Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
PLoS One ; 17(2): e0261925, 2022.
Article in English | MEDLINE | ID: mdl-35143514

ABSTRACT

PURPOSE: Vitreous humor is a complex biofluid whose composition determines its structure and function. Vitreous viscosity will affect the delivery, distribution, and half-life of intraocular drugs, and key physiological molecules. The central pig vitreous is thought to closely match human vitreous viscosity. Diffusion is inversely related to viscosity, and diffusion is of fundamental importance for all biochemical reactions. Fluorescence Recovery After Photobleaching (FRAP) may provide a novel means of measuring intravitreal diffusion that could be applied to drugs and physiological macromolecules. It would also provide information about vitreous viscosity, which is relevant to drug elimination, and delivery. METHODS: Vitreous viscosity and intravitreal macromolecular diffusion of fluorescently labelled macromolecules were investigated in porcine eyes using fluorescence recovery after photobleaching (FRAP). Fluorescein isothiocyanate conjugated (FITC) dextrans and ficolls of varying molecular weights (MWs), and FITC-bovine serum albumin (BSA) were employed using FRAP bleach areas of different diameters. RESULTS: The mean (±standard deviation) viscosity of porcine vitreous using dextran, ficoll and BSA were 3.54 ± 1.40, 2.86 ± 1.13 and 4.54 ± 0.13 cP respectively, with an average of 3.65 ± 0.60 cP. CONCLUSIONS: FRAP is a feasible and practical optical method to quantify the diffusion of macromolecules through vitreous.


Subject(s)
Fluorescence Recovery After Photobleaching/methods , Vitreous Body/metabolism , Animals , Bevacizumab/chemistry , Bevacizumab/metabolism , Dextrans/chemistry , Diffusion , Ficoll/chemistry , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/chemistry , Ranibizumab/chemistry , Ranibizumab/metabolism , Receptors, Vascular Endothelial Growth Factor/chemistry , Receptors, Vascular Endothelial Growth Factor/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Serum Albumin, Bovine/chemistry , Swine , Viscosity
2.
Mol Pharm ; 18(6): 2208-2217, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34014104

ABSTRACT

The current standard of care for antivascular endothelial growth factor (VEGF) treatment requires frequent intravitreal (IVT) injections of protein therapeutics, as a result of limited retention within the eye. A thorough understanding of the determinants of ocular pharmacokinetics (PK) and its translation across species is an essential prerequisite for developing more durable treatments. In this work, we studied the ocular PK in macaques of the protein formats that comprise today's anti-VEGF standard of care. Cynomolgus monkeys received a single IVT injection of a single-chain variable fragment (scFv, brolucizumab), antigen-binding fragment (Fab, ranibizumab), fragment crystallizable-fusion protein (Fc-fusion, aflibercept), or immunoglobulin G monoclonal antibody (IgG, VA2 CrossMAb). Drug concentrations were determined in aqueous humor samples collected up to 42 days postinjection using immunoassay methods. The ocular half-life (t1/2) was 2.28, 2.62, 3.13, and 3.26 days for scFv, Fab, Fc-fusion, and IgG, respectively. A correlation with human t1/2 values from the literature confirmed the translational significance of the cynomolgus monkey as an animal model for ocular research. The relation between ocular t1/2 and molecular size was also investigated. Size was inferred from the molecular weight (MW) or determined experimentally by dynamic light scattering. The MW and hydrodynamic radius were found to be good predictors for the ocular t1/2 of globular proteins. The analysis showed that molecular size is a determinant of ocular disposition and may be used in lieu of dedicated PK studies in animals.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Aqueous Humor/metabolism , Vitreous Body/metabolism , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/pharmacokinetics , Half-Life , Intravitreal Injections , Macaca fascicularis , Models, Animal , Molecular Weight , Ranibizumab/administration & dosage , Ranibizumab/chemistry , Ranibizumab/pharmacokinetics , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Receptors, Vascular Endothelial Growth Factor/chemistry , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics
3.
Molecules ; 26(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670650

ABSTRACT

Epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor (VEGFR) are two extensively studied membrane-bound receptor tyrosine kinase proteins that are frequently overexpressed in many cancers. As a result, these receptor families constitute attractive targets for imaging and therapeutic applications in the detection and treatment of cancer. This review explores the dynamic structure and structure-function relationships of these two growth factor receptors and their significance as it relates to theranostics of cancer, followed by some of the common inhibition modalities frequently employed to target EGFR and VEGFR, such as tyrosine kinase inhibitors (TKIs), antibodies, nanobodies, and peptides. A summary of the recent advances in molecular imaging techniques, including positron emission tomography (PET), single-photon emission computerized tomography (SPECT), computed tomography (CT), magnetic resonance imaging (MRI), and optical imaging (OI), and in particular, near-IR fluorescence imaging using tetrapyrrolic-based fluorophores, concludes this review.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Molecular Targeted Therapy , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , ErbB Receptors/metabolism , Humans , Molecular Imaging , Peptides/chemistry , Peptides/pharmacology , Protein Multimerization , Receptors, Vascular Endothelial Growth Factor/chemistry , Receptors, Vascular Endothelial Growth Factor/metabolism
4.
ACS Chem Biol ; 16(3): 548-556, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33621466

ABSTRACT

We report a general approach to engineering multivalent d-proteins with antibody-like activities in vivo. Mirror-image phage display and structure-guided design were utilized to create a d-protein that uses receptor mimicry to antagonize vascular endothelial growth factor A (VEGF-A). Selections against the d-protein form of VEGF-A using phage-displayed libraries of two different domain scaffolds yielded two proteins that bound distinct receptor interaction sites on VEGF-A. X-ray crystal structures of the d-protein/VEGF-A complexes were used to guide affinity maturation and to construct a heterodimeric d-protein VEGF-A antagonist with picomolar activity. The d-protein VEGF-A antagonist prevented vascular leakage in a rabbit eye model of wet age-related macular degeneration and slowed tumor growth in the MC38 syngeneic mouse tumor model with efficacies comparable to those of approved antibody drugs, and in contrast with antibodies, the d-protein was non-immunogenic during treatment and following subcutaneous immunizations.


Subject(s)
Antineoplastic Agents/chemistry , Neoplasms/drug therapy , Peptides/chemistry , Receptors, Vascular Endothelial Growth Factor/chemistry , Retinal Vessels/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacology , Bevacizumab/pharmacology , Binding Sites , Drug Evaluation, Preclinical , Eye/drug effects , Female , Humans , Mice , Models, Molecular , Peptide Library , Peptides/pharmacology , Protein Binding , Protein Conformation , Protein Multimerization , Rabbits , Receptors, Vascular Endothelial Growth Factor/metabolism
5.
Am J Ophthalmol ; 224: 36-42, 2021 04.
Article in English | MEDLINE | ID: mdl-33309691

ABSTRACT

PURPOSE: To review the biosimilars of anti-vascular endothelial growth factor agents for retinal diseases and provide an update about their development. DESIGN: Literature review. METHODS: A comprehensive literature review was performed for scientific articles, clinical trials, and press releases for the development of biosimilars in ophthalmology. RESULTS: To date, Razumab (Intas Pharmaceuticals Ltd, Ahmedabad, GJ, India) is the only approved biosimilar for ophthalmic use, but the landscape will rapidly change in the future with multiple biosimilar candidates, which are currently in phase 3 trials, showing promising early results. CONCLUSION: Biosimilars hold the potential to reduce the financial burden of the highly efficacious biologic therapy in retinal pathologies. However, the off-label bevacizumab may differentiate the success of biosimilars in different geographic regions.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Biosimilar Pharmaceuticals/therapeutic use , Retinal Diseases/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Biosimilar Pharmaceuticals/chemistry , Biosimilar Pharmaceuticals/pharmacology , Clinical Trials as Topic , Drug Approval , Drug Development , Humans , Ranibizumab/chemistry , Ranibizumab/pharmacology , Ranibizumab/therapeutic use , Receptors, Vascular Endothelial Growth Factor/chemistry , Receptors, Vascular Endothelial Growth Factor/therapeutic use , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use
6.
Nanotechnology ; 32(9): 095101, 2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33113518

ABSTRACT

Lower doses of capsaicin (8-methyl-N-vanillyl-6-nonenamide) have the potential to serve as an anticancer drug, however, due to its pungency, irritant effect, poor water solubility and high distribution volume often linked to various off-target effects, its therapeutic use is limited. This study aimed to determine the biodistribution and anticancer efficacy of capsaicin loaded solid lipid nanoparticles (SLNs) in human hepatocellular carcinoma in vitro. In this study, SLNs of stearic acid loaded with capsaicin was formulated by the solvent evaporation-emulsification technique and were instantly characterized for their encapsulation efficiency, morphology, loading capacity, stability, particle size, charge and in vitro drug release profile. Synthesized SLNs were predominantly spherical, 80 nm diameter particles that proved to be biocompatible with good stability in aqueous conditions. In vivo biodistribution studies of the formulated SLNs showed that 48 h after injection in the lateral tail vein, up to 15% of the cells in the liver, 1.04% of the cells in the spleen, 3.05% of the cells in the kidneys, 3.76% of the cells in the heart, 1.31% of the cells in the lungs and 0% of the cells in the brain of rats were determined. Molecular docking studies against the identified targets in HepG2 cells showed that the capsaicin is able to bind Abelson tyrosine-protein kinase, c-Src kinase, p38 MAP kinase and VEGF-receptor. Molecular dynamic simulation showed that capsaicin-VEGF receptor complex is highly stable at 50 nano seconds. The IC50 of capsaicin loaded SLNs in HepG2 cells in vitro was 21.36 µg × ml-1. These findings suggest that capsaicin loaded SLNs are stable in circulation for a period up to 3 d, providing a controlled release of loaded capsaicin and enhanced anticancer activity.


Subject(s)
Antineoplastic Agents/pharmacology , Capsaicin/pharmacology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , CSK Tyrosine-Protein Kinase/metabolism , Capsaicin/chemical synthesis , Capsaicin/pharmacokinetics , Carcinoma, Hepatocellular/drug therapy , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Design , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Lipids , Liver Neoplasms/drug therapy , Models, Molecular , Molecular Dynamics Simulation , Nanoparticles , Particle Size , Proto-Oncogene Proteins c-abl/metabolism , Rats , Receptors, Vascular Endothelial Growth Factor/chemistry , Solubility , Tissue Distribution , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Biomolecules ; 10(12)2020 12 15.
Article in English | MEDLINE | ID: mdl-33333800

ABSTRACT

Vascular endothelial growth factors (VEGFs) bind to membrane receptors on a wide variety of cells to regulate diverse biological responses. The VEGF-A family member promotes vasculogenesis and angiogenesis, processes which are essential for vascular development and physiology. As angiogenesis can be subverted in many disease states, including tumour development and progression, there is much interest in understanding the mechanistic basis for how VEGF-A regulates cell and tissue function. VEGF-A binds with high affinity to two VEGF receptor tyrosine kinases (VEGFR1, VEGFR2) and with lower affinity to co-receptors called neuropilin-1 and neuropilin-2 (NRP1, NRP2). Here, we use a structural viewpoint to summarise our current knowledge of VEGF-VEGFR activation and signal transduction. As targeting VEGF-VEGFR activation holds much therapeutic promise, we examine the structural basis for anti-angiogenic therapy using small-molecule compounds such as tyrosine kinase inhibitors that block VEGFR activation and downstream signalling. This review provides a rational basis towards reconciling VEGF and VEGFR structure and function in developing new therapeutics for a diverse range of ailments.


Subject(s)
Disease , Receptors, Vascular Endothelial Growth Factor/chemistry , Receptors, Vascular Endothelial Growth Factor/metabolism , Animals , Humans , Ligands , Protein Domains , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
8.
Sci Rep ; 10(1): 14413, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32879419

ABSTRACT

Vascular endothelial growth factor A (VEGF-A) and its binding to VEGFRs is an important angiogenesis regulator, especially the earliest-known isoform, VEGF-A165a. Yet several additional splice variants play prominent roles in regulating angiogenesis in health and in vascular disease, including VEGF-A121 and an anti-angiogenic variant, VEGF-A165b. Few studies have attempted to distinguish these forms from their angiogenic counterparts, experimentally. Previous studies of VEGF-A:VEGFR binding have measured binding kinetics for VEGFA165 and VEGF-A121, but binding kinetics of the other two pro- and all anti-angiogenic splice variants are not known. We measured the binding kinetics for VEGF-A165, -A165b, and -A121 with VEGFR1 and VEGF-R2 using surface plasmon resonance. We validated our methods by reproducing the known affinities between VEGF-A165a:VEGFR1 and VEGF-A165a:VEGFR2, 1.0 pM and 10 pM respectively, and validated the known affinity VEGF-A121:VEGFR2 as KD = 0.66 nM. We found that VEGF-A121 also binds VEGFR1 with an affinity KD = 3.7 nM. We further demonstrated that the anti-angiogenic variant, VEGF-A165b selectively prefers VEGFR2 binding at an affinity = 0.67 pM while binding VEGFR1 with a weaker affinity-KD = 1.4 nM. These results suggest that the - A165b anti-angiogenic variant would preferentially bind VEGFR2. These discoveries offer a new paradigm for understanding VEGF-A, while further stressing the need to take care in differentiating the splice variants in all future VEGF-A studies.


Subject(s)
Alternative Splicing , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Humans , Neovascularization, Physiologic/genetics , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Vascular Endothelial Growth Factor/chemistry , Receptors, Vascular Endothelial Growth Factor/genetics , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/genetics
9.
Med Hypotheses ; 144: 110009, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32758869

ABSTRACT

The outbreak of Novel Coronavirus 2019 (COVID-19) represents a global threat to the public healthcare. The viral spike (S) glycoprotein is the key molecule for viral entry through interaction with angiotensin converting enzyme 2 (ACE2) receptor molecules present on the cell membranes. Moreover, it has been established that COVID-19 interacts and infects brain cells in humans via ACE2. Therefore in the light of these known facts we hypothesized that viral S protein molecule may bind to the other overexpressed receptor molecules in glioma cells and may play some role in glioma tumorogenesis. Thus we leverage docking analysis (HEX and Z-DOCK) between viral S protein and epidermal growth factor receptors (EGFR), vascular endothelial growth factor receptors (VEGFR) and hepatocyte growth factor receptors (HGFR/c-MET) to investigate the oncogenic potential of COVID-19. Our findings suggested higher affinity of Viral S protein towards EGFR and VEGFR. Although, the data presented is preliminary and need to be validated further via molecular dynamics studies, however it paves platform to instigate further investigations on this aspect considering the aftermath of COVID-19 pandemic in oncogenic perspective.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Brain Neoplasms/etiology , COVID-19/complications , Glioma/etiology , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/genetics , Brain/virology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Transformation, Neoplastic , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Glioma/genetics , Glioma/metabolism , Humans , Models, Molecular , Molecular Docking Simulation , Neoplasm Proteins/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Conformation , Proto-Oncogene Proteins c-met/chemistry , Receptors, Vascular Endothelial Growth Factor/chemistry , SARS-CoV-2/metabolism , Up-Regulation
10.
J Pharm Biomed Anal ; 185: 113233, 2020 Jun 05.
Article in English | MEDLINE | ID: mdl-32169790

ABSTRACT

Ziv-aflibercept (ziv-AFL) is a complex fusion protein which is widely used in hospitals for the treatment of colorectal metastatic cancer. Charge variants are critical attributes for assessing post-transitional modifications (PMTs) that have to be controlled during the development and manufacture of these proteins and until their administration to patients. Cation exchange (CEX) chromatography is a charge-sensitive analytical method that is well suited for analysing charge variants in proteins. The aim of this paper is to analyse the charge variants of ziv-AFL in the medicine (Zaltrap®) when fresh and when degraded. Two CEX chromatographic methods were compared for this purpose. The former was an adaptation of the method used in the first published study in which charge variants were analysed via pH gradient elution using volatile, low ionic strength buffers with direct coupling to high-resolution Orbitrap mass spectrometry. The second method was developed and optimized during our research using the salt-mediated pH gradient mode and classical non-volatile, high ionic strength buffers which were incompatible with direct coupling with mass detection. Fresh and controlled degraded samples of ziv-AFL were used to evaluate the capacity of both CEX chromatographic strategies for detecting charge variants in ziv-AFL. In the controlled degradation study the samples of the medicine were subjected to three stress factors: temperature of 60 °C for three hours, freeze/thaw process -two cycles-, and exposure to light for twelve hours. The CEX chromatographic method with non-volatile salts in the mobile phase enabled better detection of charge variants degraded ziv-AFL samples than the method using volatile salts with lower ionic strength. In addition, the complexity of the mass spectra data generated made it impossible to identify the multicharge variant species of ziv-AFL. Although charge variants were not separated in ziv-AFL fresh sample, our results indicate that the method with non-volatile salts in the mobile phase could be used to characterize and track changes in the charge variant UV chromatographic profile of ziv-AFL in fresh and degraded samples, even though it cannot be coupled to a mass detector and there is therefore no information about mass. The increase of basic protein degraded compounds were the most important degradation pattern detected in ziv-AFL (Zaltrap®).


Subject(s)
Chemistry, Pharmaceutical/methods , Receptors, Vascular Endothelial Growth Factor/analysis , Recombinant Fusion Proteins/analysis , Cations , Chromatography, Ion Exchange/methods , Feasibility Studies , Humans , Hydrogen-Ion Concentration , Mass Spectrometry/methods , Osmolar Concentration , Proof of Concept Study , Proteolysis , Receptors, Vascular Endothelial Growth Factor/chemistry , Recombinant Fusion Proteins/chemistry
11.
Sci Rep ; 10(1): 2675, 2020 02 14.
Article in English | MEDLINE | ID: mdl-32060315

ABSTRACT

Aflibercept (AFL) is an Fc fusion protein used in the treatment of colorectal cancers and different ophthalmological diseases. There are two medicines in which AFL is the active substance: Zaltrap and Eylea, referred as ziv-AFL and AFL respectively. No proper accelerated degradation studies were published on either AFL or ziv-AFL. These studies are essential during research, development and manufacturing stages. Here, we characterized ziv-AFL and submitted it to different stress conditions: light, 60 °C, freeze-thaw cycles, changes in pH, high hypertonic solution and strong denaturing conditions. We used an array of techniques to detect aggregation (SE-HPLC/DAD and DLS), changes in secondary structure (Far-UV circular dichroism), changes in conformation or tertiary structure (Intrinsic tryptophan fluorescence) and alterations in functionality (ELISA). Results indicate that aggregation is common degradation pathway. Two different types of aggregates were detected: dimers and high molecular weight aggregates attributed to ß-amyloid-like structures. Secondary structure was maintained in most of the stress tests, while conformation was altered by almost all the tests except for the freeze-thaw cycles. Functionality, evaluated by its immunochemical reaction with VEGF, was found to be stable but with decrease when exposed to light and with likely partial inactivation of the drug when pH was altered.


Subject(s)
Angiogenesis Inhibitors/chemistry , Drug Stability , Receptors, Vascular Endothelial Growth Factor/chemistry , Recombinant Fusion Proteins/chemistry , Angiogenesis Inhibitors/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Freezing/adverse effects , Hot Temperature/adverse effects , Humans , Hydrogen-Ion Concentration , Hypertonic Solutions/adverse effects , Intravitreal Injections , Light/adverse effects , Receptors, Vascular Endothelial Growth Factor/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Vascular Endothelial Growth Factor A/genetics
12.
Spectrochim Acta A Mol Biomol Spectrosc ; 220: 117102, 2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31146207

ABSTRACT

Quantum chemical analyses were performed over enrofloxacin and boron complexes. The most stable isomer of enrofloxacin was examined at M062X/6-31+G(d) level in gas phase. Structural and spectral characterizations of enrofloxacin and its complexes were performed at same level of theory. MEP maps of studied compound were calculated via ESP charges analyses. Some quantum chemical descriptors (QCDs) were calculated to determine the non-linear optical (NLO) and biological reactivity of studied molecules. Furthermore, molecular docking calculations between boron complexes and a protein (ID: 2ITN and 2ITV) were done. ADME analyses were done in the determination of the best drug candidate. As a result, complex (3) was found as the best in the NLO applications and it was found that complex (1) and (3) have similar biological reactivity in lung cancer treatment.


Subject(s)
Boron Compounds/chemistry , Enrofloxacin/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacokinetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Boron Compounds/pharmacokinetics , Cisplatin/chemistry , Cisplatin/pharmacology , Cytochrome P-450 Enzyme Inhibitors/chemistry , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Enrofloxacin/pharmacokinetics , Hydrogen Bonding , Isomerism , Magnetic Resonance Spectroscopy , Molecular Conformation , Molecular Docking Simulation , Quantum Theory , Receptors, Vascular Endothelial Growth Factor/chemistry , Receptors, Vascular Endothelial Growth Factor/metabolism , Spectrophotometry, Infrared , Thermodynamics , Urea/chemistry
13.
J Ocul Pharmacol Ther ; 35(5): 278-282, 2019 06.
Article in English | MEDLINE | ID: mdl-30916605

ABSTRACT

Purpose: Aflibercept (Eylea™, Regeneron) is supplied in single-use glass vials along with 1 cc polycarbonate syringes. We sought to determine if storage of aflibercept for sustained periods within these syringes would result in loss of antivascular endothelial growth factor (anti-VEGF) activity. Methods: Aflibercept samples were drawn from commercially available glass vials into manufacturer-supplied 1-mL syringes and stored at 4°C. Anti-VEGF activity was assessed using enzyme-linked immunosorbent assays at the following storage durations: 0, 4, 9, 14, and 28 days. Frozen samples stored at -20°C for 28 and 56 days were also assayed. Also, a subset of aflibercept samples was stored and then diluted to 1:10 and progressively smaller concentrations and the assays repeated. Aggregation of aflibercept was tested using a dynamic light scattering assay. Results: There were no statistical differences in anti-VEGF activity among aflibercept samples of 1:1 or 1:10 dilution stored at either 4°C or -20°C at any of the storage intervals (P > 0.05). We also observed persistence of robust anti-VEGF activity for up to 14 days when diluted poststorage to 1:16,000, a concentration that would be expected after >7 vitreous half-lives within the eye (estimated at >50 days). No evidence of drug aggregation in specimens stored for 14 days was observed. Conclusions: Our findings support feasibility of prefilling and storage of aflibercept within manufacturer-supplied polycarbonate syringes for as long as 14 days before use under pharmacy-based sterile conditions, facilitating greater safety and efficiency in many clinics delivering anti-VEGF therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Polycarboxylate Cement/chemistry , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factors/antagonists & inhibitors , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/chemistry , Drug Storage , Humans , Intravitreal Injections , Receptors, Vascular Endothelial Growth Factor/chemistry , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/chemistry
14.
Wiley Interdiscip Rev Syst Biol Med ; 11(2): e1437, 2019 03.
Article in English | MEDLINE | ID: mdl-30255986

ABSTRACT

Receptor tyrosine kinases (RTKs) are cell membrane proteins that provide cells with the ability to sense proteins in their environments. Many RTKs are essential to development and organ growth. Derangement of RTKs-by mutation or by overexpression-is central to several developmental and adult disorders including cancer, short stature, and vascular pathologies. The mechanism of action of RTKs is complex and is regulated by contextual components, including the existence of multiple competing ligands and receptors in many families, the intracellular location of the RTK, the dynamic and cell-specific coexpression of other RTKs, and the commonality of downstream signaling pathways. This means that both the state of the cell and the microenvironment outside the cell play a role, which makes sense given the pivotal location of RTKs as the nexus linking the extracellular milieu to intracellular signaling and modification of cell behavior. In this review, we describe these different contextual components through the lens of systems biology, in which both computational modeling and experimental "omics" approaches have been used to better understand RTK networks. The complexity of these networks is such that using these systems biology approaches is necessary to get a handle on the mechanisms of pathology and the design of therapeutics targeting RTKs. In particular, we describe in detail three concrete examples (involving ErbB3, VEGFR2, and AXL) that illustrate how systems approaches can reveal key mechanistic and therapeutic insights. This article is categorized under: Biological Mechanisms > Cell Signaling Models of Systems Properties and Processes > Mechanistic Models Translational, Genomic, and Systems Medicine > Therapeutic Methods.


Subject(s)
Receptor Protein-Tyrosine Kinases/metabolism , ErbB Receptors/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Ligands , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Vascular Endothelial Growth Factor/chemistry , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction , Tumor Microenvironment , Vascular Diseases/metabolism , Vascular Diseases/pathology , Axl Receptor Tyrosine Kinase
15.
J Agric Food Chem ; 67(4): 1127-1137, 2019 Jan 30.
Article in English | MEDLINE | ID: mdl-30525561

ABSTRACT

Resveratrol is a polyphenol commonly found in plants and food health products, such as grape and red wine, and was identified for its binding to vascular endothelial growth factor (VEGF) by using HerboChips screening. The binding, therefore, resulted in alterations of VEGF binding to its receptor and revealed the roles of VEGF in angiogenesis. Several lines of evidence gave support to the inhibitory activities of resveratrol in VEGF-triggered angiogenesis. In human umbilical vein endothelial cells (HUVECs), compared with a VEGF-induced group, resveratrol, at a high concentration, suppressed VEGF-mediated endothelial cell proliferation, cell migration, cell invasion, and tube formation by 80 ± 9.01%, 140 ± 3.78%, 110 ± 7.51%, and 120 ± 10.26%, respectively. Moreover, resveratrol inhibited the subintestinal vessel formation in zebrafish embryo. In signaling cascades, application of resveratrol in HUVECs reduced the VEGF-triggered VEGF receptor 2 phosphorylation and c-Jun N-terminal kinase phosphorylation. Moreover, the VEGF-mediated phosphorylations of endothelial nitric oxide synthase, protein kinase B, and extracellular signal-regulated kinase were obviously decreased by (3 ± 0.37)-, (2 ± 0.27)- and (6 ± 0.23)-fold, respectively, in the presence of resveratrol at high concentration. Parallelly, the VEGF-induced reactive oxygen species formation was significantly decreased by 50 ± 7.88% to 120 ± 14.82% under resveratrol treatment. Thus, our results provided support to the antiangiogenic roles of resveratrol, as well as its related signaling mechanisms, in attenuating the VEGF-mediated responses. The present results supported possible development of resveratrol, which should be considered as a therapeutic agent in terms of prevention and clinical treatment of diseases related to angiogenesis.


Subject(s)
Angiogenesis Inhibitors/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic/drug effects , Receptors, Vascular Endothelial Growth Factor/metabolism , Resveratrol/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Phosphorylation/drug effects , Receptors, Vascular Endothelial Growth Factor/chemistry , Receptors, Vascular Endothelial Growth Factor/genetics , Resveratrol/chemistry , Resveratrol/pharmacology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/chemistry , Zebrafish
16.
Pharm Res ; 35(12): 245, 2018 Oct 29.
Article in English | MEDLINE | ID: mdl-30374744

ABSTRACT

Topical delivery of therapeutics to the posterior segment of the eye remains the "holy grail" of ocular drug delivery. As an example, anti-vascular endothelial growth factor biologics, such as ranibizumab, aflibercept, and bevacizumab, are delivered by intravitreal injection to treat neovascular age-related macular degeneration and, although these drugs have revolutionized treatment of the disease, less invasive alternatives to intravitreal injection are desired. Multiple reports in the literature have demonstrated topical delivery of both small and large molecules to the back of the eye in small animal models. Despite this progress, successful translation to larger species, and ultimately humans, has yet to be demonstrated. Selection of animal models with relevant ocular anatomy and physiology, along with appropriate experimental design, is critical to enable more relevant feasibility assessments and increased probability of successful translation.


Subject(s)
Drug Delivery Systems/methods , Eye/metabolism , Administration, Topical , Animals , Bevacizumab/administration & dosage , Bevacizumab/chemistry , Chemistry, Pharmaceutical , Humans , Intravitreal Injections , Macular Degeneration/drug therapy , Ranibizumab/administration & dosage , Ranibizumab/chemistry , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Receptors, Vascular Endothelial Growth Factor/chemistry , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Translational Research, Biomedical , Vascular Endothelial Growth Factor A/metabolism
17.
Open Biol ; 8(9)2018 09 19.
Article in English | MEDLINE | ID: mdl-30232099

ABSTRACT

Cells interact with the extracellular environment by means of receptor molecules on their surface. Receptors can bind different ligands, leading to the formation of receptor-ligand complexes. For a subset of receptors, called receptor tyrosine kinases, binding to ligand enables sequential phosphorylation of intra-cellular residues, which initiates a signalling cascade that regulates cellular function and fate. Most mathematical modelling approaches employed to analyse receptor signalling are deterministic, especially when studying scenarios of high ligand concentration or large receptor numbers. There exist, however, biological scenarios where low copy numbers of ligands and/or receptors need to be considered, or where signalling by a few bound receptor-ligand complexes is enough to initiate a cellular response. Under these conditions stochastic approaches are appropriate, and in fact, different attempts have been made in the literature to measure the timescales of receptor signalling initiation in receptor-ligand systems. However, these approaches have made use of numerical simulations or approximations, such as moment-closure techniques. In this paper, we study, from an analytical perspective, the stochastic times to reach a given signalling threshold for two receptor-ligand models. We identify this time as an extinction time for a conveniently defined auxiliary absorbing continuous time Markov process, since receptor-ligand association/dissociation events can be analysed in terms of quasi-birth-and-death processes. We implement algorithmic techniques to compute the different order moments of this time, as well as the steady-state probability distribution of the system. A novel feature of the approach introduced here is that it allows one to quantify the role played by each kinetic rate in the timescales of signal initiation, and in the steady-state probability distribution of the system. Finally, we illustrate our approach by carrying out numerical studies for the vascular endothelial growth factor and one of its receptors, the vascular endothelial growth factor receptor of human endothelial cells.


Subject(s)
Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Algorithms , Humans , Kinetics , Ligands , Markov Chains , Phosphorylation , Receptors, Vascular Endothelial Growth Factor/chemistry , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction , Stochastic Processes , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/metabolism
18.
Chemistry ; 24(44): 11461-11466, 2018 Aug 06.
Article in English | MEDLINE | ID: mdl-29799174

ABSTRACT

QK peptide is a vascular endothelial growth factor (VEGF)-mimetic molecule with significant proangiogenic activity. In particular, QK is able to bind and activate VEGF receptors (VEGFRs) to stimulate a functional response in endothelial cells. To characterize the peptide bioactivity and its molecular recognition properties, a detailed picture of the interaction between peptide QK and VEGF receptors is reported. By combining NMR spectroscopy studies in solution on the purified receptor and in the presence of intact endothelial cells, a molecular description of the binding interaction between peptide QK and VEGFR2 in the cellular context is obtained. These results reveal useful insights into the peptide biological mechanism, which opens the way to further optimization of this class of VEGF-mimicking peptides.


Subject(s)
Biomimetic Materials/chemistry , Peptides/chemistry , Receptors, Vascular Endothelial Growth Factor/chemistry , Vascular Endothelial Growth Factor A/chemistry , Endothelial Cells , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Binding , Protein Conformation
19.
Pharm Res ; 35(4): 78, 2018 Feb 28.
Article in English | MEDLINE | ID: mdl-29492680

ABSTRACT

PURPOSE: Studies were conducted to investigate dilute solutions of the monoclonal antibody (mAb) bevacizumab, mAb fragment ranibizumab and fusion protein aflibercept, develop common procedures for formulation of low concentration mAbs and identify a stabilizing formulation for anti-VEGF mAbs for use in in vitro permeation studies. METHODS: Excipient substitutions were screened. The most stabilizing formulation was chosen. Standard dilutions of bevacizumab, ranibizumab and aflibercept were prepared in PBS, manufacturer's formulation, and the new formulation. Analysis was by SE-HPLC and ELISA. Stability, disaggregation and pre-exposure tests were studied. RESULTS: When Avastin, Lucentis and Eylea are diluted in PBS or manufacturer's formulation, there is a 40-50% loss of monomer concentration and drug activity. A formulation containing 0.3% NaCl, 7.5% trehalose, 10 mM arginine and 0.04% Tween 80 at a pH of 6.78 stabilized the mAbs and minimized the drug loss. The formulation also disaggregates mAb aggregation while preserving the activity. Degassing the formulation increases recovery. CONCLUSIONS: We developed a novel formulation that significantly stabilizes mAbs under unfavorable conditions such as low concentration or body temperature. The formulation allows for tissue permeation experimentation. The formulation also exhibits a disaggregating effect on mAbs, which can be applied to the manufacture/packaging of mAbs and bioassay reagents.


Subject(s)
Angiogenesis Inhibitors/chemistry , Biological Products/chemistry , Drug Compounding/methods , Excipients/chemistry , Bevacizumab/chemistry , Biological Assay/methods , Drug Stability , Protein Aggregates , Ranibizumab/chemistry , Receptors, Vascular Endothelial Growth Factor/chemistry , Recombinant Fusion Proteins/chemistry , Solutions , Temperature
20.
Sci Rep ; 8(1): 2101, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29391560

ABSTRACT

Macular edema due to neovascular age-related macular degeneration, diabetes or retinal vein occlusion can cause central vision loss. Intravitreal treatment with antibody-based biopharmaceutical compounds designed to neutralize vascular endothelial growth factor (VEGF) has proven to be an efficient strategy to ameliorate macular edema and restore visual acuity. At the same time, the use of anti-VEGF drugs places an economic burden on the health care system; the drugs are expensive, and repeated injections are usually required to maintain the therapeutic effect. Thus, there is an unmet need for more cost-effective procedures. We here describe how the most recently approved anti-VEGF drug, aflibercept, can be compounded into prefilled sterile syringes and stored for up to 4 weeks without compromising its quality, stability or functional properties, including VEGF and neonatal Fc receptor (FcRn) binding. The novel compounding method for repackaging of aflibercept in sterile plastic syringes can greatly reduce both cost and time spent per patient in the injection room.


Subject(s)
Drug Compounding/standards , Immunoglobulin Fc Fragments/metabolism , Receptors, Vascular Endothelial Growth Factor/chemistry , Receptors, Vascular Endothelial Growth Factor/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Syringes/standards , Vascular Endothelial Growth Factor A/metabolism , Drug Stability , Humans , Immunoglobulin Fc Fragments/chemistry , Vascular Endothelial Growth Factor A/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...