Therapeutic effect of apatinib-loaded nanoparticles on diabetes-induced retinal vascular leakage

Ji Hoon Jeong, Hong Khanh Nguyen, Jung Eun Lee, Wonhee Suh, Ji Hoon Jeong, Hong Khanh Nguyen, Jung Eun Lee, Wonhee Suh

Abstract

Apatinib, a novel and selective inhibitor of vascular endothelial growth factor (VEGF) receptor 2, has been demonstrated recently to exhibit anticancer efficacy by inhibiting the VEGF signaling pathway. Given the importance of VEGF in retinal vascular leakage, the present study was designed to investigate whether apatinib-loaded polymeric nanoparticles inhibit VEGF-mediated retinal vascular hyperpermeability and block diabetes-induced retinal vascular leakage. For the delivery of water-insoluble apatinib, the drug was encapsulated in nanoparticles composed of human serum albumin (HSA)-conjugated polyethylene glycol (PEG). In vitro paracellular permeability and transendothelial electric resistance assays showed that apatinib-loaded HSA-PEG (Apa-HSA-PEG) nanoparticles significantly inhibited VEGF-induced endothelial hyperpermeability in human retinal microvascular endothelial cells. In addition, they substantially reduced the VEGF-induced junctional loss and internalization of vascular endothelial-cadherin, a major component of endothelial junction complexes. In vivo intravitreal injection of Apa-HSA-PEG nanoparticles in mice blocked VEGF-induced retinal vascular leakage. These in vitro and in vivo data indicated that Apa-HSA-PEG nanoparticles efficiently blocked VEGF-induced breakdown of the blood-retinal barrier. In vivo experiments with streptozotocin-induced diabetic mice showed that an intravitreal injection of Apa-HSA-PEG nanoparticles substantially inhibited diabetes-induced retinal vascular leakage. These results demonstrated, for the first time, that apatinib-loaded nanoparticles may be a promising therapeutic agent for the prevention and treatment of diabetes-induced retinal vascular disorders.

Keywords: permeability; retinal vascular endothelial cells; vascular endothelial growth factor.

Figures

Figure 1
Figure 1
Preparation and characterization of Apa-HSA-PEG nanoparticles. Notes: (A) Schematic diagram of the preparation of Apa-HSA-PEG nanoparticles. (B) TEM images and (C) particle size distribution of Apa-HSA-PEG nanoparticles. All scale bars =500 µm. Abbreviations: Apa-HSA-PEG, apatinib-loaded human serum albumin-conjugated polyethylene glycol; TEM, transmission electron microscope; NHS, N-hydroxysuccinimide; RT, room temperature; THF, tetrahydrofuran.
Figure 2
Figure 2
Apa-HSA-PEG nanoparticles block VEGF-induced hyperpermeability in HRMECs. Notes: (A) Cytotoxicity of Apa-HSA-PEG nanoparticles and apatinib was evaluated using the Cell Counting Kit-8 assay (n=6). HRMECs were incubated with apatinib (1 and 5 µM) or Apa-HSA-PEG nanoparticles (960 and 4.8 µg corresponding to 1 and 5 µM apatinib, respectively) for 1–2 days. (B and C) Endothelial permeability was evaluated by measuring the (B) passage of FITC-dextran and (C) TEER in a HRMEC monolayer. HRMECs pretreated with apatinib (1 µM) or Apa-HSA-PEG nanoparticles (6.1 µg corresponding to 1 µM apatinib), and untreated cells were stimulated with rhVEGF (50 ng/mL). (B) FITC-dextran permeability is expressed as the fold change ± SEM with respect to the PBS control (*P<0.05 vs PBS control, #P<0.05 vs VEGF, n=6). (C) In the TEER experiment, specified reagents were added to the upper chamber at time zero, and serial changes in electrical resistance were measured 30 and 60 minutes later. Electrical resistance is expressed as the fold change ± SEM relative to the PBS control at each time point (*P<0.05 vs PBS control, #P<0.05 vs VEGF, n=6). Abbreviations: Apa-HSA-PEG, apatinib-loaded human serum albumin-conjugated polyethylene glycol; VEGF, vascular endothelial growth factor; HRMECs, human retinal microvascular endothelial cells; FITC, fluorescein isothiocyanate; TEER, transendothelial electrical resistance; rhVEGF, recombinant human VEGF; SEM, standard error of the mean; PBS, phosphate-buffered saline; NS, nonsignificant.
Figure 3
Figure 3
Apa-HSA-PEG nanoparticles prevent VEGF-induced internalization of VE-cadherin. Notes: (A) Representative immunofluorescent images of VE-cadherin. (B) Quantification of internalized VE-cadherin in HRMECs (means ± SEM, *P<0.05 vs PBS, #P<0.05 vs VEGF, n=4). Cells pretreated with Apa-HSA-PEG nanoparticles (6.1 µg corresponding to 1 µM apatinib) or PBS were stimulated with rhVEGF (50 ng/mL). Arrowheads in the no wash image indicate the disappearance of VE-cadherin (green) at endothelial junctions that were stained positively for anti-ZO1 IgGs (red). Arrowheads in the acid wash image indicate internalized VE-cadherin (green) in endosomes that were stained positively for EEA1 (red). Nuclei are shown in blue (DAPI). Scale bars =25 µm. Abbreviations: Apa-HSA-PEG, apatinib-loaded human serum albumin-conjugated polyethylene glycol; EEA1, early endosome antigen 1; VEGF, vascular endothelial growth factor; VE, vascular endothelial; HRMECs, human retinal microvascular endothelial cells; SEM, standard error of the mean; PBS, phosphate-buffered saline; rhVEGF, recombinant human VEGF; DAPI, 4′,6-diamidino-2-phenylindole; VE-cad, VE-cadherin.
Figure 4
Figure 4
Apa-HSA-PEG nanoparticles block VEGF-induced retinal vascular leakage in vivo. Notes: (A) Representative images of FITC-dextran-perfused retinal whole mounts. (B) Quantitative analysis of extravasated EB dye in the retinal tissues after intravitreal injection of rhVEGF (100 ng) and/or Apa-HSA-PEG nanoparticles (580 ng). Vascular leakage of EB dye in treated eyes was normalized relative to that in each contralateral control eye (means ± SEM, *P<0.05 vs PBS control, #P<0.05 vs VEGF, n=5). Scale bar =200 µm. Abbreviations: Apa-HSA-PEG, apatinib-loaded human serum albumin-conjugated polyethylene glycol; VEGF, vascular endothelial growth factor; FITC, fluorescein isothiocyanate; EB, Evans Blue; rhVEGF, recombinant human VEGF; SEM, standard error of the mean; PBS, phosphate-buffered saline.
Figure 5
Figure 5
Intravitreal injection of Apa-HSA-PEG nanoparticles inhibits diabetes-induced retinal vascular leakage in STZ-induced diabetic mice. Notes: (A) STZ-induced diabetic mice received an intravitreal injection of Apa-HSA-PEG nanoparticles (61 ng for the low-dose group [Low], 610 ng for the high-dose group [High]). An equal volume of PBS was injected into the contralateral eye as a control. One day after injection, retinal vascular leakage of EB dye was measured and expressed relative to that in each contralateral control eye (means ± SEM, **P<0.01 vs contralateral PBS control, n=8). (B) Blood glucose and body weights of mice used in this experiment were measured before (Nondiabetes) and 2 weeks after (Diabetes) the initial STZ injection (*P<0.05 vs Nondiabetes, n=8). Abbreviations: Apa-HSA-PEG, apatinib-loaded human serum albumin-conjugated polyethylene glycol; STZ, streptozotocin; PBS, phosphate-buffered saline; EB, Evans Blue; SEM, standard error of the mean.

References

    1. Klaassen I, Van Noorden CJ, Schlingemann RO. Molecular basis of the inner blood-retinal barrier and its breakdown in diabetic macular edema and other pathological conditions. Prog Retin Eye Res. 2013;34:19–48.
    1. Aiello LP, Avery RL, Arrigg PG, et al. Vascular endothelial growth factor in ocular fluid of patients with diabetic retinopathy and other retinal disorders. N Engl J Med. 1994;331:1480–1487.
    1. Weis SM, Cheresh DA. Pathophysiological consequences of VEGF-induced vascular permeability. Nature. 2005;437:497–504.
    1. Dejana E, Tournier-Lasserve E, Weinstein BM. The control of vascular integrity by endothelial cell junctions: molecular basis and pathological implications. Dev Cell. 2009;16:209–221.
    1. Ho AC, Scott IU, Kim SJ, et al. Anti-vascular endothelial growth factor pharmacotherapy for diabetic macular edema: a report by the American Academy of Ophthalmology. Ophthalmology. 2012;119:2179–2188.
    1. Roviello G, Ravelli A, Polom K, et al. Apatinib: a novel receptor tyrosine kinase inhibitor for the treatment of gastric cancer. Cancer Lett. 2016;372:187–191.
    1. Tian S, Quan H, Xie C, et al. YN968D1 is a novel and selective inhibitor of vascular endothelial growth factor receptor-2 tyrosine kinase with potent activity in vitro and in vivo. Cancer Sci. 2011;102:1374–1380.
    1. Li J, Qin S, Xu J, et al. Apatinib for chemotherapy-refractory advanced metastatic gastric cancer: results from a randomized, placebo-controlled, parallel-arm, phase II trial. J Clin Oncol. 2013;31:3219–3225.
    1. Li J, Zhao X, Chen L, et al. Safety and pharmacokinetics of novel selective vascular endothelial growth factor receptor-2 inhibitor YN968D1 in patients with advanced malignancies. BMC Cancer. 2010;10:529.
    1. Kim JY, Choi JS, Song SH, et al. Stem cell factor is a potent endothelial permeability factor. Arterioscler Thromb Vasc Biol. 2014;34:1459–1467.
    1. Kompella UB, Amrite AC, Pacha Ravi R, Durazo SA. Nanomedicines for back of the eye drug delivery, gene delivery, and imaging. Prog Retin Eye Res. 2013;36:172–198.
    1. Diebold Y, Calonge M. Applications of nanoparticles in ophthalmology. Prog Retin Eye Res. 2010;29:596–609.
    1. Xu Q, Kambhampati SP, Kannan RM. Nanotechnology approaches for ocular drug delivery. Middle East Afr J Ophthalmol. 2013;20:26–37.
    1. Kragh-Hansen U, Chuang VT, Otagiri M. Practical aspects of the ligand-binding and enzymatic properties of human serum albumin. Biol Pharm Bull. 2002;25:695–704.
    1. Sparreboom A, Scripture CD, Trieu V, et al. Comparative preclinical and clinical pharmacokinetics of a cremophor-free, nanoparticle albumin-bound paclitaxel (ABI-007) and paclitaxel formulated in Cremophor (Taxol) Clin Cancer Res. 2005;11:4136–4143.

Source: PubMed

3
Subscribe