Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 14: 1285923, 2023.
Article in English | MEDLINE | ID: mdl-38035094

ABSTRACT

Intra-operative fluorescence imaging has demonstrated its ability to improve tumor lesion identification. However, the limited tissue penetration of the fluorescent signals hinders the detection of deep-lying or occult lesions. Integrating fluorescence imaging with SPECT and/or intra-operative gamma-probing synergistically combines the deep tissue penetration of gamma rays for tumor localization with the precision of fluorescence imaging for precise tumor resection. In this study, we detail the use of a genetically encoded multifunctional handle, henceforth referred to as a GEM-handle, for the development of fluorescent/radioactive bimodal single-domain antibody (sdAb)-based tracers. A sdAb that targets the urokinase plasminogen activator receptor (uPAR) was engineered to carry a GEM-handle containing a carboxy-terminal hexahistidine-tag and cysteine-tag. A two-step labeling strategy was optimized and applied to site-specifically label IRDye800CW and 99mTc to the sdAb. Bimodal labeling of the sdAbs proved straightforward and successful. 99mTc activity was however restricted to 18.5 MBq per nmol fluorescently-labeled sdAb to prevent radiobleaching of IRDye800CW without impeding SPECT/CT imaging. Subsequently, the in vivo biodistribution and tumor-targeting capacity of the bimodal tracer were evaluated in uPAR-positive tumor-bearing mice using SPECT/CT and fluorescence imaging. The bimodal sdAb showed expected renal background signals due to tracer clearance, along with slightly elevated non-specific liver signals. Four hours post-injection, both SPECT/CT and fluorescent images achieved satisfactory tumor uptake and contrast, with significantly higher values observed for the anti-uPAR bimodal sdAb compared to a control non-targeting sdAb. In conclusion, the GEM-handle is a convenient method for designing and producing bimodal sdAb-based tracers with adequate in vivo characteristics.


Subject(s)
Neoplasms , Single-Domain Antibodies , Animals , Mice , Fluorescent Dyes , Tissue Distribution , Tomography, Emission-Computed, Single-Photon/methods , Neoplasms/diagnostic imaging
2.
Int Rev Cell Mol Biol ; 369: 143-199, 2022.
Article in English | MEDLINE | ID: mdl-35777863

ABSTRACT

Cancer is a heterogeneous disease, requiring treatment tailored to the unique phenotype of the patient's tumor. Monoclonal antibodies (mAbs) and variants thereof have enabled targeted therapies to selectively target cancer cells. Cancer cell-specific mAbs have been used for image-guided surgery and targeted delivery of radionuclides or toxic agents, improving classical treatment strategies. Cancer cell-specific mAbs can further inhibit tumor cell growth or can stimulate immune-mediated destruction of cancer cells, a feature that has also been achieved through mAb-mediated manipulation of immune cells and pathways. Drawbacks of mAbs and their variants, together with the discovery of camelid heavy chain-only antibodies and the many advantageous features of their variable domains, referred to as VHHs, single domain antibodies or nanobodies (Nbs), resulted in the exploration of Nbs as an alternative targeting moiety. We therefore review the state-of-the-art as well as novel exploitation strategies of Nbs for targeted cancer therapy.


Subject(s)
Neoplasms , Single-Domain Antibodies , Antibodies, Monoclonal , Humans , Neoplasms/drug therapy , Single-Domain Antibodies/genetics , Single-Domain Antibodies/therapeutic use
3.
Nature ; 604(7904): 195-201, 2022 04.
Article in English | MEDLINE | ID: mdl-35355017

ABSTRACT

Hyaluronan is an acidic heteropolysaccharide comprising alternating N-acetylglucosamine and glucuronic acid sugars that is ubiquitously expressed in the vertebrate extracellular matrix1. The high-molecular-mass polymer modulates essential physiological processes in health and disease, including cell differentiation, tissue homeostasis and angiogenesis2. Hyaluronan is synthesized by a membrane-embedded processive glycosyltransferase, hyaluronan synthase (HAS), which catalyses the synthesis and membrane translocation of hyaluronan from uridine diphosphate-activated precursors3,4. Here we describe five cryo-electron microscopy structures of a viral HAS homologue at different states during substrate binding and initiation of polymer synthesis. Combined with biochemical analyses and molecular dynamics simulations, our data reveal how HAS selects its substrates, hydrolyses the first substrate to prime the synthesis reaction, opens a hyaluronan-conducting transmembrane channel, ensures alternating substrate polymerization and coordinates hyaluronan inside its transmembrane pore. Our research suggests a detailed model for the formation of an acidic extracellular heteropolysaccharide and provides insights into the biosynthesis of one of the most abundant and essential glycosaminoglycans in the human body.


Subject(s)
Hyaluronan Synthases , Hyaluronic Acid , Phycodnaviridae , Cryoelectron Microscopy , Hyaluronan Synthases/metabolism , Phycodnaviridae/enzymology , Polymers
4.
Methods Mol Biol ; 2446: 395-407, 2022.
Article in English | MEDLINE | ID: mdl-35157285

ABSTRACT

Near-infrared fluorescence molecular imaging has become an established preclinical technique to investigate molecular processes in vivo and to study novel therapies. Furthermore, fluorescence molecular imaging is gaining significant interest from clinicians as an intra-operative guidance tool. This technique makes use of targeted fluorescent tracers as contrast agents that recognize specific biomarkers expressed at the site of disease. Single-domain antibodies have shown to possess excellent properties for in vivo imaging in comparison to conventional antibodies. In this chapter, we describe a method for site-specific conjugation of a near-infrared fluorophore to single-domain antibodies by exploiting cysteine-maleimide chemistry. As opposed to random conjugation, site-specific conjugation results in a homogenously labeled fluorescent tracer and avoids inference with antigen binding.


Subject(s)
Single-Domain Antibodies , Surgery, Computer-Assisted , Cell Line, Tumor , Fluorescent Dyes/chemistry , Molecular Imaging/methods , Optical Imaging/methods , Single-Domain Antibodies/chemistry
5.
Biomolecules ; 11(3)2021 02 26.
Article in English | MEDLINE | ID: mdl-33652977

ABSTRACT

Intraoperative guidance using targeted fluorescent tracers can potentially provide surgeons with real-time feedback on the presence of tumor tissue in resection margins. To overcome the limited depth penetration of fluorescent light, combining fluorescence with SPECT/CT imaging and/or gamma-ray tracing has been proposed. Here, we describe the design and preclinical validation of a novel bimodal nanobody-tracer, labeled using a "multifunctional single attachment point" (MSAP) label, integrating a Cy5 fluorophore and a diethylenetriaminepentaacetic acid (DTPA) chelator into a single structure. After conjugation of the bimodal MSAP to primary amines of the anti-HER2 nanobody 2Rs15d and 111In-labeling of DTPA, the tracer's characteristics were evaluated in vitro. Subsequently, its biodistribution and tumor targeting were assessed by SPECT/CT and fluorescence imaging over 24 h. Finally, the tracer's ability to identify small, disseminated tumor lesions was investigated in mice bearing HER2-overexpressing SKOV3.IP1 peritoneal lesions. [111In]In-MSAP.2Rs15d retained its affinity following conjugation and remained stable for 24 h. In vivo SPECT/CT and fluorescence images showed specific uptake in HER2-overexpressing tumors with low background. High tumor-to-muscle ratios were obtained at 1h p.i. and remained 19-fold on SPECT/CT and 3-fold on fluorescence images over 24 h. In the intraperitoneally disseminated model, the tracer allowed detection of larger lesions via nuclear imaging, while fluorescence enabled accurate removal of submillimeter lesions. Bimodal nuclear/fluorescent nanobody-tracers can thus be conveniently designed by conjugation of a single-molecule MSAP-reagent carrying a fluorophore and chelator for radioactive labeling. Such tracers hold promise for clinical applications.


Subject(s)
Single-Domain Antibodies/chemistry , Surgery, Computer-Assisted , Animals , CHO Cells , Cell Line, Tumor , Cricetulus , Humans , Mice , Neoplasms/diagnostic imaging , Neoplasms/pathology , Radiopharmaceuticals/chemistry , Tissue Distribution , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Xenograft Model Antitumor Assays
6.
Chem Biol Interact ; 284: 126-136, 2018 Mar 25.
Article in English | MEDLINE | ID: mdl-29458015

ABSTRACT

Type 2 diabetes mellitus (T2DM) is characterised not only by hyperglycaemia and insulin resistance but also an impaired balance between the processes of coagulation and fibrinolysis. The aim of this study was to examine the effects of metformin, a widely-used oral anti-diabetic drug, phenformin and eight sulfenamide and sulfonamide derivatives of metformin on several haemostasis parameters. Thrombin Time (TT) tests were performed according to the available commercial method. The activity of factor X was conducted based on deficient plasma factor X. The activity of two main enzymes involved in haemostasis, thrombin and plasmin, was measured spectrophotometrically with chromogenic substrates. Protein C and antithrombin III (AT) activity assays using chromogenic substrates were conducted to determine the effect of the derivatives of metformin on these both naturally occurring anticoagulants. Two of the compounds, sulfenamide with hexyl tail and para-nitro-benzenesulfonamide significantly shortened TT. ortho-nitro sulfonamide at a concentration of 0.3-1.5 µmol/mL contributed to a significant decrease in the activity of factor X. However, sulfenamides with cyclohexyl, butyl and branched ethyl-hexyl tails at 1.5 of µmol/mL increased its activity, and simultaneously shortened PT. Additionally, ortho-nitro-benzenesulfonamide at concentrations of 1.5 µmol/mL was found to significantly decrease reaction velocity (↓ dA/dt) in the thrombin activity assay. On contrary, it was noticed that branched sulfenamide at the concentration of 1.5 µmol/mL significantly increased the enzymatic activity of plasmin. Metformin, phenformin and octyl and butyl sulfenamides were associated with a significant increase in the activity of AT. Hexyl sulfenamide and para-nitro- as well as para-trifluoro-ortho-nitro-benzenesulfonamide contributed to the decrease in the activity of protein C, while the other tested compounds did not affect its activity. In conclusion, 2-nitro-benzenesulfonamide derivative of metformin presents highly beneficial anticoagulant properties. This compound is therefore promising candidate for further in vitro and in vivo studies.


Subject(s)
Anticoagulants/chemistry , Blood Coagulation/drug effects , Fibrinolysis/drug effects , Metformin/chemistry , Sulfamerazine/chemistry , Sulfonamides/chemistry , Anticoagulants/pharmacology , Antithrombin III/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Factor X/metabolism , Humans , Metformin/pharmacology , Protein C/metabolism , Sulfamerazine/pharmacology , Sulfonamides/pharmacology , Thrombin Time
7.
Oxid Med Cell Longev ; 2017: 7303096, 2017.
Article in English | MEDLINE | ID: mdl-28770024

ABSTRACT

The results of epidemiological and pathophysiological studies suggest that type 2 diabetes mellitus (T2DM) may predispose to Alzheimer's disease (AD). The two conditions present similar glucose levels, insulin resistance, and biochemical etiologies such as inflammation and oxidative stress. The diabetic state also contributes to increased acetylcholinesterase (AChE) activity, which is one of the factors leading to neurodegeneration in AD. The aim of this study was to assess in vitro the effects of metformin, phenformin, and metformin sulfenamide prodrugs on the activity of human AChE and butyrylcholinesterase (BuChE) and establish the type of inhibition. Metformin inhibited 50% of the AChE activity at micromolar concentrations (2.35 µmol/mL, mixed type of inhibition) and seemed to be selective towards AChE since it presented low anti-BuChE activity. The tested metformin prodrugs inhibited cholinesterases (ChE) at nanomolar range and thus were more active than metformin or phenformin. The cyclohexyl sulfenamide prodrug demonstrated the highest activity towards both AChE (IC50 = 890 nmol/mL, noncompetitive inhibition) and BuChE (IC50 = 28 nmol/mL, mixed type inhibition), while the octyl sulfenamide prodrug did not present anti-AChE activity, but exhibited mixed inhibition towards BuChE (IC50 = 184 nmol/mL). Therefore, these two bulkier prodrugs were concluded to be the most selective compounds for BuChE over AChE. In conclusion, it was demonstrated that biguanides present a novel class of inhibitors for AChE and BuChE and encourages further studies of these compounds for developing both selective and nonselective inhibitors of ChEs in the future.


Subject(s)
Butyrylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Metformin/chemistry , Prodrugs/chemistry , Sulfamerazine/chemistry , Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/enzymology , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Humans , Male , Metformin/pharmacology , Prodrugs/pharmacology , Sulfamerazine/pharmacology
8.
Eur J Pharmacol ; 811: 208-221, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28606852

ABSTRACT

Although metformin, an oral anti-diabetic drug, has been found to have multidirectional effects over the past decade, it is characterised by unfavourable pharmacokinetic properties. This study discusses the effects of metformin, phenformin and three prodrugs of metformin on the haemostasis and integrity of Red Blood Cells (RBCs). The influence of examined biguanide derivatives on haemostasis was evaluated spectrophotometrically by clot formation and lysis test (CL-test) at 405nm. The extrinsic and intrinsic coagulation pathway were examined by measuring the PT (Prothrombin Time) and aPTT (Activated Partial Tromboplastin Time). Haemolysis assay, microscopy and flow cytometry studies were used to assess the effect of the tested compounds on RBCs. Although none of the tested biguanide derivatives significantly influenced the overall potential of clot formation and fibrinolysis (CLAUC constants), statistically significant changes were seen in the values of the kinetic parameters of fibrinolysis. Furthermore, only prodrug 2, with an 8-carbon alkyl chain, unfavourably affected RBCs by interaction with the erythrocyte membrane leading to significant haemolysis. Our results provide a further insight into the effects of metformin and its prodrugs on haemostasis and RBCs and underscore the necessity for further research.


Subject(s)
Erythrocyte Membrane/drug effects , Hemostasis/drug effects , Metformin/metabolism , Prodrugs/pharmacology , Blood Coagulation/drug effects , Humans , Prodrugs/metabolism
9.
Curr Pharm Des ; 23(17): 2532-2550, 2017.
Article in English | MEDLINE | ID: mdl-27908266

ABSTRACT

Metformin, a synthetic biguanide, is currently one of the most frequently recommended medications for type 2 diabetes treatment around the world. This review presents the latest discoveries in the pharmacokinetics of metformin, especially the role of transporters (e.g. Organic Cation Transporters OCTs, Multidrug and Toxin Extrusion transporters MATE) in oral absorption, distribution, elimination and biochemical effects of metformin in humans. We also review the associations between genetic variations of metformin transporters, their pharmacokinetics and drug efficacy or drug responses. In the second part of this paper, we highlight the current knowledge on novel metformin actions including favourable effects on lipid profile (e.g. decreasing plasma triglycerides (TG) and low density lipoprotein (LDL) cholesterol levels) and the cardiovascular system (e.g. decline in systolic and diastolic blood pressure, and vasoprotective effects). Furthermore, we provide an up-to-date overview of multidirectional activities of metformin, including the effects on coagulation and fibrinolysis, polycystic ovary syndrome, as well as the anti-ageing and antiinflammatory properties. Over the past two decades, metformin's antineoplastic properties have been drawing increasing attention of scientists; herein, we outline the state-of-the-art discoveries concerning metformin use in the field of oncology. Finally, we review the newly synthesized derivatives and pro-drugs of metformin and other biguanides.


Subject(s)
Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/pharmacokinetics , Metformin/pharmacology , Metformin/pharmacokinetics , Carrier Proteins/genetics , Diabetes Mellitus, Type 2/drug therapy , Genetic Variation , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...