Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Affiliation country
Publication year range
1.
Eur J Nucl Med Mol Imaging ; 46(12): 2569-2580, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31388723

ABSTRACT

PURPOSE: Targeting fibroblast activation protein (FAP) is a new diagnostic approach allowing the visualization of tumor stroma. Here, we applied FAP-specific PET imaging to gliomas. We analyzed the target affinity and specificity of two FAP ligands (FAPI-02 and FAPI-04) in vitro, and the pharmacokinetics and biodistribution in mice in vivo. Clinically, we used 68Ga-labeled FAPI-02/04 for PET imaging in 18 glioma patients (five IDH-mutant gliomas, 13 IDH-wildtype glioblastomas). METHODS: For binding studies with 177Lu-radiolabeled FAPI-02/04, we used the glioblastoma cell line U87MG, FAP-transfected fibrosarcoma cells, and CD26-transfected human embryonic kidney cells. For pharmacokinetic and biodistribution studies, U87MG-xenografted mice were injected with 68Ga-labeled compounds followed by small-animal PET imaging and 177Lu-labeled FAPI-02/04, respectively. Clinical PET/CT scans were performed 30 min post intravenous administration of 68Ga-FAPI-02/04. PET and MRI scans were co-registrated. Immunohistochemistry was done on 14 gliomas using a FAP-specific antibody. RESULTS: FAPI-02 and FAPI-04 showed high binding specificity to FAP. FAPI-04 demonstrated higher tumor accumulation and delayed elimination compared with FAPI-02 in preclinical studies. IDH-wildtype glioblastomas and grade III/IV, but not grade II, IDH-mutant gliomas showed elevated tracer uptake. In glioblastomas, we observed spots with increased uptake in projection on contrast-enhancing areas. Immunohistochemistry showed FAP-positive cells with mainly elongated cell bodies and perivascular FAP-positive cells in glioblastomas and an anaplastic IDH-mutant astrocytoma. CONCLUSIONS: Using FAP-specific PET imaging, increased tracer uptake in IDH-wildtype glioblastomas and high-grade IDH-mutant astrocytomas, but not in diffuse astrocytomas, may allow non-invasive distinction between low-grade IDH-mutant and high-grade gliomas. Therefore, FAP-specific imaging in gliomas may be useful for follow-up studies although further clinical evaluation is required.


Subject(s)
Gelatinases/metabolism , Glioblastoma/genetics , Glioblastoma/pathology , Isocitrate Dehydrogenase/genetics , Membrane Proteins/metabolism , Positron Emission Tomography Computed Tomography , Serine Endopeptidases/metabolism , Acebutolol , Adult , Animals , Biological Transport , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Endopeptidases , Female , Glioblastoma/diagnostic imaging , Glioblastoma/metabolism , Humans , Ligands , Mice , Middle Aged , Mutation , Naphthols , Neoplasm Grading , Radioactive Tracers , Triazines , Young Adult
2.
Mol Imaging ; 12(8)2013.
Article in English | MEDLINE | ID: mdl-24447616

ABSTRACT

The extracellular domain of human carbonic anhydrase IX (CA IX) is extended by a proteoglycan-like region (PGLR). The aim of the present study was the development of novel molecules with specificity for PGLR, which may be used for tumor targeting and imaging. PGLR was chemically synthesized, and phage display biopanning was performed. The identified ligand PGLR-P1 was labeled with 125I and characterized for target binding and metabolic stability. In vitro characterization included kinetic, competition, and internalization studies on CA IX-positive renal cell carcinoma SKRC 52 cells. The CA IX-negative cell lines HEK293 wt and BxPC3 were used as negative controls. In vitro binding experiments revealed an increasing affinity of 125I-PGLR-P1 to SKRC 52 cells but not to negative control HEK293 wt and BxPC3 cells. Internalization studies indicated an exclusive cell membrane binding. Biodistribution analysis demonstrated a higher accumulation in SKRC 52 tumors than in most normal tissues after perfusion. In vivo blocking led to a significant decrease in tumor uptake. Our findings indicate that PGLR-P1 is a promising lead structure for the development of new peptide-based ligands targeting the PGLR of CA IX and reveal challenges that need to be considered for peptide-related molecular imaging.


Subject(s)
Antigens, Neoplasm/chemistry , Antigens, Neoplasm/metabolism , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/metabolism , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Proteoglycans/metabolism , Animals , Binding Sites , Carbonic Anhydrase IX , Cell Line, Tumor , Cell Membrane/metabolism , Chromatography, High Pressure Liquid , Flow Cytometry , HEK293 Cells , Humans , Iodine Radioisotopes , Mass Spectrometry , Mice , Mice, Inbred BALB C , Peptide Library , Proteoglycans/chemistry
3.
Org Biomol Chem ; 11(16): 2706-11, 2013 Apr 28.
Article in English | MEDLINE | ID: mdl-23474823

ABSTRACT

Peptides are excellent alternatives to small molecules and proteinaceous drugs. Their high medicinal potential for diagnostic and therapeutic applications has prompted the development of tumor targeting peptides. Despite its excellent tumor binding capacity, FROP-DOTA (H-Glu-Asn-Tyr-Glu-Leu-Met-Asp-Leu-Leu-Ala-Tyr-Leu-Lys(DOTA)-NH2), a peptide that we had identified in phage display libraries, revealed slow binding kinetics. Consequently, biodistribution studies showed that its excretion forestalled a significant tumor accumulation. The aim of this study was to investigate whether the conjugation of PEG to FROP-DOTA resulted in a derivative with a prolonged residence time in the blood. A synthetic method for the PEGylation of the tumor specific peptide FROP-DOTA was developed. Thereafter, binding studies were done in vitro and a biodistribution was performed in tumor bearing animals. These were compared to the data obtained with FROP-DOTA. The binding kinetics of the PEGylated FROP-DOTA was even slower than that of FROP-DOTA. Biodistribution studies of the labeled conjugate in mice bearing human FRO82-2 tumors showed a time dependent increased uptake of the PEGylated peptide with a high retention (at 24 h p.i. 76% of the maximal activity concentration persisted in the tumor). The highest uptake values were determined at 120 min p.i. reaching 2.3%ID/g tumor as compared to 0.06%ID/g observed for the non-PEGylated derivative at 135 min p.i. Apparently, PEGylation provides a substantially improved stabilization in the circulation which allowed a stable tumor accumulation.


Subject(s)
Drug Delivery Systems , Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Neoplasms/drug therapy , Peptides/chemistry , Peptides/pharmacokinetics , Polyethylene Glycols/chemistry , Amino Acid Sequence , Animals , Female , Heterocyclic Compounds, 1-Ring/blood , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neoplasms/metabolism , Peptide Library , Peptides/blood , Tissue Distribution
4.
Methods ; 55(3): 215-22, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21640826

ABSTRACT

Peptides, proteins and antibodies are promising candidates as carriers for radionuclides in endoradiotherapy. This novel class of pharmaceuticals offers a great potential for the targeted therapy of cancer. The fact that some receptors are overexpressed in several tumor types and can be targeted by small peptides, proteins or antibodies conjugated to radionuclides has been used in the past for the development of peptide endoradiotherapeutic agents such as (90)Y-DOTATOC or radioimmunotherapy of lymphomas with Zevalin. These procedures have been shown to be powerful options for the treatment of cancer patients. Design of new peptide libraries and scaffolds combined with biopanning techniques like phage and ribosome display may lead to the discovery of new specific ligands for target structures overexpressed in malignant tumors. Display methods are high throughput systems which select for high affinity binders. These methods allow the screening of a vast amount of potential binding motifs which may be exposed to either cells overexpressing the target structures or in a cell-free system to the protein itself. Labelling these binders with radionuclides creates new potential tracers for application in diagnosis and endoradiotherapy. This review highlights the advantages and problems of phage and ribosome display for the identification and evaluation of new tumor specific peptides.


Subject(s)
Biotechnology/methods , Drug Discovery/methods , Neoplasms/drug therapy , Peptides/administration & dosage , Animals , Biotechnology/trends , Drug Carriers/administration & dosage , Drug Discovery/trends , Humans , Neoplasms/diagnosis , Neoplasms/metabolism , Peptides/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism
5.
Angew Chem Int Ed Engl ; 51(52): 13136-9, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23150222

ABSTRACT

Frankenstein's peptide: the grafting of the binding domain from miniprotein Min-23 into the sunflower trypsin inhibitor (SFTI-I) peptide scaffold preserved its in vitro and in vivo binding specificity and proteolytic stability. The combination of these peptides was shown to be tumor-specific with a good binding affinity for delta-like ligand 4 (Dll4) protein. The use of SFTI-I as a peptide scaffold is ideal for hit-to-lead development.


Subject(s)
Peptides/metabolism , Trypsin Inhibitors/metabolism , Amino Acid Sequence , Cell Line, Tumor , Fluorescent Dyes/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Microscopy, Confocal , Molecular Sequence Data , Neoplasms/pathology , Peptide Library , Peptides/chemistry , Protein Binding , Surface Plasmon Resonance , Trypsin Inhibitors/chemistry
6.
Molecules ; 16(2): 1559-78, 2011 Feb 14.
Article in English | MEDLINE | ID: mdl-21321528

ABSTRACT

The transfer of peptides identified through the phage display technology to clinical applications is difficult. Major drawbacks are the metabolic degradation and label instability. The aim of our work is the optimization of DUP-1, a peptide which was identified by phage display to specifically target human prostate carcinoma. To investigate the influence of chelate conjugation, DOTA was coupled to DUP-1 and labeling was performed with ¹¹¹In. To improve serum stability cyclization of DUP-1 and targeted D-amino acid substitution were carried out. Alanine scanning was performed for identification of the binding site and based on the results peptide fragments were chemically synthesized. The properties of modified ligands were investigated in in vitro binding and competition assays. In vivo biodistribution studies were carried out in mice, carrying human prostate tumors subcutaneously. DOTA conjugation resulted in different cellular binding kinetics, rapid in vivo renal clearance and increased tumor-to-organ ratios. Cyclization and D-amino acid substitution increased the metabolic stability but led to binding affinity decrease. Fragment investigation indicated that the sequence NRAQDY might be significant for target-binding. Our results demonstrate challenges in optimizing peptides, identified through phage display libraries, and show that careful investigation of modified derivatives is necessary in order to improve their characteristics.


Subject(s)
Indium Radioisotopes/chemistry , Peptides/chemistry , Prostatic Neoplasms/chemistry , Amino Acid Sequence , Animals , Cell Line, Tumor , Chelating Agents/chemistry , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Peptide Library , Peptides/genetics , Tissue Distribution
7.
J Nucl Med ; 62(2): 160-167, 2021 02.
Article in English | MEDLINE | ID: mdl-33127618

ABSTRACT

Fibroblast activation protein (FAP), a membrane-anchored peptidase, is highly expressed in cancer-associated fibroblasts in more than 90% of epithelial tumors and contributes to progression and worse prognosis of different cancers. Therefore, FAP is considered a promising target for radionuclide-based approaches for diagnosis and treatment of tumors and for the diagnosis of nonmalignant diseases associated with a remodeling of the extracellular matrix. Accordingly, a variety of quinolone-based FAP inhibitors (FAPIs) coupled to chelators were developed displaying specific binding to human and murine FAP with a rapid and almost complete internalization. Because of a high tumor uptake and a very low accumulation in normal tissues, as well as a rapid clearance from the circulation, a high contrast is obtained for FAPI PET/CT imaging even at 10 min after tracer administration. Moreover, FAPI PET/CT provides advantages over 18F-FDG PET/CT in several tumor entities for initial staging and detection of tumor recurrence and metastases, including peritonitis carcinomatosa.


Subject(s)
Gelatinases/metabolism , Membrane Proteins/metabolism , Molecular Imaging/methods , Serine Endopeptidases/metabolism , Endopeptidases , Humans , Positron Emission Tomography Computed Tomography , Tomography, Emission-Computed, Single-Photon
8.
Curr Opin Chem Biol ; 63: 145-151, 2021 08.
Article in English | MEDLINE | ID: mdl-34004409

ABSTRACT

Targeted therapy of cancer is considered as promising alternative approach to conventional chemotherapy and radiotherapy. Recent advancements in biotechnology have significantly improved the identification of novel radiopharmaceuticals allowing for more accurate imaging and therapeutic targeting of epithelial tumors. The successful development of radiotracers critically depends on the selection and validation of the tumor-specific target structure, the technical approach employed for the identification of a target-specific ligand, and the evaluation and improvement of the binding properties and the pharmacokinetic profile of the ligand by biotechnological procedures or chemical modification, respectively. Employing rational design of a quinoline-based fibroblast activation protein inhibitor (FAPI) and 'high-through put' display technology using a sunflower trypsin inhibitor1-based peptide library, several FAPI derivatives and a novel αvß6 integrin-binding peptide (SFITGv6) were identified. FAPI and SFITGv6 represent powerful radiopharmaceuticals for diagnostic imaging and/or endoradiotherapy of FAP- and αvß6 integrin-expressing epithelial tumors, respectively.


Subject(s)
Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Neoplasms/diagnosis , Neoplasms/radiotherapy , Precision Medicine/methods , Radiopharmaceuticals/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Diagnostic Imaging/methods , Endopeptidases , Enzyme Inhibitors/pharmacokinetics , Helianthus/chemistry , Humans , Ligands , Membrane Proteins/antagonists & inhibitors , Molecular Targeted Therapy , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , Peptide Library , Peptides/chemistry , Peptides/pharmacokinetics , Positron-Emission Tomography/methods , Quinolines/chemistry , Quinolines/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Tomography, Emission-Computed, Single-Photon/methods , Trypsin Inhibitors/chemistry , Trypsin Inhibitors/pharmacokinetics
9.
EJNMMI Radiopharm Chem ; 6(1): 26, 2021 Aug 21.
Article in English | MEDLINE | ID: mdl-34417894

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts are found in the stroma of epithelial tumors. They are characterized by overexpression of the fibroblast activation protein (FAP), a serine protease which was already proven as attractive target for chelator-based theranostics. Unfortunately, the value of gallium-68 labeled tracers is limited by their batch size and the short nuclide half-life. To overcome this drawback, radiolabeling with aluminum fluoride complexes and 6-fluoronicotinamide derivatives of the longer-lived nuclide fluorine-18 was established. The novel compounds were tested for their FAP-specific binding affinity. Uptake and binding competition were studied in vitro using FAP expressing HT-1080 cells. HEK cells transfected with the closely related dipeptidyl peptidase-4 (HEK-CD26) were used as negative control. Small animal positron emission tomography imaging and biodistribution experiments were performed in HT-1080-FAP xenografted nude mice. [18F]AlF-FAPI-74 was selected for PET/CT imaging in a non-small cell lung cancer (NSCLC) patient. RESULTS: In vitro, 18F-labeled FAPI-derivatives demonstrated high affinity (EC50 = < 1 nm to 4.2 nm) and binding of up to 80% to the FAP-expressing HT1080 cells while no binding to HEK-CD26 cells was observed. While small animal PET imaging revealed unfavorable biliary excretion of most of the 18F-labeled compounds, the NOTA bearing compounds [18F]AlF-FAPI-74 and -75 achieved good tumor-to-background ratios, as a result of their preferred renal excretion. These two compounds showed the highest tumor accumulation in PET imaging. The organ distribution values of [18F]AlF-FAPI-74 were in accordance with the small animal PET imaging results. Due to its less complex synthesis, fast clearance and low background values, [18F]AlF-FAPI-74 was chosen for clinical imaging. PET/CT of a patient with metastasized non-small cell lung cancer (NSCLC), enabled visualization of the primary tumor and its metastases at the hepatic portal and in several bones. This was accompanied by a rapid clearance from the blood pool and low background in healthy organs. CONCLUSION: [18F]AlF-labeled FAPI derivatives represent powerful tracers for PET. Owing to an excellent performance in PET imaging, FAPI-74 can be regarded as a promising precursor for [18F]AlF-based FAP-imaging.

10.
J Nucl Med ; 62(2): 201-207, 2021 02.
Article in English | MEDLINE | ID: mdl-32591493

ABSTRACT

68Ga-fibroblast activation protein inhibitors (FAPIs) 2, 4, and 46 have already been proposed as promising PET tracers. However, the short half-life of 68Ga (68 min) creates problems with manufacture and delivery. 18F (half-life, 110 min) labeling would result in a more practical large-scale production, and a cold-kit formulation would improve the spontaneous availability. The NOTA chelator ligand FAPI-74 can be labeled with both 18F-AlF and 68Ga. Here, we describe the in vivo evaluation of 18F-FAPI-74 and a proof of mechanism for 68Ga-FAPI-74 labeled at ambient temperature. Methods: In 10 patients with lung cancer, PET scans were acquired at 10 min, 1 h, and 3 h after administration of 259 ± 26 MBq of 18F-FAPI-74. Physiologic biodistribution and tumor uptake were semiquantitatively evaluated on the basis of SUV at each time point. Absorbed doses were evaluated using OLINDA/EXM, version 1.1, and QDOSE dosimetry software with the dose calculator IDAC-Dose, version 2.1. Identical methods were used to evaluate one examination after injection of 263 MBq of 68Ga-FAPI-74. Results: The highest contrast was achieved in primary tumors, lymph nodes, and distant metastases at 1 h after injection, with an SUVmax of more than 10. The effective dose per a 100-MBq administered activity of 18F-FAPI-74 was 1.4 ± 0.2 mSv, and for 68Ga-FAPI-74 it was 1.6 mSv. Thus, the radiation burden of a diagnostic 18F-FAPI-74 PET scan is even lower than that of PET scans with 18F-FDG and other 18F tracers; 68Ga-FAPI-74 is comparable to other 68Ga ligands. FAPI PET/CT supported target volume definition for guiding radiotherapy. Conclusion: The high contrast and low radiation burden of FAPI-74 PET/CT favor multiple clinical applications. Centralized large-scale production of 18F-FAPI-74 or decentralized cold-kit labeling of 68Ga-FAPI-74 allows flexible routine use.


Subject(s)
Aluminum Compounds/chemistry , Fluorides/chemistry , Gallium Radioisotopes/chemistry , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Positron Emission Tomography Computed Tomography , Aged , Biological Transport , Female , Humans , Isotope Labeling , Male , Middle Aged , Radiometry , Temperature , Tissue Distribution
11.
Eur J Nucl Med Mol Imaging ; 37(12): 2286-97, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20680269

ABSTRACT

PURPOSE: Dysregulation of histone acetylation associated with an up-regulation of histone deacetylase (HDAC) activity is common in malignant tumours. Therefore, HDAC inhibitors were developed whose effects on proliferation and apoptosis have been shown in different tumour entities. Since non-iodide-concentrating thyroid carcinomas represent a therapeutic problem, this study addressed the effects of the HDAC inhibitor MS-275 on thyroid carcinoma cells. METHODS: After the antiproliferative effect of MS-275 had been proven in different human and rat thyroid carcinoma cell lines, FRO82-2, SW1736 and FTC133 cells were further investigated with respect to changes in apoptosis, cell cycle and metabolism by the annexin V/propidium iodide assay, FACS analysis and uptake experiments employing 3-O-methyl-D-(3H)glucose, fluoro-2-deoxy-D-glucose2 [5,6-(3)H] and 14C-aminoisobutyric acid (AIB). The induction of iodide transport and gene expression were investigated in 125iodide uptake experiments and real-time polymerase chain reaction (PCR). RESULTS: MS-275 induced a concentration- and time-dependent inhibition of proliferation in the thyroid carcinoma cell lines with varying IC50 values. In FRO82-2, SW1736 and FTC133 cells characterized by low, moderate and high sensitivity an up-regulation of p21CIP/WAF1 expression and G1 and/or G2 phase arrest were observed upon MS-275 exposure corresponding to the sensitivity of individual cell lines. In addition, high MS-275 concentrations increased the apoptotic cell fraction of FTC133 and SW1736 cells, whereas resistance to apoptosis and simultaneous up-regulation of Bcl-2 gene expression were observed in FRO82-2 cells. MS-275 treatment also mediated a concentration-dependent decrease of 3H-FDG uptake and an increased 3-O-methyl-D-(3H)glucose uptake in all thyroid carcinoma cell lines after 24 h, an increased uptake of both tracers in FTC133 cells after 48 h, and restored the functional activity of the sodium-iodide symporter in SW1736 and FTC133 cells up to 20- and 45-fold. CONCLUSION: MS-275 exerts dose-dependent antiproliferative effects including growth arrest, differentiation and apoptosis in some thyroid carcinoma cell lines and might, therefore, be considered for the treatment of anaplastic and non-iodide-concentrating thyroid carcinomas.


Subject(s)
Benzamides/therapeutic use , Pyridines/therapeutic use , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Female , Histone Deacetylase Inhibitors/therapeutic use , Humans , Male , Radionuclide Imaging , Rats , Treatment Outcome
12.
Mol Pharm ; 7(6): 2224-31, 2010 Dec 06.
Article in English | MEDLINE | ID: mdl-20845937

ABSTRACT

Cell-penetrating peptides (CPPs) are able to penetrate the cell membrane carrying cargoes such as peptides, proteins, oligonucleotides, siRNAs, radioisotopes, liposomes, and nanoparticles. Consequently, many delivery approaches have been developed to use CPPs as tools for drug delivery. However, until now a systematic analysis of their in vivo properties including potential tumor binding specificity for drug targeting purposes has not been conducted. Ten of the most commonly applied CPPs were obtained by solid phase peptide synthesis and labeled with (111)In or (68)Ga. Uptake studies were conducted using a panel of six tumor cell lines of different origin. The stability of the peptides was examined in human serum. Biodistribution experiments were conducted in nude mice bearing human prostate carcinoma. Finally, positron emission tomography (PET) measurements were performed in male Wistar rats. The in vitro uptake studies revealed high cellular uptake values, but no specificity toward any of the cell lines. The biodistribution in PC-3 tumor-bearing nude mice showed a high transient accumulation in well-perfused organs and a rapid clearance from the blood. All of the CPPs revealed a relatively low accumulation rate in the brain. The highest uptake values were observed in the liver (with a maximal uptake of 51 %ID/g observed for oligoarginine (R(9))) and the kidneys (with a maximal uptake of 94 %ID/g observed for NLS). The uptake values in the PC-3 tumor were low at all time points, indicating a lack of tumor specific accumulation for all peptides studied. A micro-PET imaging study with (68)Ga-labeled penetratin, Tat and transportan(10) (TP(10)) confirmed the organ distribution data. These data reveal that CPPs do not show evidence for application in tumor targeting purposes in vivo. However, CPPs readily penetrate into most organs and show rapid clearance from the circulation. The high uptake rates observed in vitro and the relatively low specificity in vivo imply that CPPs would be better suited for topical application in combination with cargoes which show passive targeting and dominate the pharmacokinetic behavior. In conclusion, CPPs are suitable as drug carriers for in vivo application provided that their pharmacokinetic properties are also considered in design of CPP drug delivery systems.


Subject(s)
Cell-Penetrating Peptides/pharmacokinetics , Neoplasms, Experimental/metabolism , Animals , Cell-Penetrating Peptides/blood , Cell-Penetrating Peptides/chemistry , Female , Humans , Male , Mice , Mice, Nude , Neoplasms, Experimental/chemistry , Positron-Emission Tomography , Rats , Rats, Wistar , Staining and Labeling , Tissue Distribution , Tumor Cells, Cultured
13.
J Control Release ; 322: 200-208, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32184098

ABSTRACT

Monoclonal antibodies (mAbs) are increasingly exploited as vehicles for the targeted delivery of cytotoxic drugs. In antibody-drug conjugates (ADCs) antibodies specifically deliver cytotoxic compounds to cancer cells. Here, we present a technology for elevating the intracellular delivery of antibodies by the conjugation of tetrameric cell-penetrating peptides (tCPPs). The solid phase synthesis of tCPPs and their application in a chemical modification strategy for mAbs provides constructs that attain up to fourfold elevated internalization rates while retaining the mAbs target specificity. The antigen independent internalization is accompanied by beneficial pharmacokinetics limiting off-target accumulation. Applicability was proven for matuzumab, trastuzumab and the ADC Kadcyla®. Cytotoxicity studies of tCPP-conjugates of Kadcyla® resulted in a sixfold increased cytotoxicity proving the potential of chemical modification strategies to extend the applicability of biologicals. This constitutes a significant step towards next-generation antibody-based therapeutics.


Subject(s)
Antineoplastic Agents , Cell-Penetrating Peptides , Immunoconjugates , Antibodies, Monoclonal , Chemical Engineering , Trastuzumab
14.
J Nucl Med ; 61(10): 1507-1513, 2020 10.
Article in English | MEDLINE | ID: mdl-32169911

ABSTRACT

Most epithelial tumors recruit fibroblasts and other nonmalignant cells and activate them into cancer-associated fibroblasts. This often leads to overexpression of the membrane serine protease fibroblast-activating protein (FAP). It has already been shown that DOTA-bearing FAP inhibitors (FAPIs) generate high-contrast images with PET/CT scans. Since SPECT is a lower-cost and more widely available alternative to PET, 99mTc-labeled FAPIs represent attractive tracers for imaging applications in a larger number of patients. Furthermore, the chemically homologous nuclide 188Re is available from generators, which allows FAP-targeted endoradiotherapy. Methods: For the preparation of 99mTc-tricarbonyl complexes, a chelator was selected whose carboxylic acids can easily be converted into various derivatives in the finished product, enabling a platform strategy based on the original tracer. The obtained 99mTc complexes were investigated in vitro by binding and competition experiments on FAP-transfected HT-1080 (HT-1080-FAP) or on mouse FAP-expressing (HEK-muFAP) and CD26-expressing (HEKCD26) HEK cells and characterized by planar scintigraphy and organ distribution studies in tumor-bearing mice. Furthermore, a first-in-humans application was done on 2 patients with ovarian and pancreatic cancer, respectively. Results:99mTc-FAPI-19 showed specific binding to recombinant FAP-expressing cells with high affinity. Unfortunately, liver accumulation, biliary excretion, and no tumor uptake were observed on planar scintigraphy for a HT-1080-FAP-xenotransplanted mouse. To improve the pharmacokinetic properties, hydrophilic amino acids were attached to the chelator moiety of the compound. The resulting 99mTc-labeled FAPI tracers revealed excellent binding properties (≤45% binding; >95% internalization), high affinity (half-maximal inhibitory concentration, 6.4-12.7 nM), and significant tumor uptake (≤5.4% injected dose per gram of tissue) in biodistribution studies. The lead candidate 99mTc-FAPI-34 was applied for diagnostic scintigraphy and SPECT of patients with metastasized ovarian and pancreatic cancer for follow-up to therapy with 90Y-FAPI-46. 99mTc-FAPI-34 accumulated in the tumor lesions, as also shown on PET/CT imaging using 68Ga-FAPI-46. Conclusion:99mTc-FAPI-34 represents a powerful tracer for diagnostic scintigraphy, especially when PET imaging is not available. Additionally, the chelator used in this compound allows labeling with the therapeutic nuclide 188Re, which is planned for the near future.


Subject(s)
Gelatinases/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Radioisotopes/therapeutic use , Radiopharmaceuticals/pharmacokinetics , Rhenium/therapeutic use , Single Photon Emission Computed Tomography Computed Tomography/methods , Technetium/pharmacokinetics , Animals , Cells, Cultured , Drug Design , Endopeptidases , Humans , Liver/metabolism , Mice , Mice, Inbred BALB C , Quinolines/pharmacokinetics , Serine Endopeptidases
15.
J Nucl Med ; 50(3): 426-34, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19223422

ABSTRACT

UNLABELLED: Combination therapy has improved the quality of life for patients with squamous cell carcinomas of the head and neck (HNSCCs) but has not decisively changed prognosis. Targeted therapies, which enhance accumulation of the drug in the tumor, may be realized using tumor-specific binding peptides. This paper identifies and characterizes an HNSCC affine peptide. METHODS: From a phage library comprising 10(9) different displayed peptides, 1 peptide was enriched after 5 in vitro selection rounds on HNO223 tumor cells. Subsequently, the gained peptide sequence H(2)N-SPRGDLAVLGHKY-CONH(2) (HBP-1) was synthesized as an amide and labeled with (125)I. In vitro studies for binding kinetics and competition were performed with 5 different HNSCC cell lines. Furthermore, the stability of the peptide was evaluated in human serum. The in vivo biodistribution of (131)I-labeled peptide was determined in HNSCC tumor-bearing nude mice. The results were further validated in human HNSCC tumor tissue sections using fluorescence-labeled HBP-1. Competition experiments were performed to determine the binding sequence and validate the target. RESULTS: The HBP-1 motif was enriched in 62% of all phages sequenced. Labeled (125)I-HBP-1 showed binding to 5 different HNSCC cell lines and a maximum binding to HNO97 cells, with 11% of the applied dose per 10(6) cells and an inhibitory concentration of 50% of 38.9 nM. Stability experiments in human serum showed a half-life of 55 min. In 2 different HNSCC tumor xenografts, (131)I-HBP-1 accumulated rapidly, with stable uptake until 45 min after intravenous application. Peptide immunohistochemistry of HNSCC tissue sections exhibited tumor staining by HBP-1, whereas normal tissue remained negative. Sequence mutation and competition experiments revealed that the intrinsic RGD motif in combination with the intrinsic LXXL motif is responsible for the binding ability of HBP1. The RGDLXXL sequence within this peptide is known and indicates that binding occurs via the alpha(v)beta(6) rather than the alpha(v)beta(3) integrin. CONCLUSION: Within the sequence of HBP-1 is a RGDLXXL motif, and most likely it is targeting the alpha(v)beta(6) receptor of the integrin family of cell adhesion receptors. HBP-1 represents a promising lead structure for the development of targeted therapies or diagnostic procedures in patients with HNSCC.


Subject(s)
Head and Neck Neoplasms/diagnostic imaging , Oligopeptides/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Amino Acid Sequence , Animals , Cell Line, Tumor , Head and Neck Neoplasms/metabolism , Humans , Iodine Radioisotopes , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Neoplasm Transplantation , Peptide Library , Radionuclide Imaging
17.
EJNMMI Radiopharm Chem ; 4(1): 16, 2019 Jul 25.
Article in English | MEDLINE | ID: mdl-31659499

ABSTRACT

Tumors form a complex environment consisting of a variety of non-malignant cells. Especially cancer-associated fibroblasts have been shown to have an important role for different aspects of malignant tumors such as migration, metastasis, resistance to chemotherapy and immunosuppression. Therefore, a targeting of these cells may be useful for both imaging and therapy. In this respect, an interesting target is the fibroblast activation protein (FAP) which is expressed in activated fibroblasts, but not in quiescent fibroblasts, giving the opportunity to use this membrane-anchored enzyme as a target for radionuclide-based approaches for diagnosis and treatment of tumors and for the diagnosis of non-malignant disease associated with a remodelling of the extracellular matrix.

18.
J Nucl Med ; 60(10): 1421-1429, 2019 10.
Article in English | MEDLINE | ID: mdl-30850501

ABSTRACT

Cancer-associated fibroblasts constitute a vital subpopulation of the tumor stroma and are present in more than 90% of epithelial carcinomas. The overexpression of the serine protease fibroblast activation protein (FAP) allows a selective targeting of a variety of tumors by inhibitor-based radiopharmaceuticals (FAPIs). Of these compounds, FAPI-04 has been recently introduced as a theranostic radiotracer and demonstrated high uptake into different FAP-positive tumors in cancer patients. To enable the delivery of higher doses, thereby improving the outcome of a therapeutic application, several FAPI variants were designed to further increase tumor uptake and retention of these tracers. Methods: Novel quinoline-based radiotracers were synthesized by organic chemistry and evaluated in radioligand binding assays using FAP-expressing HT-1080 cells. Depending on their in vitro performance, small-animal PET imaging and biodistribution studies were performed on HT-1080-FAP tumor-bearing mice. The most promising compounds were used for clinical PET imaging in 8 cancer patients. Results: Compared with FAPI-04, 11 of 15 FAPI derivatives showed improved FAP binding in vitro. Of these, 7 compounds demonstrated increased tumor uptake in tumor-bearing mice. Moreover, tumor-to-normal-organ ratios were improved for most of the compounds, resulting in images with higher contrast. Notably two of the radiotracers, FAPI-21 and -46, displayed substantially improved ratios of tumor to blood, liver, muscle, and intestinal uptake. A first diagnostic application in cancer patients revealed high intratumoral uptake of both radiotracers already 10 min after administration but a higher uptake in oral mucosa, salivary glands, and thyroid for FAPI-21. Conclusion: Chemical modification of the FAPI framework enabled enhanced FAP binding and improved pharmacokinetics in most of the derivatives, resulting in high-contrast images. Moreover, higher doses of radioactivity can be delivered while minimizing damage to healthy tissue, which may improve therapeutic outcome.


Subject(s)
Gelatinases/chemistry , Membrane Proteins/chemistry , Neoplasms/diagnostic imaging , Neoplasms/therapy , Quinolines/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Serine Endopeptidases/chemistry , Animals , Cell Line , Cell Line, Tumor , Chelating Agents/pharmacology , Endopeptidases , Humans , Image Processing, Computer-Assisted , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Positron-Emission Tomography , Protein Binding , Quinolines/chemistry , Solvents , Treatment Outcome
19.
Nuklearmedizin ; 58(4): 309-318, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31075798

ABSTRACT

INTRODUCTION: Integrin αvß6 shows a high expression rate in several cancer entities. As it is absent in most healthy adult tissues, it represents a promising target for tumor targeting with peptidic radiotracers. This study was performed to pave the way of the recently published αvß6-binding peptide SFLAP3 for the clinical application in patients with pancreatic cancer. METHODS: The expression of integrin αvß6 on several pancreatic cancer cell lines was assessed using flow cytometry and cell binding assays. The affinity was determined in competition binding assays followed by internalization and efflux studies. To increase the affinity, the binding sequence was modified and trimerization of the SFLAP3 peptide was achieved by oxime ligation. PET and biodistribution assays were conducted in Capan-2 tumor bearing mice. Finally, a first pancreatic tumor patient was examined with 68Ga-DOTA-SFLAP3. RESULTS: Flow cytometric analysis and IN VITRO: cell binding revealed high expression of integrin αvß6 on most pancreatic tumor cell lines. Modification of SFLAP3 led to compounds with improved IN VITRO: binding properties. Unfortunately, these superior properties could not be transferred into improved pharmacokinetics. Consequently, the first pancreatic tumor patient was examined with 68Ga-DOTA-SFLAP3. The PET revealed specific accumulation (with SUV(max) values in the metastases ranging from 5 to 10) and a long retention in the tumor. CONCLUSION: SFLAP3 showed high affinity to integrin αvß6 on pancreatic cancer cell lines. The IN VITRO: performance could be confirmed in tumor bearing mice and by PET imaging. These data suggest that DOTA-SFLAP3 is a promising tracer for targeting αvß6-expressing pancreatic tumors.


Subject(s)
Antigens, Neoplasm/metabolism , Integrins/metabolism , Oligopeptides/chemistry , Pancreatic Neoplasms/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Mice , Mice, Inbred BALB C , Oligopeptides/metabolism , Oligopeptides/pharmacokinetics , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/pathology , Radioactive Tracers , Tissue Distribution , Pancreatic Neoplasms
20.
Mol Imaging Biol ; 21(5): 973-983, 2019 10.
Article in English | MEDLINE | ID: mdl-30671741

ABSTRACT

PURPOSE: Targeted therapies are regarded as promising approaches to increase 5-year survival rate of non-small cell lung cancer (NSCLC) patients. Here, we investigated the clinical value of the αvß6 integrin-specific peptide SFITGv6 as a diagnostic reagent targeting NSCLC. METHODS: Affinity and binding properties of [125I]SFITGv6 or [177Lu]SFITGv6 for αvß6 integrin-expressing NSCLC cell lines were evaluated in cell culture experiments including competition, kinetic, internalization, and efflux. To confirm αvß6 integrin specificity in vivo small-animal positron emission tomography (PET) imaging using [68Ga]SFITGv6 as radiotracer and biodistribution of [177Lu]SFITGv6 in NCI-H2009 and NCI-H322 tumor-bearing mice was performed. Finally, to distinguish between benign and malignant lesions [68Ga]SFITGv6 was applied as radiotracer for PET/x-ray computed tomography (CT) imaging of NSCLC patients with unclear diagnosis upon routinely performed 2-deoxy-2-[18F]flouro-D-glucose ([18F]FDG)-PET/CT. The biodistribution of the SFITGv6-ligand in different organs and tumor lesions of NSCLC patients was quantified 1 h and 3 h after injection measuring standard uptake values (SUV)max. RESULTS: In vitro experiments revealed a significant time-dependent SFITGv6 binding of up to 33 % to αvß6 integrin-expressing the cell lines NCI-H2009, NCI-H322, NCI-H292, NCI-H358, and high affinity (IC50-mean 3.1 nM) to NCI-H2009 and NCI-H322. Moreover, a fast internalization of approximately 66 % by NCI-H2009 and NCI-H322 cells was observed. Small-animal PET imaging and biodistribution experiments of NCI-H2009 and NCI-H322 xenografts demonstrated an increased tumor-specific accumulation of SFITGv6 40 to 60 min after injection. Finally, PET/CT scans of NSCLC patients after [18F] FDG injection followed by [68Ga]SFITGv6 application revealed correlating images. Comparing the uptake of [68Ga]SFITGv6 and [18F] FDG both PET/CT-examinations presented with significantly increased SUVmax values in histologically proven NSCLC lesions, but a generally higher accumulation of [18F] FDG was noticed. CONCLUSIONS: Even if SFITGv6 demonstrates excellent affinity and specificity for αvß6 integrin-expressing NSCLC cell lines and several NSCLC xenografts [18F]FDG-PET/CT provides an advantage over [68Ga]SFITGv6-PET/CT for the diagnosis of NSCLC patients.


Subject(s)
Antigens, Neoplasm/metabolism , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Integrins/metabolism , Lung Neoplasms/diagnostic imaging , Peptide Fragments/metabolism , Positron Emission Tomography Computed Tomography , Animals , Cell Line, Tumor , Fluorodeoxyglucose F18 , Gallium Radioisotopes/chemistry , Humans , Mice, Inbred BALB C , Mice, Nude , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL