 of 121 operated levels (accuracy 93%) and in 19 of 25 operated sides (accuracy 76%). For CT/MRI, sensitivities of 60% and 73% and accuracies of 90% and 80% were found per level and side, respectively. In the six patients with seven tumor-involved neck levels and sides, immuno-PET and (18)F-fluoro-2-deoxy-d-glucose PET gave comparable diagnostic results. CONCLUSION: In this study, immuno-PET with (89)Zr-cmAb U36 performed at least as good as CT/MRI for detection of HNSCC lymph node metastases.


Subject(s)
Antibodies, Monoclonal , Carcinoma, Squamous Cell/diagnosis , Head and Neck Neoplasms/diagnosis , Positron-Emission Tomography/methods , Radioisotopes , Zirconium , Aged , Antibodies, Monoclonal/administration & dosage , Carcinoma, Squamous Cell/secondary , Carcinoma, Squamous Cell/surgery , False Positive Reactions , Female , Fluorodeoxyglucose F18 , Head and Neck Neoplasms/secondary , Head and Neck Neoplasms/surgery , Humans , Isotope Labeling , Lymphatic Metastasis , Male , Middle Aged , Neck Dissection , Sensitivity and Specificity
16.
J Nucl Med ; 46(11): 1898-906, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16269605

ABSTRACT

UNLABELLED: Immuno-PET as a scouting procedure before radioimmunotherapy (RIT) aims at confirming tumor targeting and accurately estimating radiation dose delivery to both tumor and normal tissues and might therefore be of value for selection of patient candidates for RIT. A prerequisite for this approach is that PET radioimmunoconjugates and RIT radioimmunoconjugates must show a similar biodistribution. In the present study, we evaluated the potential of the long-lived positron emitter (89)Zr to predict biodistribution of the residualizing therapeutic radiometals (88)Y (as a substitute for (90)Y) and (177)Lu when labeled to the monoclonal antibody (mAb) cetuximab via different types of chelates. Cetuximab was selected as a model mAb because it abundantly internalizes after binding to the epidermal growth factor receptor. METHODS: Cetuximab was labeled with (89)Zr using succinylated desferrioxamine B (N-sucDf). The chelates p-benzyl isothiocyanate-1,4,7,10-tetraazacyclododecane-1,4,7, 10-tetraacetic acid (p-SCN-Bz-DOTA) and p-isothiocyanatobenzyl diethylenetriaminepentaacetic acid (p-SCN-Bz-DTPA) were both used for radiolabeling with (88)Y and (177)Lu. For measurement of the in vitro stability of each of the 5 radioimmunoconjugates, samples were incubated in freshly prepared human serum at 37 degrees C up to 16 d. Biodistribution was assessed at 24, 48, 72, and 144 h after intraperitoneal coinjection of the PET and RIT conjugates in nude mice bearing the squamous cell carcinoma xenograft line A431. RESULTS: Cetuximab premodification with N-sucDf, p-SCN-Bz-DOTA, or p-SCN-Bz-DTPA resulted in chelate-to-mAb molar ratios of about 1. After radiolabeling and purification, the radiochemical purity and immunoreactive fraction of the conjugates always exceeded 97% and 93%, respectively. All conjugates were stable in serum, showing a radioactivity release of less than 5% until day 7. From day 7 until day 16, an enhanced release was observed for the (89)Zr-N-sucDf, (88)Y-p-SCN-Bz-DTPA, and (177)Lu-p-SCN-Bz-DTPA conjugates. The coinjected PET and RIT conjugates showed similar biodistributions, except for the thighbone and sternum. For example, the (89)Zr-N-sucDf conjugate showed a 2.0-2.5 times higher radioactivity accretion in the thighbone than did the RIT conjugates at 72 h after injection. CONCLUSION: In view of the advantages of PET over SPECT, (89)Zr-immuno-PET is a promising modality for in vivo scouting of (90)Y- and (177)Lu-labeled mAbs, although care should be taken when estimating bone marrow doses.


Subject(s)
Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/metabolism , Lutetium/pharmacokinetics , Positron-Emission Tomography/methods , Yttrium Radioisotopes/pharmacokinetics , Zirconium/pharmacokinetics , Animals , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Drug Delivery Systems/methods , Female , Humans , Lutetium/therapeutic use , Metabolic Clearance Rate , Mice , Mice, Nude , Organ Specificity , Radioisotopes/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution , Yttrium Radioisotopes/therapeutic use
17.
Eur J Nucl Med Mol Imaging ; 31(12): 1645-52, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15290121

ABSTRACT

PURPOSE: Monoclonal antibodies (MAbs) labelled with 124I are an attractive option for quantitative imaging with positron emission tomography (PET) in a scouting procedure prior to 131I-radioimmunotherapy (131I-RIT). In this study, three important items in the labelling of MAbs with 124I were introduced to obtain optimal and reproducible product quality: restoration of radiation-induced inorganic deterioration of the starting 124I solution, radiation protection during and after 124I labelling, and synchronisation of the I/MAb molar ratio. METHODS: A new method was applied, using an NaIO3/NaI carrier mix, realising in one step >90% restoration of deteriorated 124I into the iodide form and chemical control over the I/MAb molar ratio. Chimeric MAb (cMAb) U36 and the murine MAbs 425 and E48 were labelled with 124I using the so-called Iodogen-coated MAb method, as this method provides optimal quality conjugates under challenging radiation conditions. As a standardising condition, NaIO3/NaI carrier mix was added at a stoichiometric I/MAb molar ratio of 0.9. For comparison, MAbs were labelled with 131I and with a mixture of 124I, 123I, 126I and 130I. RESULTS: Labelling with 124I in this setting resulted in overall yields of >70%, a radiochemical purity of >95%, and preservation of MAb integrity and immunoreactivity, including at the patient dose level (85 MBq). No significant quality differences were observed when compared with 131I products, while the iodine isotope mixture gave exactly the same labelling efficiency for each of the isotopes, excluding a different chemical reactivity of 124I-iodide. The scouting performance of 124I-cMAb U36 labelled at the patient dose level was evaluated in biodistribution studies upon co-injection with 131I-labelled cMAb U36, and by PET imaging in nude mice bearing the head and neck cancer xenograft line HNX-OE. 124I-cMAb and 131I-cMAb U36 labelled with a synchronised I/MAb molar ratio gave fully concordant tissue uptake values. Selective tumour uptake was confirmed with immuno-PET, revealing visualisation of 15 out of 15 tumours. CONCLUSION: These results pave the way for renewed evaluation of the potential of 124I-immuno-PET for clinical applications.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/metabolism , Iodine Radioisotopes/pharmacokinetics , Positron-Emission Tomography/methods , Animals , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Female , Humans , Image Enhancement/methods , Iodine Radioisotopes/therapeutic use , Metabolic Clearance Rate , Mice , Mice, Nude , Organ Specificity , Prognosis , Radioimmunotherapy/methods , Radiopharmaceuticals/pharmacokinetics , Radiotherapy Planning, Computer-Assisted/methods , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
18.
Cancer Biother Radiopharm ; 18(4): 655-61, 2003 Aug.
Article in English | MEDLINE | ID: mdl-14503961

ABSTRACT

Antibody-PET imaging might be of value for the selection of radioimmunotherapy (RIT) candidates to confirm tumor targeting and to estimate radiation doses to tumor and normal tissues. One of the requirements to be set for such a scouting procedure is that the biodistributions of the diagnostic and therapeutic radioimmunoconjugates should be similar. In the present study we evaluated the potential of the positron emitters zirconium-89 ((89)Zr) and iodine-124 ((124)I) for this approach, as these radionuclides have a relatively long half-life that matches with the kinetics of MAbs in vivo (t(1/2) 3.27 and 4.18 days, respectively). After radiolabeling of the head and neck squamous cell carcinoma (HNSCC)-selective chimeric antibody (cMAb) U36, the biodistribution of two diagnostic (cMAb U36-N-sucDf-(89)Zr and cMAb U36-(124)I) and three therapeutic radioimmunoconjugates (cMAb U36-p-SCN-Bz-DOTA-(88)Y-with (88)Y being substitute for (90)Y, cMAb U36-(131)I, and cMAb U36-MAG3-(186)Re) was assessed in mice with HNSCC-xenografts, at 24, 48, and 72 hours after injection. Two patterns of biodistribution were observed, one pattern matching for (89)Zr- and (88)Y-labeled cMAb U36 and one pattern matching for (124)I-, (131)I-, and (186)Re-cMAb U36. The most remarkable differences between both patterns were observed for uptake in tumor and liver. Tumor uptake levels were 23.2 +/- 0.5 and 24.1 +/- 0.7%ID/g for the (89)Zr- and (88)Y-cMAb U36 and 16.0 +/- 0.8, 15.7 +/- 0.79 and 17.1 +/- 1.6%ID/g for (124)I-, (131)I-, and (186)Re-cMAb U36-conjugates, respectively, at 72 hours after injection. For liver these values were 6.9 +/- 0.8 ((89)Zr), 6.2 +/- 0.8 ((88)Y), 1.7 +/- 0.1 ((124)I), 1.6 +/- 0.1 ((131)I), and 2.3 +/- 0.1 ((186)Re), respectively. These preliminary data justify the further development of antibody-PET with (89)Zr-labeled MAbs for scouting of therapeutic doses of (90)Y-labeled MAbs. In such approach (124)I-labeled MAbs are most suitable for scouting of (131)I- and (186)Re-labeled MAbs.


Subject(s)
Immunoconjugates/pharmacokinetics , Iodine Radioisotopes , Radioisotopes , Zirconium , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Female , Glycoproteins/immunology , Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Humans , Hyaluronan Receptors/immunology , Immunoconjugates/chemistry , Isotope Labeling , Mice , Mice, Nude , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics , Rhenium , Time Factors , Tissue Distribution , Tomography, Emission-Computed , Xenograft Model Antitumor Assays , Yttrium Radioisotopes
19.
Genes Chromosomes Cancer ; 33(2): 168-77, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11793443

ABSTRACT

Ample molecular data are available on the progression from normal mucosa to invasive head and neck squamous cell carcinoma (HNSCC), but information on further genetic progression to metastatic disease is scarce. To obtain insight into the metastatic process, we compared 23 primary HNSCCs with 25 corresponding lymph node metastases (LNMs) and 10 corresponding distant metastases (DMs) with respect to TP53 mutations and patterns of loss of heterozygosity (LOH) based on 26 microsatellite markers on six chromosome arms (3p, 9p, 17p, 13q, 8p, and 18q). In 18 of the 23 patients, a TP53 mutation was detected in the primary tumor, and in all cases the same TP53 mutation was present in the corresponding LNM or DM. In nine of 20 patients with LNMs and three of seven patients with DMs, the LOH pattern of metastasis differed from that of the corresponding primary tumor by at least one marker. Microsatellite markers located on chromosome arms 13q, 8p, and 18q were most frequently discordant, providing evidence that alterations at these chromosomes occur late in HNSCC carcinogenesis. Moreover, evidence was found that DMs had developed directly from the primary tumor and not from LNMs. Remarkably, we observed that the mutational status of the TP53 gene is associated significantly with the degree of genetic differences between primary HNSCCs and corresponding metastases. All patients with TP53 wild-type primary tumors showed significantly more discordant LOH patterns in the corresponding LNMs and DMs than patients with TP53-mutated tumors. The percentages were 100% versus 27% (LNMs) and 100% versus 0% (DMs), respectively (P = 0.008 and P = 0.029; two-sided Fisher exact test). This finding suggests that TP53-mutated tumors need fewer additional genetic alterations to develop metastases compared with TP53 wild-type primary tumors.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/secondary , Chromosome Aberrations , Genes, p53/genetics , Head and Neck Neoplasms/genetics , Aged , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Carcinoma, Squamous Cell/pathology , DNA Mutational Analysis , Female , Head and Neck Neoplasms/pathology , Heart Neoplasms/genetics , Heart Neoplasms/pathology , Heart Neoplasms/secondary , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/secondary , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Loss of Heterozygosity/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lymph Nodes/pathology , Male , Microsatellite Repeats/genetics , Middle Aged , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/secondary , Splenic Neoplasms/genetics , Splenic Neoplasms/pathology , Splenic Neoplasms/secondary
SELECTION OF CITATIONS
SEARCH DETAIL
...