1
|
Dervenis N, Dervenis P, Agorogiannis E. Neovascular age-related macular degeneration: disease pathogenesis and current state of molecular biomarkers predicting treatment response-a scoping review. BMJ Open Ophthalmol 2024; 9:e001516. [PMID: 38341189 PMCID: PMC10862285 DOI: 10.1136/bmjophth-2023-001516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Age-related macular degeneration is a major cause of blindness, and the development of anti-vascular endothelial growth factor (VEGF) intravitreal treatments has revolutionised the management of the disease. At the same time, new challenges and unmet needs arose due to the limitations of the current therapeutic options. Neovascularisation development during the course of the disease has a complex pathogenetic mechanism, and several biomarkers and their association with treatment outcomes have been investigated. We reviewed the relevant literature about neovascularisation development and biomarkers related to response to treatment. Improving our knowledge on the field can improve patient outcomes and offer personalised care.
Collapse
Affiliation(s)
- Nikolaos Dervenis
- Aristotle University of Thessaloniki, Thessaloniki, Greece
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | | | | |
Collapse
|
2
|
Salas A, Badia A, Fontrodona L, Zapata M, García-Arumí J, Duarri A. Neovascular Progression and Retinal Dysfunction in the Laser-Induced Choroidal Neovascularization Mouse Model. Biomedicines 2023; 11:2445. [PMID: 37760886 PMCID: PMC10525599 DOI: 10.3390/biomedicines11092445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/22/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
The mouse model of laser-induced choroidal neovascularization (LI-CNV) has been widely used to study neovascular age-related macular degeneration; however, it still lacks a comprehensive characterization. Here, CNV was induced in the eyes of 12-week-old C57BL/6J male mice by argon laser irradiation. We studied the CNV lesion progression of an LI-CNV mouse cohort by using multimodal imaging (color fundus, optical coherence tomography (OCT), and fluorescence angiography, focal electroretinography features for 14 days, and related cytokines, angiogenic factors, and reactive gliosis for 5 days. CNV lesions involving the rupture of the Bruch's membrane were confirmed using funduscopy and OCT after laser photocoagulation. During the initial stage, from the CNV induction until day 7, CNV lesions presented leakage observed by using fluorescence angiography and a typical hyperreflective area with cell infiltration, subretinal leakage, and degeneration of photoreceptors observed through OCT. This correlated with decreased retinal responses to light. Moreover, inflammatory and angiogenic markers were reduced to basal levels in the first 5 days of CNV progression. In contrast, reactive gliosis and the VEGF expression in retinal sections were sustained, with infiltration of endothelial cells in the subretinal space. In the second stage, between days 7 and 14 post-induction, we observed stabilization of the CNV lesions, a hyperfluorescent area corresponding to the formation of fibrosis, and a partial rescue of retinal function. These findings suggest that the LI-CNV lesion development goes through an acute phase during the first seven days following induction, and then the CNV lesion stabilizes. According to these results, this model is suitable for screening anti-inflammatory and anti-angiogenic drugs in the early stages of LI-CNV. At the same time, it is more convenient for screening anti-fibrotic compounds in the later stages.
Collapse
Affiliation(s)
- Anna Salas
- Ophthalmology Research Group, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain
| | - Anna Badia
- Ophthalmology Research Group, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain
| | - Laura Fontrodona
- Ophthalmology Research Group, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain
| | - Miguel Zapata
- Ophthalmology Research Group, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain
- Department of Ophthalmology, Vall d’Hebron Hospital Universitari, 08035 Barcelona, Spain
| | - José García-Arumí
- Ophthalmology Research Group, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain
- Department of Ophthalmology, Vall d’Hebron Hospital Universitari, 08035 Barcelona, Spain
| | - Anna Duarri
- Ophthalmology Research Group, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain
| |
Collapse
|
3
|
Lima e Silva R, Mirando AC, Tzeng SY, Green JJ, Popel AS, Pandey NB, Campochiaro PA. Anti-angiogenic collagen IV-derived peptide target engagement with α vβ 3 and α 5β 1 in ocular neovascularization models. iScience 2023; 26:106078. [PMID: 36844452 PMCID: PMC9947312 DOI: 10.1016/j.isci.2023.106078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/22/2022] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
AXT107, a collagen-derived peptide that binds integrins αvβ3 and α5β1 with high affinity, suppresses vascular endothelial growth factor (VEGF) signaling, promotes angiopoietin 2-induced Tie2 activation, and suppresses neovascularization (NV) and vascular leakage. Immunohistochemical staining for αvβ3 and α5β1 was markedly increased in NV compared with normal retinal vessels. After intravitreous injection of AXT107, there was no staining with an anti-AXT107 antibody on normal vessels but robust staining of NV that co-localized with αvβ3 and α5β1. Likewise, after intravitreous injection, fluorescein amidite-labeled AXT107 co-localized with αvβ3 and α5β1 on NV but not normal vessels. AXT107 also co-localized with αv and α5 at cell-cell junctions of human umbilical vein endothelial cells (HUVECs). AXT107-integrin binding was demonstrated by ex vivo cross-linking/pull-down experiments. These data support the hypothesis that AXT107 therapeutic activity is mediated through binding αvβ3 and α5β1 which are markedly upregulated on endothelial cells in NV providing selective targeting of diseased vessels which has therapeutic and safety benefits.
Collapse
Affiliation(s)
- Raquel Lima e Silva
- Department of Ophthalmology and The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adam C. Mirando
- AsclepiX Therapeutics, Inc., Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephany Y. Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jordan J. Green
- Department of Ophthalmology and The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Niranjan B. Pandey
- AsclepiX Therapeutics, Inc., Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter A. Campochiaro
- Department of Ophthalmology and The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Therapeutic Effects of Fenofibrate Nano-Emulsion Eye Drops on Retinal Vascular Leakage and Neovascularization. BIOLOGY 2021; 10:biology10121328. [PMID: 34943243 PMCID: PMC8698460 DOI: 10.3390/biology10121328] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 01/08/2023]
Abstract
Macular edema caused by retinal vascular leakage and ocular neovascularization are the leading causes of severe vision loss in diabetic retinopathy (DR) and age-related macular degeneration (AMD) patients. Oral administration of fenofibrate, a PPARα agonist, has shown therapeutic effects on macular edema and retinal neovascularization in diabetic patients. To improve the drug delivery to the retina and its efficacy, we have developed a nano-emulsion-based fenofibrate eye drop formulation that delivered significantly higher amounts of the drug to the retina compared to the systemic administration, as measured by liquid chromatography-mass spectrometer (LC-MS). The fenofibrate eye drop decreased leukocytes adherent to retinal vasculature and attenuated overexpression of multiple inflammatory factors in the retina of very low-density lipoprotein receptor knockout (Vldlr-/-) mice, a model manifesting AMD phenotypes, and streptozotocin-induced diabetic rats. The fenofibrate eye drop also reduced retinal vascular leakage in these models. The laser-induced choroidal neovascularization was also alleviated by the fenofibrate eye drop. There were no detectable ocular toxicities associated with the fenofibrate eye drop treatment. These findings suggest that fenofibrate can be delivered efficiently to the retina through topical administration of the nano-emulsion eye drop, which has therapeutic potential for macular edema and neovascularization.
Collapse
|
5
|
Inhibitory effect of nintedanib on VEGF secretion in retinal pigment epithelial cells induced by exposure to a necrotic cell lysate. PLoS One 2019; 14:e0218632. [PMID: 31386668 PMCID: PMC6684070 DOI: 10.1371/journal.pone.0218632] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/05/2019] [Indexed: 11/19/2022] Open
Abstract
Necrosis is a form of cell death that results in rupture of the plasma membrane and the release of cellular contents, and it can give rise to sterile inflammation in the retina and other tissues. The secretion of vascular endothelial growth factor (VEGF) by retinal pigment epithelial (RPE) cells contributes to retinal homeostasis as well as to pathological angiogenesis. We have now examined the effect of a necrotic cell lysate prepared from human RPE cells (NLR) on the release of VEGF by healthy RPE cells. We found that NLR markedly increased the release of VEGF from RPE cells and that this effect was attenuated by nintedanib, a multiple receptor tyrosine kinase inhibitor, whereas it was unaffected by inhibitors of NF-κB signaling or of caspase-1. NLR also induced the phosphorylation of extracellular signal-regulated kinase (Erk) and signal transducer and activator of transcription 3 (Stat3) in a manner sensitive to inhibition by nintedanib, although inhibitors of Erk and Stat3 signaling pathways did not affect NLR-induced VEGF secretion. In addition, nintedanib attenuated the development of choroidal neovascularization in mice. Our results have thus shown that a necrotic lysate of RPE cells induced VEGF secretion from healthy RPE cells and that this effect was mediated by receptor tyrosine kinase signaling. They therefore suggest that VEGF secretion by healthy RPE cells is a potential therapeutic target for retinal diseases associated with sterile inflammation and pathological angiogenesis.
Collapse
|
6
|
Brain signalling systems: A target for treating type I diabetes mellitus. Brain Res Bull 2019; 152:191-201. [PMID: 31325597 DOI: 10.1016/j.brainresbull.2019.07.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/08/2019] [Accepted: 07/15/2019] [Indexed: 01/26/2023]
Abstract
From early to later stages of Type I Diabetes Mellitus (TIDM), signalling molecules including brain indolamines and protein kinases are altered significantly, and that has been implicated in the Metabolic Disorders (MD) as well as impairment of retinal, renal, neuronal and cardiovascular systems. Considerable attention has been focused to the effects of diabetes on these signalling systems. However, the exact pathophysiological mechanisms of these signals are not completely understood in TIDM, but it is likely that hyperglycemia, acidosis, and insulin resistance play significant roles. Insulin maintains normal glycemic levels and it acts by binding to its receptor, so that it activates the receptor's tyrosine kinase activity, resulting in phosphorylation of several substrates. Those substrates provide binding/interaction sites for signalling molecules, including serine/threonine kinases and indolamines. For more than two decades, our research has been focused on the mechanisms of protein kinases, CaM Kinase and Serotonin transporter mediated alterations of indolamines in TIDM. In this review, we have also discussed how discrete areas of brain respond to insulin or some of the pharmacological agents that triggers or restores these signalling molecules, and it may be useful for the treatment of specific region wise changes/disorders of diabetic brain.
Collapse
|
7
|
Luisi J, Liu W, Zhang W, Motamedi M. En-Face Optical Coherence Tomography Angiography for Longitudinal Monitoring of Retinal Injury. APPLIED SCIENCES (BASEL, SWITZERLAND) 2019; 9:2617. [PMID: 34671487 PMCID: PMC8525491 DOI: 10.3390/app9132617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
A customized Optical Coherence Tomography Angiography (OCTA) algorithm and Orthogonal OCT (en-face and B-Scans) were used for longitudinal assessment of retina murine vascular and tissue remodeling comparing photoreceptor ablation and laser-induced Choroidal Neovascularization (CNV). In the mouse model, we utilized a combined OCTA/OCT technique to image and quantify morphological and vascular features of laser lesions over time. This approach enabled us to monitor and correlate the dynamics of retina vascular and tissue remodeling as evidenced by swelling, edema, and scarring. From the OCT B-Scans, three stages of inflammatory progression were identified: the early response occurring within hours to day 3, the transition phase from 3-7 days, and the late stage of 7-21 days entering either the resolving phase or chronic phase, respectively. For the case of CNV, en-face OCTA revealed a transient non-perfusion of inner retina capillaries, specifically Deep Vascular Plexus (DVP), which corresponded to growth in lesions of a height of 200 μm or greater. Non-perfusion first occurred at 24 hours, persisted during edema and CNV formation days 7-14. In contrast, the acute inflammation induced photoreceptor damage, but no detectable alterations to the microvasculature were observed. We demonstrated that the en-face OCTA system is capable of visualizing capillary networks (~5 μm) and the corresponding tissue remodeling and growth dynamics allowing for separating acute injury from CNV. For the first time, by using OCTA we observed the presence of the 5-10 μm capillary non-perfusion present in DVP as part of CNV formation and the associated wound healing in the retina.
Collapse
Affiliation(s)
- Jonathan Luisi
- Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Wei Liu
- Ophthalmology and Visual Science, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Wenbo Zhang
- Ophthalmology and Visual Science, University of Texas Medical Branch, Galveston, TX 77555, USA
- Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Massoud Motamedi
- Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, TX 77555, USA
- Ophthalmology and Visual Science, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
8
|
Mg–Zn–Mn alloy extract induces the angiogenesis of human umbilical vein endothelial cells via FGF/FGFR signaling pathway. Biochem Biophys Res Commun 2019; 514:618-624. [DOI: 10.1016/j.bbrc.2019.04.198] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 04/29/2019] [Indexed: 11/21/2022]
|
9
|
Manley PW, Caravatti G, Furet P, Roesel J, Tran P, Wagner T, Wartmann M. Comparison of the Kinase Profile of Midostaurin (Rydapt) with That of Its Predominant Metabolites and the Potential Relevance of Some Newly Identified Targets to Leukemia Therapy. Biochemistry 2018; 57:5576-5590. [PMID: 30148617 DOI: 10.1021/acs.biochem.8b00727] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The multitargeted protein kinase inhibitor midostaurin is approved for the treatment of both newly diagnosed FLT3-mutated acute myeloid leukemia (AML) and KIT-driven advanced systemic mastocytosis. AML is a heterogeneous malignancy, and investigational drugs targeting FLT3 have shown disparate effects in patients with FLT3-mutated AML, probably as a result of their inhibiting different targets and pathways at the administered doses. However, the efficacy and side effects of drugs do not just reflect the biochemical and pharmacodynamic properties of the parent compound but are often comprised of complex cooperative effects between the properties of the parent and active metabolites. Following chronic dosing, two midostaurin metabolites attain steady-state plasma trough levels greater than that of the parent drug. In this study, we characterized these metabolites and determined their profiles as kinase inhibitors using radiometric transphosphorylation assays. Like midostaurin, the metabolites potently inhibit mutant forms of FLT3 and KIT and several additional kinases that either are directly involved in the deregulated signaling pathways or have been implicated as playing a role in AML via stromal support, such as IGF1R, LYN, PDPK1, RET, SYK, TRKA, and VEGFR2. Consequently, a complex interplay between the kinase activities of midostaurin and its metabolites is likely to contribute to the efficacy of midostaurin in AML and helps to engender the distinctive effects of the drug compared to those of other FLT3 inhibitors in this malignancy.
Collapse
Affiliation(s)
- Paul W Manley
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research , Novartis International AG , CH-4002 Basel , Switzerland
| | - Giorgio Caravatti
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research , Novartis International AG , CH-4002 Basel , Switzerland
| | - Pascal Furet
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research , Novartis International AG , CH-4002 Basel , Switzerland
| | - Johannes Roesel
- Oncology Disease Area, Novartis Institutes for Biomedical Research , Novartis International AG , CH-4002 Basel , Switzerland
| | - Phi Tran
- Department of Drug Metabolism and Pharmacokinetics , Novartis Institutes for Biomedical Research , East Hanover , New Jersey 07936 , United States
| | - Trixie Wagner
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research , Novartis International AG , CH-4002 Basel , Switzerland
| | - Markus Wartmann
- Oncology Disease Area, Novartis Institutes for Biomedical Research , Novartis International AG , CH-4002 Basel , Switzerland
| |
Collapse
|
10
|
Ragauskas S, Kielczewski E, Vance J, Kaja S, Kalesnykas G. In Vivo Multimodal Imaging and Analysis of Mouse Laser-Induced Choroidal Neovascularization Model. J Vis Exp 2018. [PMID: 29443029 DOI: 10.3791/56173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Laser-induced choroidal neovascularization (CNV) is a well-established model to mimic the wet form of age-related macular degeneration (AMD). In this protocol, we aim to guide the reader not simply through the technical considerations of generating laser-induced lesions to trigger neovascular processes, but rather focus on the powerful information that can be obtained from multimodal longitudinal in vivo imaging throughout the follow-up period. The laser-induced mouse CNV model was generated by a diode laser administration. Multimodal in vivo imaging techniques were used to monitor CNV induction, progression and regression. First, spectral domain optical coherence tomography (SD-OCT) was performed immediately after the lasering to verify a break of Bruch's membrane. Subsequent in vivo imaging using fluorescein angiography (FA) confirmed successful damage of Bruch's membrane from serial images acquired at the choroidal level. Longitudinal follow-up of CNV proliferation and regression on days 5, 10, and 14 after the lasering was performed using both SD-OCT and FA. Simple and reliable grading of leaky CNV leasions from FA images is presented. Automated segmentation for measurement of total retinal thickness, combined with manual caliber application for measurement of retinal thickness at CNV sites, allow unbiased evaluation of the presence of edema. Finally, histological verification of CNV is performed using isolectin GS-IB4 staining on choroidal flatmounts. The staining is thresholded, and the isolectin-positive area is calculated with ImageJ. This protocol is especially useful in therapeutics studies requiring high-throughput-like screening of CNV pathology as it allows fast, multimodal, and reliable classification of CNV pathology and retinal edema. In addition, high resolution SD-OCT enables the recording of other pathological hallmarks, such as the accumulation of subretinal or intraretinal fluid. However, this method does not provide a possibility to automate CNV volume analysis from SD-OCT images, which has to be performed manually.
Collapse
Affiliation(s)
| | | | | | - Simon Kaja
- Experimentica Ltd.; Department of Ophthalmology, Stritch School of Medicine, Loyola University Chicago
| | | |
Collapse
|
11
|
Yan YJ, Bao LL, Zhang LJ, Bian J, Hu TS, Zheng MZ, Chen DY, Yu XH, Chen ZL. Inhibition of Laser-Induced Choroidal Neovascularization by Hematoporphyrin Dimethylether-Mediated Photodynamic Therapy in Rats. Biol Pharm Bull 2017; 40:2088-2095. [PMID: 29199233 DOI: 10.1248/bpb.b17-00319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study aimed to investigate the effect of hematoporphyrin dimethylether (HDME)-mediated photodynamic therapy for laser-induced choroidal neovascularization (CNV) in adult Brown Norway rats. HDME was administered via tail vein at 14 d after the laser photocoagulation, and the rats received irradiance with a laser light at 570 nm at 15 min after injection. CNV was evaluated by fundus photography, fundus fluorescein angiography, optical coherence tomography, and hematoxylin and eosin staining. We found that CNV was occurred at 7 d after photocoagulation and reaching peak activity at 14 d after photocoagulation. There is a significant reduction in the total area of the fluorescein leakage and the number of strong fluorescein leakage spots on 7 d after HDME-mediated photodynamic therapy (PDT). The results suggest that HDME-mediated PDT inhibits laser-induced CNV in rats, representing a promising therapy for wet age-related macular degeneration.
Collapse
Affiliation(s)
- Yi-Jia Yan
- Department of Pharmaceutical Science & Technology, College of Chemistry and Biology, Donghua University.,Department of Pharmacology, Shanghai Xianhui Pharmaceutical Co., Ltd
| | | | - Li-Jun Zhang
- Department of Pharmaceutical Science & Technology, College of Chemistry and Biology, Donghua University
| | | | - Tai-Shan Hu
- Department of Pharmacology, Shanghai Xianhui Pharmaceutical Co., Ltd
| | - Mei-Zhen Zheng
- Department of Pharmacology, Shanghai Xianhui Pharmaceutical Co., Ltd
| | - Dan-Ye Chen
- Department of Pharmaceutical Science & Technology, College of Chemistry and Biology, Donghua University
| | - Xin-Hai Yu
- Department of Pharmaceutical Science & Technology, College of Chemistry and Biology, Donghua University
| | - Zhi-Long Chen
- Department of Pharmaceutical Science & Technology, College of Chemistry and Biology, Donghua University
| |
Collapse
|
12
|
Tahiri H, Omri S, Yang C, Duhamel F, Samarani S, Ahmad A, Vezina M, Bussières M, Vaucher E, Sapieha P, Hickson G, Hammamji K, Lapointe R, Rodier F, Tremblay S, Royal I, Cailhier JF, Chemtob S, Hardy P. Lymphocytic Microparticles Modulate Angiogenic Properties of Macrophages in Laser-induced Choroidal Neovascularization. Sci Rep 2016; 6:37391. [PMID: 27874077 PMCID: PMC5118818 DOI: 10.1038/srep37391] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 10/27/2016] [Indexed: 11/13/2022] Open
Abstract
Pathological choroidal neovascularization (CNV) is the common cause of vision loss in patients with age-related macular degeneration (AMD). Macrophages possess potential angiogenic function in CNV. We have demonstrated that human T lymphocyte-derived microparticles (LMPs) exert a potent antiangiogenic effect in several pathological neovascularization models. In this study, we investigated the alteration of proangiogenic properties of macrophages by LMPs treatment in vitro and in vivo models. LMPs regulated the expression of several angiogenesis-related factors in macrophages and consequently stimulated their antiangiogenic effects evidenced by the suppression of the proliferation of human retinal endothelial cells in co-culture experiments. The involvement of CD36 receptor in LMPs uptake by macrophages was demonstrated by in vitro assays and by immunostaining of choroidal flat mounts. In addition, ex vivo experiments showed that CD36 mediates the antiangiogenic effect of LMPs in murine and human choroidal explants. Furthermore, intravitreal injection of LMPs in the mouse model of laser-induced CNV significantly suppressed CNV in CD36 dependent manner. The results of this study suggested an ability of LMPs to alter the gene expression pattern of angiogenesis-related factors in macrophages, which provide important information for a new therapeutic approach for efficiently interfering with both vascular and extravascular components of CNV.
Collapse
Affiliation(s)
- Houda Tahiri
- Department of Pharmacology, Université de Montréal, Montréal, QC, Canada.,Research Center CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Samy Omri
- Research Center Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC, Canada
| | - Chun Yang
- Research Center CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - François Duhamel
- Department of Pharmacology, Université de Montréal, Montréal, QC, Canada
| | - Suzanne Samarani
- Departments of Microbiology and Immunology, Université de Montréal, Montréal, QC, Canada
| | - Ali Ahmad
- Departments of Microbiology and Immunology, Université de Montréal, Montréal, QC, Canada
| | - Mark Vezina
- Charles River Laboratories, Senneville, Montreal, QC, Canada
| | | | - Elvire Vaucher
- School of Optometry, Université de Montréal, Montréal, QC, Canada
| | - Przemyslaw Sapieha
- Research Center Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC, Canada.,Department of Ophthalmology, Université de Montréal, Montréal, QC, Canada
| | - Gilles Hickson
- Department of Pathology and Cell Biology, Université de Montréal, Montréal, QC, Canada
| | - Karim Hammamji
- Department of Ophthalmology, Université de Montréal, Montréal, QC, Canada
| | - Réjean Lapointe
- Institut du Cancer de Montréal, CRCHUM-Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Francis Rodier
- Institut du Cancer de Montréal, CRCHUM-Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Department of Medicine, Université de Montréal, Montréal, QC, Canada.,Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montréal, QC, Canada
| | - Sophie Tremblay
- University of British Columbia, Vancouver, BC, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada
| | - Isabelle Royal
- Institut du Cancer de Montréal, CRCHUM-Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Jean-François Cailhier
- Institut du Cancer de Montréal, CRCHUM-Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Sylvain Chemtob
- Department of Pharmacology, Université de Montréal, Montréal, QC, Canada.,Research Center CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada.,Research Center Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC, Canada.,Department of Ophthalmology, Université de Montréal, Montréal, QC, Canada.,Department of Pediatrics, Université de Montréal, Montréal, QC, Canada
| | - Pierre Hardy
- Department of Pharmacology, Université de Montréal, Montréal, QC, Canada.,Research Center CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada.,Department of Pediatrics, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
13
|
Chang KC, Shieh B, Petrash JM. Aldose reductase mediates retinal microglia activation. Biochem Biophys Res Commun 2016; 473:565-71. [PMID: 27033597 DOI: 10.1016/j.bbrc.2016.03.122] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 03/26/2016] [Indexed: 01/10/2023]
Abstract
Retinal microglia (RMG) are one of the major immune cells in charge of surveillance of inflammatory responses in the eye. In the absence of an inflammatory stimulus, RMG reside predominately in the ganglion layer and inner or outer plexiform layers. However, under stress RMG become activated and migrate into the inner nuclear layer (INL) or outer nuclear layer (ONL). Activated RMG in cell culture secrete pro-inflammatory cytokines in a manner sensitive to downregulation by aldose reductase inhibitors. In this study, we utilized CX3CR1(GFP) mice carrying AR mutant alleles to evaluate the role of AR on RMG activation and migration in vivo. When tested on an AR(WT) background, IP injection of LPS induced RMG activation and migration into the INL and ONL. However, this phenomenon was largely prevented by AR inhibitors or in AR null mice, or was exacerbated in transgenic mice that over-express AR. LPS-induced increases in ocular levels of TNF-α and CX3CL-1 in WT mice were substantially lower in AR null mice or were reduced by AR inhibitor treatment. These studies demonstrate that AR expression in RMG may contribute to the proinflammatory phenotypes common to various eye diseases such as uveitis and diabetic retinopathy.
Collapse
Affiliation(s)
- Kun-Che Chang
- Department of Ophthalmology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Biehuoy Shieh
- Department of Ophthalmology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - J Mark Petrash
- Department of Ophthalmology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
14
|
Wigg JP, Zhang H, Yang D. A Quantitative and Standardized Method for the Evaluation of Choroidal Neovascularization Using MICRON III Fluorescein Angiograms in Rats. PLoS One 2015; 10:e0128418. [PMID: 26024231 PMCID: PMC4449229 DOI: 10.1371/journal.pone.0128418] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 04/27/2015] [Indexed: 01/18/2023] Open
Abstract
Introduction In-vivo imaging of choroidal neovascularization (CNV) has been increasingly recognized as a valuable tool in the investigation of age-related macular degeneration (AMD) in both clinical and basic research applications. Arguably the most widely utilised model replicating AMD is laser generated CNV by rupture of Bruch’s membrane in rodents. Heretofore CNV evaluation via in-vivo imaging techniques has been hamstrung by a lack of appropriate rodent fundus camera and a non-standardised analysis method. The aim of this study was to establish a simple, quantifiable method of fluorescein fundus angiogram (FFA) image analysis for CNV lesions. Methods Laser was applied to 32 Brown Norway Rats; FFA images were taken using a rodent specific fundus camera (Micron III, Phoenix Laboratories) over 3 weeks and compared to conventional ex-vivo CNV assessment. FFA images acquired with fluorescein administered by intraperitoneal injection and intravenous injection were compared and shown to greatly influence lesion properties. Utilising commonly used software packages, FFA images were assessed for CNV and chorioretinal burns lesion area by manually outlining the maximum border of each lesion and normalising against the optic nerve head. Net fluorescence above background and derived value of area corrected lesion intensity were calculated. Results CNV lesions of rats treated with anti-VEGF antibody were significantly smaller in normalised lesion area (p<0.001) and fluorescent intensity (p<0.001) than the PBS treated control two weeks post laser. The calculated area corrected lesion intensity was significantly smaller (p<0.001) in anti-VEGF treated animals at 2 and 3 weeks post laser. The results obtained using FFA correlated with, and were confirmed by conventional lesion area measurements from isolectin stained choroidal flatmounts, where lesions of anti-VEGF treated rats were significantly smaller at 2 weeks (p = 0.049) and 3 weeks (p<0.001) post laser. Conclusion The presented method of in-vivo FFA quantification of CNV, including acquisition variable corrections, using the Micron III system and common use software establishes a reliable method for detecting and quantifying CNV enabling longitudinal studies and represents an important alternative to conventional CNV quantification methods.
Collapse
Affiliation(s)
- Jonathan P. Wigg
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Hong Zhang
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
- Eye Hospital, Harbin Medical University, Nangang District, Harbin, Heilongjiang Province, China
- * E-mail:
| | - Dong Yang
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| |
Collapse
|
15
|
Li L, Eter N, Heiduschka P. The microglia in healthy and diseased retina. Exp Eye Res 2015; 136:116-30. [PMID: 25952657 DOI: 10.1016/j.exer.2015.04.020] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 04/28/2015] [Accepted: 04/29/2015] [Indexed: 12/25/2022]
Abstract
The microglia are the immune cells of the central nervous system and, also the retina. They fulfil several tasks of surveillance in the healthy retina. In case of an injury or disease, microglia become activated and tries to repair the damage. However, in a lot of cases it does not work, and microglia deteriorate the situation by releasing toxic and pro-inflammatory compounds. Moreover, they further promote degenerative processes by attacking and phagocytosing damaged neurones and photoreceptors that otherwise would possibly have the chance to survive. Such deleterious action of the microglia has been observed in degeneration of retinal ganglion cells and photoreceptors, and it takes place in hereditary diseases, infections as well as in case of traumatic or light injuries. Therefore, a number of attempts has been undertaken so far to inhibit the microglia, with varying success. The task remains to study behaviour of the microglia and their interaction with other retinal cell populations in more detail with respect to released factors and expressed receptors including the time points of the corresponding events. The goal has to be to find a better balance between helpful and detrimental actions of the microglia.
Collapse
Affiliation(s)
- Lu Li
- University of Münster Medical School, Department of Ophthalmology, Domagkstr. 15, D-48149 Münster, Germany
| | - Nicole Eter
- University of Münster Medical School, Department of Ophthalmology, Domagkstr. 15, D-48149 Münster, Germany
| | - Peter Heiduschka
- University of Münster Medical School, Department of Ophthalmology, Domagkstr. 15, D-48149 Münster, Germany.
| |
Collapse
|
16
|
Abstract
This is a summary of current and emerging pharmacologic therapies utilized in the treatment of diabetic retinopathy (DR). Current therapies, such as ranibizumab, bevacizumab, triamcinolone acetonide, and fluocinolone acetonide, inhibit angiogenesis and inflammation and may be used alone or in combination with laser treatment. Emerging therapies aim to reduce oxidative stress or inhibit other signal transduction pathways, including the protein kinase C cascade and aldose reductase pathway. Future therapies may target other molecules crucial to the pathogenesis of DR, including hepatocyte growth factors and matrix metalloproteinase 9. Finally, the emergence of novel mechanisms of medication delivery may also be on the horizon.
Collapse
Affiliation(s)
- Vaidehi S. Dedania
- Department of Ophthalmology, Albany Medical Center, Lions Eye Institute, Albany, NY 12208, USA
| | - Sophie J. Bakri
- Department of Ophthalmology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
17
|
Wang L, Yang Z, Yu Y, Cui C, Guan H, Chen H. Blockage of tissue factor ameliorates the lesion of laser-induced choroidal neovascularization in mice. Exp Eye Res 2014; 127:117-23. [DOI: 10.1016/j.exer.2014.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/16/2014] [Accepted: 07/08/2014] [Indexed: 01/16/2023]
|
18
|
Tosi GM, Marigliani D, Romeo N, Toti P. Disease pathways in proliferative vitreoretinopathy: an ongoing challenge. J Cell Physiol 2014; 229:1577-83. [PMID: 24604697 DOI: 10.1002/jcp.24606] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 01/16/2014] [Indexed: 11/08/2022]
Abstract
Despite remarkable advances in vitreoretinal surgery, proliferative vitreoretinopathy (PVR) remains a common cause of severe visual loss or blindness. One of the critical reasons for PVR-induced blindness is tractional retinal detachment due to the formation of contractile preretinal fibrous membranes. This membrane formation is characterized by the proliferation and migration of cells and the excessive synthesis and deposition of extracellular matrix proteins. Herein we present the disease pathways of PVR, reviewing the role of both systemic and intraocular cells as well as molecular mediators. A chronological sequence of events leading to PVR is also hypothesized. Better understanding of the pathogenesis of PVR is needed in order to improve disease management. Efforts should be oriented towards greater cooperation between basic researchers and clinicians, aimed at matching the different clinical scenarios with the biological markers of the disease.
Collapse
Affiliation(s)
- Gian Marco Tosi
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | | | | | | |
Collapse
|
19
|
Sweigard JH, Yanai R, Gaissert P, Saint-Geniez M, Kataoka K, Thanos A, Stahl GL, Lambris JD, Connor KM. The alternative complement pathway regulates pathological angiogenesis in the retina. FASEB J 2014; 28:3171-82. [PMID: 24668752 DOI: 10.1096/fj.14-251041] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A defining feature in proliferative retinopathies is the formation of pathological neovessels. In these diseases, the balance between neovessel formation and regression determines blindness, making the modulation of neovessel growth highly desirable. The role of the immune system in these retinopathies is of increasing interest, but it is not completely understood. We investigated the role of the alternative complement pathway during the formation and resolution of aberrant neovascularization. We used alternative complement pathway-deficient (Fb(-/-)) mice and age- and strain-matched control mice to assess neovessel development and regression in an oxygen-induced retinopathy (OIR) mouse model. In the control mice, we found increased transcription of Fb after OIR treatment. In the Fb(-/-) mice, we prepared retinal flatmounts and identified an increased number of neovessels, peaking at postnatal day 17 (P17; P=0.001). Subjecting human umbilical vein endothelial cells (HUVECs) to low oxygen, mimicking a characteristic of neovessels, decreased the expression of the complement inhibitor Cd55. Finally, using laser capture microdissection (LCM) to isolate the neovessels after OIR, we found decreased expression of Cd55 (P=0.005). Together, our data implicate the alternative complement pathway in facilitating neovessel clearance by down-regulating the complement inhibitor Cd55 specifically on neovessels, allowing for their targeted removal while leaving the established vasculature intact.-Sweigard, J. H., Yanai, R., Gaissert, P., Saint-Geniez, M., Kataoka, K., Thanos, A., Stahl, G. L., Lambris, J. D., Connor, K. M. The alternative complement pathway regulates pathological angiogenesis in the retina.
Collapse
Affiliation(s)
| | - Ryoji Yanai
- Angiogenesis Laboratory, Department of Ophthalmology, and
| | | | | | - Keiko Kataoka
- Angiogenesis Laboratory, Department of Ophthalmology, and
| | | | - Gregory L Stahl
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; and
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kip M Connor
- Angiogenesis Laboratory, Department of Ophthalmology, and
| |
Collapse
|
20
|
Microvascular complications and diabetic retinopathy: recent advances and future implications. Future Med Chem 2013; 5:301-14. [PMID: 23464520 DOI: 10.4155/fmc.12.206] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Retinal microvascular alterations have been observed during diabetic retinopathy (DR) due to the retinal susceptibility towards subtle pathological alterations. Therefore, retinal microvascular pathology is essential to understand the nature of retinal degenerations during DR. In this review, the role of retinal microvasculature complications during progression of DR, along with recent efforts to normalize such alterations for better therapeutic outcome, will be underlined. In addition, current therapeutics and future directions for advancement of standard treatment for DR patients will be discussed.
Collapse
|
21
|
Menne J, Shushakova N, Bartels J, Kiyan Y, Laudeley R, Haller H, Park JK, Meier M. Dual inhibition of classical protein kinase C-α and protein kinase C-β isoforms protects against experimental murine diabetic nephropathy. Diabetes 2013; 62:1167-74. [PMID: 23434935 PMCID: PMC3609593 DOI: 10.2337/db12-0534] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Activation of protein kinase C (PKC) has been implicated in the pathogenesis of diabetic nephropathy with proteinuria and peritubular extracellular matrix production. We have previously shown that the PKC isoforms α and β mediate different cellular effects. PKC-β contributes to hyperglycemia-induced renal matrix production, whereby PKC-α is involved in the development of albuminuria. We further tested this hypothesis by deletion of both isoforms and used a PKC inhibitor. We analyzed the phenotype of nondiabetic and streptozotocin (STZ)-induced diabetic homozygous PKC-α/β double-knockout mice (PKC-α/β(-/-)). After 8 weeks of diabetes mellitus, the high-glucose-induced renal and glomerular hypertrophy as well as transforming growth factor-β1) and extracellular matrix production were diminished in the PKC-α/β(-/-) mice compared with wild-type controls. Urinary albumin/creatinine ratio also was significantly reduced, however, it was not completely abolished in diabetic PKC-α/β(-/-) mice. Treatment with CGP41252, which inhibits PKC-α and PKC-β, is able to prevent the development of albuminuria and to reduce existing albuminuria in type 1 (STZ model) or type 2 (db/db model) diabetic mice. These results support our hypothesis that PKC-α and PKC-β contribute to the pathogenesis of diabetic nephropathy, and that dual inhibition of the classical PKC isoforms is a suitable therapeutic strategy in the prevention and treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Jan Menne
- Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
- Phenos GmbH, Hannover, Germany
- Corresponding authors: Jan Menne, , and Hermann Haller,
| | - Nelli Shushakova
- Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
- Phenos GmbH, Hannover, Germany
| | - Janina Bartels
- Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Yulia Kiyan
- Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Robert Laudeley
- Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
- Corresponding authors: Jan Menne, , and Hermann Haller,
| | - Joon-Keun Park
- Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Matthias Meier
- Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| |
Collapse
|
22
|
|
23
|
Pennesi ME, Neuringer M, Courtney RJ. Animal models of age related macular degeneration. Mol Aspects Med 2012; 33:487-509. [PMID: 22705444 DOI: 10.1016/j.mam.2012.06.003] [Citation(s) in RCA: 289] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Age related macular degeneration (AMD) is the leading cause of vision loss of those over the age of 65 in the industrialized world. The prevalence and need to develop effective treatments for AMD has lead to the development of multiple animal models. AMD is a complex and heterogeneous disease that involves the interaction of both genetic and environmental factors with the unique anatomy of the human macula. Models in mice, rats, rabbits, pigs and non-human primates have recreated many of the histological features of AMD and provided much insight into the underlying pathological mechanisms of this disease. In spite of the large number of models developed, no one model yet recapitulates all of the features of human AMD. However, these models have helped reveal the roles of chronic oxidative damage, inflammation and immune dysregulation, and lipid metabolism in the development of AMD. Models for induced choroidal neovascularization have served as the backbone for testing new therapies. This article will review the diversity of animal models that exist for AMD as well as their strengths and limitations.
Collapse
Affiliation(s)
- Mark E Pennesi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| | | | | |
Collapse
|
24
|
Tan H, Mu G, Zhu W, Liu J, Wang F. Down-regulation of vascular endothelial growth factor and up-regulation of pigment epithelium derived factor make low molecular weight heparin-endostatin and polyethylene glycol-endostatin potential candidates for anti-angiogenesis drug. Biol Pharm Bull 2011; 34:545-50. [PMID: 21467643 DOI: 10.1248/bpb.34.545] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim was to study the effects and action mechanism of endostatin (ES), low molecular weight heparin-endostatin (LMWH-ES) and polyethylene glycol-endostatin (PEG-ES) on endothelial cell proliferation, choroidal neovascularization and zebrafish angiogenesis. Three-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazoliumbromide was used to study the effects of ES and its derivatives on endothelial cell proliferation in vitro. Choroidal neovascularization model was used to evaluate the effects of ES and its derivatives on choroidal neovascularization in vivo. Western blotting was employed to study the effects of ES and its derivatives on the expression of vascular endothelial growth factor (VEGF) and pigment epithelium derived factor (PEDF) in chorioid tissues. Zebrafish model was also used to study the anti-angiogenesis activities of ES and its derivatives. The results showed that ES and its derivatives could significantly inhibit endothelial cell proliferation in vitro (p<0.05), suppress choroidal neovascularization by down-regulating expression of VEGF and up-regulating expression of PEDF in chorioid tissues, and restrain angiogenesis in zebrafish. ES showed better activity in inhibiting endothelial cell proliferation in vitro (p<0.05), but LMWH-ES and PEG-ES showed higher activity in inhibiting choroidal neovascularization in vivo (p<0.05) and angiogenesis in zebrafish (p<0.05). These results indicate that LMWH-endostatin and PEG-endostatin are potential candidates for anti-angiogenesis drug.
Collapse
Affiliation(s)
- Haining Tan
- Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, China
| | | | | | | | | |
Collapse
|
25
|
Abstract
Diseases complicated by abnormal growth of vessels or excessive leakage are the most prevalent cause of moderate or severe vision loss in developed countries. Recent progress unraveling the molecular pathogenesis of several of these disease processes has led to new drug therapies that have provided major benefits to patients. However, those treatments often require frequent intraocular injections, and despite monthly injections, some patients have a suboptimal response. Gene transfer of antiangiogenic proteins is an alternative approach that has the potential to provide long-term suppression of neovascularization (NV) and/or excessive vascular leakage in the eye. Studies in animal models of ocular NV have demonstrated impressive results with a number of transgenes, and a clinical trial in patients with advanced neovascular age-related macular degeneration has provided proof-of-concept. Two ongoing clinical trials, one using an adeno-associated viral (AAV) vector to express a vascular endothelial growth factor-binding protein and another using a lentiviral vector to express endostatin and angiostatin, will provide valuable information that should help to inform future trials and provide a foundation on which to build.
Collapse
Affiliation(s)
- P A Campochiaro
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21287-9277, USA.
| |
Collapse
|
26
|
Zhang L, Shen X, Lu Q, Zhou Q, Gu J, Gan R, Zhang H, Sun X, Xie B. A potential therapeutic strategy for inhibition of ocular neovascularization with a new endogenous protein: rhEDI-8t. Graefes Arch Clin Exp Ophthalmol 2011; 250:731-9. [PMID: 21881847 DOI: 10.1007/s00417-011-1765-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 06/15/2011] [Accepted: 07/28/2011] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Endogenous angiogenesis inhibitors act as natural negative feedback in the focal area during the neovascularization process, and have less interference on physiological angiogenesis, and thus fewer negative side-effects. These inhibitors are potential candidates to combine with or substitutes for current popular anti-angiogenesis treatments to have synergistic effect. In this study, the effects of recombinant endothelial growth inhibitor protein (rhEDI-8t), a novel endogenous protein originated from collagen VIII, was investigated on ocular neovascularization (NV). Endostatin, a well-identified endogenous angiogenesis inhibitor, was compared in parallel and served as a positive control. METHODS The inhibitory effect of rhEDI-8t on vascular endothelial cells was evaluated by a human umbilical vascular endothelial cells (HUVEC) proliferation test and a bovine aortic endothelial cells (BAEC) migration experiment. The effect of rhEDI-8t on ocular NV was further investigated in mice with choroidal neovascularization (choroidal NV) induced by laser, ischemic retinopathy and transgenic mice with expression of VEGF in photoreceptors (rho/VEGF) respectively. RESULTS RhEDI-8t inhibited the growth of HUVECs and migration of BAECs stimulated by basic fibroblast growth factor (bFGF). Mice intravitreally treated with rhEDI-8t showed a significant reduction of choroidal NV, retinal NV and subretinal NV. CONCLUSION Endogenous angiogenesis inhibitor rhEDI-8t showed a potent anti-angiogenesis effect in both in vitro and in vivo experiments. It contributed to the suppression of ocular NV. The study suggested that rhEDI-8t could be a subsidiary potent therapeutic medicine in addition to anti-VEGF therapy in future clinical anti-angiogenesis treatment.
Collapse
Affiliation(s)
- Ling Zhang
- The Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197, Ruijin Er Road, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
DeNiro M, Al-Mohanna FH, Al-Mohanna FA. Inhibition of reactive gliosis prevents neovascular growth in the mouse model of oxygen-induced retinopathy. PLoS One 2011; 6:e22244. [PMID: 21779402 PMCID: PMC3136522 DOI: 10.1371/journal.pone.0022244] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 06/20/2011] [Indexed: 11/19/2022] Open
Abstract
Retinal neovascularization (NV) is a major cause of blindness in ischemic retinopathies. Previous investigations have indicated that ischemia upregulates GFAP and PDGF-B expression. GFAP overexpression is a hallmark of reactive gliosis (RG), which is the major pathophysiological feature of retinal damage. In addition, PDGF-B has been implicated in proliferative retinopathies. It was the aim of this study to gain insights on the possible pharmacological interventions to modulate PDGF-B and GFAP expression, and its influence on RG and NV. We used an array of assays to evaluate the effects of YC-1, a small molecule inhibitor of HIF-1 and a novel NO-independent activator of soluble guanylyl cyclase (sGC), on RG and NV, in vivo and in vitro. When compared to the DMSO-treated retinas, dual-intravitreal injections of YC-1, in vivo: (1) suppressed the development and elongation of neovascular sprouts in the retinas of the oxygen-induced retinopathy (OIR) mouse model; and (2) reduced ischemia-induced overexpression of GFAP and PDGF-B at the message (by 64.14±0.5% and 70.27±0.04%) and the protein levels (by 65.52±0.02% and 57.59±0.01%), respectively. In addition, at 100 µM, YC-1 treatment downregulated the hypoxia-induced overexpression of GFAP and PDGF-B at the message level in rMC-1 cells (by 71.42±0.02% and 75±0.03%), and R28 cells (by 58.62±0.02% and 50.00±0.02%), respectively; whereas, the protein levels of GFAP and PDGF-B were reduced (by 78.57±0.02% and 77.55±0.01%) in rMC-1 cells, and (by 81.44±0.02% and 79.16±0.01%) in R28 cells, respectively. We demonstrate that YC-1 reversed RG during ischemic retinopathy via impairing the expression of GFAP and PDGF-B in glial cells. This is the first investigation that delves into the reversal of RG during ischemic retinal vasculopathies. In addition, the study reveals that YC-1 may exert promising therapeutic effects in the treatment of retinal and neuronal pathologies.
Collapse
Affiliation(s)
- Michael DeNiro
- Research Department, King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia.
| | | | | |
Collapse
|
28
|
Gu L, Chen H, Tuo J, Gao X, Chen L. Inhibition of experimental choroidal neovascularization in mice by anti-VEGFA/VEGFR2 or non-specific siRNA. Exp Eye Res 2010; 91:433-9. [PMID: 20599960 DOI: 10.1016/j.exer.2010.06.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Revised: 05/11/2010] [Accepted: 06/20/2010] [Indexed: 11/15/2022]
Abstract
Choroidal neovascularization (CNV) is one of the severe pathological consequences of the end-stage of age-related macular degeneration (AMD). Several lines of evidence implicate increased levels of vascular endothelial growth factor (VEGF) in the retinas of AMD patients. Current available agents for the inhibition of VEGF protein such as bevacizumab show significant promise for the treatment of exudative AMD. However, this compound still has limited efficacy and requires multiple administrations; thus, it is associated with a variety of ocular complications, including endophthalmitis and retinal detachment. In this study, we used anti-VEGFA/VEGFR2 or non-specific siRNA and evaluated their suppression of laser-induced choroidal neovascularization (CNV) in mice. Male adult C57BL/6J mice were used in the study. The mice were subjected to laser rupture of Bruch's membrane to induce CNV and then randomized to five groups with six mice per group. The five groups were blank control, vehicle control (5% glucose solution, GS), VEGFA.siRNA, VEGFR2.siRNA, and non-specific siRNA. Two days after laser photocoagulation, each group, with the exception of the blank control group, received 1 microl of the appropriate agent by intravitreal injection to both eyes. Seven days later, after taking fundus photography and fundus fluorescein angiography (FFA), the mice were killed for tissue sampling. Six eyes from three mice in each group were used for choroidal flatmounts to examine CNV. Six eyes from three mice in each group were subjected to RNA extraction for VEGF mRNA quantification by qRT-PCR. Retinal tissue from 2 mice without laser treatment was harvested as the assay reference. The incidence of burns which showed fluorescein leakage was 80.0% in blank control, 75.0% in GS, 55.0% in VEGFA.siRNA, 40.0% in VEGFR2.siRNA and 30.0% in non-specific siRNA group. The flatmounted specimens showed that the retinal pigment epithelium (RPE) was visualized as a uniform hexagonal array in nonlasered areas. On day 7 after laser burn, well-defined isolectin-B4 labeled CNV networks were shown within the burn spots of the 2 control groups. The inhibitory effects of the 3 siRNAs on CNV formation were statistically significant as compared to the 2 control groups. Compared to the control groups, the ocular expression of VEGF mRNA was decreased significantly in the 3 siRNA treated groups. The areas of CNV correlated with the expression of VEGF mRNA. Anti-VEGFA/VEGFR2 or non-specific siRNA can inhibit CNV and attenuate VEGF mRNA expression in a laser-induced mouse model of CNV. We conclude that the siRNAs may be an option for treating patients with exudative AMD, and more studies are needed to test the possible side-effects of the treatment.
Collapse
Affiliation(s)
- Liping Gu
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | | | | | | | | |
Collapse
|
29
|
Miki A, Miki K, Ueno S, Wersinger DMB, Berlinicke C, Shaw GC, Usui S, Wang Y, Zack DJ, Campochiaro PA. Prolonged blockade of VEGF receptors does not damage retinal photoreceptors or ganglion cells. J Cell Physiol 2010; 224:262-72. [PMID: 20232317 DOI: 10.1002/jcp.22129] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
It has recently been reported that relatively short-term inhibition of vascular endothelial growth factor (VEGF) signaling can cause photoreceptor cell death, a potentially clinically important finding since VEGF blockade has become an important modality of treatment of ocular neovascularization and macular edema. However, in a set of studies in which we achieved extended and complete blockage of VEGF-induced vascular leakage through retinal expression of a VEGF binding protein, we did not observe any toxicity to retinal neurons. To follow-up on these apparently discrepant findings, we designed a set of experiments with the kinase inhibitor SU4312, which blocks phosphorylation of VEGF receptors, to look directly for evidence of VEGF inhibition-related retinal toxicity. Using transgenic mice with sustained expression of VEGF in photoreceptors, we determined that periocular injection of 3 microg of SU4312 every 5 days markedly suppressed subretinal neovascularization, indicating effective blockade of VEGF signaling. Wild-type mice given periocular injections of 5 microg of SU4312 every 5 days for up to 12 weeks showed normal scotopic and photopic electroretinograms (ERGs), no TUNEL stained cells in the retina, and no reduction in outer nuclear layer thickness. Incubation of cultured ganglion cells or retinal cultures containing photoreceptors with high doses of SU4312 did not reduce cell viability. These data suggest that blocking VEGF signaling in the retina for up to 12 weeks does not damage photoreceptors nor alter ERG function and should reassure patients who are receiving frequent injections of VEGF antagonists for choroidal and retinal vascular diseases.
Collapse
Affiliation(s)
- Akiko Miki
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-9277, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Grossniklaus HE, Kang SJ, Berglin L. Animal models of choroidal and retinal neovascularization. Prog Retin Eye Res 2010; 29:500-19. [PMID: 20488255 DOI: 10.1016/j.preteyeres.2010.05.003] [Citation(s) in RCA: 268] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There have been numerous types of animal models of choroidal neovascularization (CNV) and retinal neovascularization (RNV). Understanding the pathobiology of CNV and RNV is important when evaluating and utilizing these models. Both CNV and RNV are dynamic processes. A break or defect in Bruchs' membrane is necessary for CNV to develop. This may be induced with a laser, mechanically via surgery, or in the setting of transgenic mice. Some of the transgenic mouse models spontaneously develop RNV and/or retinal angiomatous proliferation (RAP)-like lesions. The pathogenesis of RNV is well-known and is generally related to ischemic retinopathy. Models of oxygen-induced retinopathy (OIR) closely resemble retinopathy of prematurity (ROP). The streptozotocin (STZ) rat model develops features similar to diabetic retinopathy. This review summarizes general categories and specific examples of animal models of CNV and RNV. There are no perfect models of CNV or RNV and individual investigators are encouraged to choose the model that best suits their needs.
Collapse
|
31
|
Yoshida T, Zhang H, Iwase T, Shen J, Semenza GL, Campochiaro PA. Digoxin inhibits retinal ischemia-induced HIF-1alpha expression and ocular neovascularization. FASEB J 2010; 24:1759-67. [PMID: 20065104 DOI: 10.1096/fj.09-145664] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Digoxin and other cardiac glycosides inhibit hypoxia-inducible factor-1 (HIF-1) transcriptional activity in cultured cells and suppress tumor xenograft growth. We tested the hypothesis that digoxin reduces HIF-1 levels in ischemic tissue in vivo and suppresses neovascularization. Well-established murine models of ocular neovascularization were used to test our hypothesis. In mice with ischemic retinopathy, intraocular or intraperitoneal injection of digoxin markedly reduced retinal levels of HIF-1alpha protein and mRNAs encoding multiple hypoxia-regulated proangiogenic proteins and their receptors. Daily intraperitoneal injection of 2 mg/kg starting at postnatal day (P) 12 or a single intravitreous injection of 100 ng of digoxin at P12 reduced retinal neovascularization by >70% at P17. Digoxin also reduced the number of CXCR4(+) cells and F4/80(+) macrophages in ischemic retina and significantly reduced choroidal neovascularization at Bruch's membrane rupture sites. Digoxin suppresses retinal and choroidal neovascularization by reducing HIF-1alpha levels, which blocks several proangiogenic pathways. Since digoxin suppresses multiple pathways in addition to VEGF signaling, it may provide advantages over specific VEGF antagonists for treatment of patients with retinal and choroidal diseases complicated by neovascularization and/or excessive vascular permeability. It may also be useful for treatment of neovascular diseases in other tissues.
Collapse
Affiliation(s)
- Tsunehiko Yoshida
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | |
Collapse
|
32
|
Bandello F, Pognuz R, Polito A, Pirracchio A, Menchini F, Ambesi M. Diabetic macular edema: classification, medical and laser therapy. Semin Ophthalmol 2009; 18:251-8. [PMID: 15513012 DOI: 10.1080/08820530390895262] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE to propose and describe a new classification of diabetic macular edema (DME) based on its etiopathogenetic features and to present efficacy and safety data on medical and laser treatments currently used and under investigation. METHODS available literature on DME has been reviewed and information provided by contact lens biomicroscopy, fluorescein angiography and optical coherence tomomography has been combined; published data from trials and reports investigating effectiveness of medical and laser treatments were also analyzed. RESULTS DME was classified according to its main etiopathogenetic components: prevalently retinovascular, tractional and with taut attached posterior hyaloid, each of them having peculiar aspects on fundus, angiographic and tomographic examination. Focal laser treatment remains the only effective intervention, as shown by a large randomized controlled trial, but new less-invasive photocoagulation strategies (i.e., light laser photocoagulation and subthreshold micropulse diode laser) as well as pharmacological approaches (i.e., antioxidants, PKC inhibitors, aldose-reductase and AGE inhibitors, VEGF inhibitors, intravitreal triamcinolone) are emerging. CONCLUSIONS based on our review a classification of DME was developed that may prove useful in making the appropriate treatment decision; the new laser and pharmacological treatments currently investigated are promising and need to be confirmed by large, controlled clinical trials.
Collapse
Affiliation(s)
- F Bandello
- Department of Ophthalmology, University of Udine, 33100 Udine, Italy.
| | | | | | | | | | | |
Collapse
|
33
|
Agostini H, Boden K, Unsöld A, Martin G, Hansen L, Fiedler U, Esser N, Marmé D. A Single Local Injection of Recombinant VEGF Receptor 2 But Not of Tie2 Inhibits Retinal Neovascularization in the Mouse. Curr Eye Res 2009; 30:249-57. [PMID: 15823917 DOI: 10.1080/02713680590923249] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE The purpose of this study was to develop pharmacological therapeutic alternatives for ischemia-induced proliferative retinopathy. METHODS C57BL/6J mice were placed in 76% oxygen on postnatal day 7 (P7) for 5 days. On P12 recombinant, chimeric vascular endothelial growth factor (sVEGF-R2) or sTie2 was injected intravitreally in one eye. The fellow eye received a control injection. On P17, retinal wholemounts were prepared after perfusion with fluorescein-dextran to quantify the retinopathy. RESULTS A single intravitreal injection of sVEGF-R2 reduced pathologic vascular changes significantly (p = 0.02). No significant effect was observed after intravitreal application of sTie2 (p = 0.07), although Ang-2 was upregulated in control animals without treatment as neovascularization developed and Ang-1 was constantly transcribed (ratio PCR). CONCLUSIONS sVEGF-R2 interferes with VEGF signaling via VEGF-R2 receptor. Thus, local application of soluble receptors for angiogenic factors is a possible therapy for proliferative retinopathy. Receptors with a wide range of ligands might prove more useful for local application than those binding few or antagonistic ligands.
Collapse
|
34
|
Identification of ARIA regulating endothelial apoptosis and angiogenesis by modulating proteasomal degradation of cIAP-1 and cIAP-2. Proc Natl Acad Sci U S A 2009; 106:8227-32. [PMID: 19416853 DOI: 10.1073/pnas.0806780106] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Endothelial apoptosis is a pivotal process for angiogenesis during embryogenesis as well as postnatal life. By using a retrovirus-mediated signal sequence trap method, we identified a previously undescribed gene, termed ARIA (apoptosis regulator through modulating IAP expression), which regulates endothelial apoptosis and angiogenesis. ARIA was expressed in blood vessels during mouse embryogenesis, as well as in endothelial cells both in vitro and in vivo. ARIA is a unique protein with no homology to previously reported conserved domain structures. Knockdown of ARIA in HUVECs by using small interfering RNA significantly reduced endothelial apoptosis without affecting either cell migration or proliferation. ARIA knockdown significantly increased inhibitor of apoptosis (cIAP)-1 and cIAP-2 protein expression, although their mRNA expression was not changed. Simultaneous knockdown of cIAP-1 and cIAP-2 abolished the antiapoptotic effect of ARIA knockdown. Using yeast 2-hybrid screening, we identified the interaction of ARIA with 20S proteasome subunit alpha-7. Thereafter, we found that cIAP-1 and cIAP-2 were degraded by proteasomes in endothelial cells under normal condition. Overexpression of ARIA significantly reduced cIAP-1 expression, and this reduction was abolished by proteasomal inhibition in BAECs. Also, knockdown of ARIA demonstrated an effect similar to proteasomal inhibition with respect to not only expression but also subcellular localization of cIAP-1 and cIAP-2. In vivo angiogenesis studied by Matrigel-plug assay, mouse ischemic retinopathy model, and tumor xenograft model was significantly enhanced by ARIA knockdown. Together, our data indicate that ARIA is a unique factor regulating endothelial apoptosis, as well as angiogenesis, presumably through modulating proteasomal degradation of cIAP-1 and cIAP-2 in endothelial cells.
Collapse
|
35
|
Yin OQP, Wang Y, Schran H. A mechanism-based population pharmacokinetic model for characterizing time-dependent pharmacokinetics of midostaurin and its metabolites in human subjects. Clin Pharmacokinet 2009; 47:807-16. [PMID: 19026036 DOI: 10.2165/0003088-200847120-00005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND AND OBJECTIVE Midostaurin, a novel potent inhibitor of protein kinase C enzyme and class III receptor tyrosine kinases, including Fms-like tyrosine kinase-3 (FLT3) and c-KIT, shows time-dependent pharmacokinetics in human subjects, presumably due to enzyme auto-induction. The purpose of this study was to develop a mechanism-based population pharmacokinetic model to describe the plasma concentration profiles of midostaurin and its metabolites and to characterize the time course of auto-induction. SUBJECTS AND METHODS Data from 37 diabetic patients who received oral doses of midostaurin (25 mg twice daily, 50 mg twice daily or 75 mg twice daily) for 28 days were analysed using nonlinear mixed-effects modelling. The structural model included a gut compartment for drug input and central and peripheral compartments for midostaurin, with drug output from the central compartment to either of two compartments for the midostaurin metabolites CGP62221 and CGP52421. Different enzyme induction sub-models were evaluated to account for the observed time-dependent decrease in midostaurin concentrations. RESULTS An enzyme turnover model, with CGP62221 formation (CL(1)) being a linear process but CGP52421 formation (CL(2)) being inducible, was found to be most appropriate. In the pre-induced state, CL(1) and CL(2) of midostaurin were determined to be 1.47 L/h and 0.501 L/h, respectively. At the end of 28 days of dosing, CL(2) was increased by 5.2-, 6.6- and 6.9-fold in the 25 mg, 50 mg and 75 mg groups, respectively, resulting in a 2.1- to 2.5-fold increase in total clearance of midostaurin. The final model estimated a mean maximum fold of induction (E(max)) of 8.61 and a concentration producing 50% of the E(max) (EC(50)) of 1700 ng/mL (approximately 2.9 micromol/L) for CGP52421-mediated enzyme induction. CONCLUSIONS The population pharmacokinetic model that was developed was able to describe the time-dependent pharmacokinetic profiles of midostaurin and its auto-induction mechanism. Thus it may be useful for designing an appropriate dosage regimen for midostaurin. The unique feature of this model included a precursor compartment that was able to capture the time delays of auto-induction. The use of such precursor extension in the model may be applicable to other drugs showing long time delays in enzyme auto-induction.
Collapse
Affiliation(s)
- Ophelia Q P Yin
- Oncology Clinical Pharmacology, Novartis Pharmaceuticals Corporation, Florham Park, New Jersey, USA
| | | | | |
Collapse
|
36
|
Wang Y, Yin OQP, Graf P, Kisicki JC, Schran H. Dose- and time-dependent pharmacokinetics of midostaurin in patients with diabetes mellitus. J Clin Pharmacol 2008; 48:763-75. [PMID: 18508951 DOI: 10.1177/0091270008318006] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Midostaurin is a novel potent inhibitor of both protein kinase C and the major receptor for vascular endothelial growth factor involved in angiogenesis, presenting a rationale for its use in diabetic retinopathy. This study evaluated the safety and pharmacokinetics of midostaurin following multiple oral doses of midostaurin for 28 days at 4 dose levels (25 mg bid, 50 mg bid, 75 mg bid, 75 mg tid), as well as a single oral 100-mg dose in patients with diabetes mellitus (n = 9-13 per dose cohort). Pharmacokinetic parameters were determined on days 1 and 28 based on the plasma concentrations of midostaurin and its metabolites, CGP62221 and CGP52421. The plasma exposures (C(max) and AUC(0-tau)) of midostaurin and metabolites increased less than proportionally over the dose range of 25 to 100 mg, showing a 2.2-fold increase after the first dose. Midostaurin concentrations increased during the first 3 to 6 days of dosing, then declined with time (by 30%-50%) until a steady state was achieved, representing an average accumulation factor (R) of 1.7. CGP62221 showed a similar concentration-time pattern as midostaurin (R = 2.5), but CGP52421 accumulated significantly (R = 18.8). A high-fat meal was found to significantly increase the C(max) and AUC(0-12 h) of midostaurin by 1.5-fold (P = .04) and 1.8-fold (P = .01), respectively, compared with taking the drug after an overnight fast. Midostaurin administered at 50 to 225 mg/day appeared to be generally safe in this group of patients. The most common treatment-related adverse events (eg, loose stools, nausea, vomiting, and headache) were found to be dose related, and the frequency increased markedly above the 150-mg/day dose level.
Collapse
Affiliation(s)
- Yanfeng Wang
- Oncology Clinical Pharmacology, Novartis Pharmaceuticals Corporation, 180 Park Ave, Florham Park, NJ 07932-0675, USA
| | | | | | | | | |
Collapse
|
37
|
Ueno S, Pease ME, Wersinger DMB, Masuda T, Vinores SA, Licht T, Zack DJ, Quigley H, Keshet E, Campochiaro PA. Prolonged blockade of VEGF family members does not cause identifiable damage to retinal neurons or vessels. J Cell Physiol 2008; 217:13-22. [PMID: 18543272 DOI: 10.1002/jcp.21445] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Several ocular diseases complicated by neovascularization are being treated by repeated intraocular injections of vascular endothelial growth factor (VEGF) antagonists. While substantial benefits have been documented, there is concern that unrecognized damage may be occurring, because blockade of VEGF may damage the fenestrated vessels of the choroicapillaris and deprive retinal neurons of input from a survival factor. One report has suggested that even temporary blockade of all isoforms of VEGF-A results in significant loss of retinal ganglion cells. In this study, we utilized double transgenic mice with doxycycline-inducible expression of soluble VEGF receptor 1 coupled to an Fc fragment (sVEGFR1Fc), a potent antagonist of several VEGF family members, including VEGF-A, to test the effects of VEGF blockade in the retina. Expression of sVEGFR1Fc completely blocked VEGF-induced retinal vascular permeability and significantly suppressed the development of choroidal neovascularization at rupture sites in Bruch's membrane, but did not cause regression of established choroidal neovascularization. Mice with constant expression of sVEGFR1Fc in the retina for 7 months had normal electroretinograms and normal retinal and choroidal ultrastructure including normal fenestrations in the choroicapillaris. They also showed no significant difference from control mice in the number of ganglion cell axons in optic nerve cross sections and the retinal level of mRNA for 3 ganglion cell-specific genes. These data indicate that constant blockade of VEGF for up to 7 months has no identifiable deleterious effects on the retina or choroid and support the use of VEGF antagonists in the treatment of retinal diseases.
Collapse
Affiliation(s)
- Shinji Ueno
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Inhibition of choroidal neovascularization by blocking vascular endothelial growth factor receptor tyrosine kinase. Jpn J Ophthalmol 2008; 52:91-98. [DOI: 10.1007/s10384-007-0506-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2006] [Accepted: 10/26/2007] [Indexed: 10/22/2022]
|
39
|
|
40
|
Furlani BA, Meyer CH, Rodrigues EB, Maia M, Farah ME, Penha FM, Holz FG. Emerging pharmacotherapies for diabetic macular edema. Expert Opin Emerg Drugs 2007; 12:591-603. [DOI: 10.1517/14728214.12.4.591] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
41
|
Hernández C, Simó R. Strategies for blocking angiogenesis in diabetic retinopathy: from basic science to clinical practice. Expert Opin Investig Drugs 2007; 16:1209-26. [PMID: 17685870 DOI: 10.1517/13543784.16.8.1209] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Proliferative diabetic retinopathy (PDR) demands both more effective and less expensive biologically based treatments. Our understanding of the pathophysiology of the disease is increasing as new biochemical pathways are identified. Most reports emphasize proangiogenic stimuli, with the natural inhibitory elements receiving little attention. There are two therapeutic strategies for blocking retinal angiogenesis in PDR: systemic drug administration (protein kinase C inhibitors and somatostatin analogs) or local therapies (anti-vascular endothelial growth factor strategies, anti-inflammatory agents, gene therapy and stem cell therapy). This review mainly focuses on the role of local therapies, especially intravitreous delivery, in the management of PDR. The potential for adverse effect are also discussed. The availability of these new strategies or the combination of them will not only be beneficial in treating PDR but may also result in a shift towards treating earlier stages of diabetic retinopathy, thus easing the burden of this devastating disease.
Collapse
Affiliation(s)
- Cristina Hernández
- Hospital Universitari Vall d'Hebron, Diabetes and Metabolism Research Unit, Endocrinology Division, Pg. Vall d'Hebron, Barcelona, Spain
| | | |
Collapse
|
42
|
Lima e Silva R, Shen J, Hackett SF, Kachi S, Akiyama H, Kiuchi K, Yokoi K, Hatara MC, Lauer T, Aslam S, Gong YY, Xiao WH, Khu NH, Thut C, Campochiaro PA. The SDF‐1/CXCR4 ligand/receptor pair is an important contributor to several types of ocular neovascularization. FASEB J 2007; 21:3219-30. [PMID: 17522382 DOI: 10.1096/fj.06-7359com] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Hypoxia causes increased expression of several proteins that have the potential to promote neovascularization. Vascular endothelial growth factor (VEGF) is up-regulated by hypoxia in the retina and plays a central role in the development of several types of ocular neovascularization, but the effects of other hypoxia-regulated proteins are less clear. Stromal-derived factor-1 (SDF-1) and its receptor, CXCR4, have hypoxia response elements in the promoter regions of their genes and are increased in hypoxic liver and heart. In this study, we found that SDF-1 and CXCR4 are increased in hypoxic retina, with SDF-1 localized in glial cells primarily near the surface of the retina and CXCR4 localized in bone marrow-derived cells. Glial cells also expressed CXCR4, which suggested the possibility of autocrine stimulation, but influx of bone marrow-derived cells is the major source of increased levels of CXCR4. High levels of VEGF in the retina in the absence of hypoxia also increased levels of Cxcr4 and Sdf1 mRNA. CXCR4 antagonists reduced influx of bone marrow-derived cells into ischemic retina and strongly suppressed retinal neovascularization, VEGF-induced subretinal neovascularization, and choroidal neovascularization. These data suggest that SDF-1 and CXCR4 contribute to the involvement of bone marrow-derived cells and collaborate with VEGF in the development of several types of ocular neovascularization. They provide new targets for therapeutic intervention that may help to bolster and supplement effects obtained with VEGF antagonists.
Collapse
Affiliation(s)
- Raquel Lima e Silva
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287-9277, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Anecortave acetate is a unique angiostatic agent. Although derived from the glucocorticoid cortisol acetate, it was designed to be devoid of glucocorticoid activity while retaining efficacious anti-angiogenic activity. These modifications have led to a new class of anti-angiogenic agents, the angiostatic cortisenes. Anecortave acetate has broad based angiostatic activity, inhibiting neovascularization at multiple steps. Unlike several other angiostatic agents that selectively target only one angiogenic factor, anecortave acetate inhibits neovascularization induced by many different angiogenic factors.
Collapse
Affiliation(s)
- Abbot F Clark
- Discovery Research, Alcon Research, Ltd., Fort Worth, Texas 76134, USA
| |
Collapse
|
44
|
Dorrell M, Uusitalo-Jarvinen H, Aguilar E, Friedlander M. Ocular neovascularization: basic mechanisms and therapeutic advances. Surv Ophthalmol 2007; 52 Suppl 1:S3-19. [PMID: 17240254 DOI: 10.1016/j.survophthal.2006.10.017] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The vast majority of diseases that cause catastrophic loss of vision do so as a result of ocular neovascularization. During normal retinal vascular development, vascular endothelial cells proliferate and migrate through the extracellular matrix in response to a variety of cytokines, leading to the formation of new blood vessels in a highly ordered fashion. During abnormal neovascularization of the iris, retina, or choroid, angiogenesis is unregulated and usually results in the formation of dysfunctional blood vessels. When these newly formed vessels leak fluid, hemorrhage, or are associated with fibrous proliferation, retinal edema, retinal/vitreous hemorrhage, or traction retinal detachments may occur resulting in potentially catastrophic loss of vision. In this review, we will briefly discuss the scope of the clinical problem and the general underlying principles of angiogenesis. We will focus on recent laboratory advances that have led to the development of therapeutics useful in the treatment of neovascular eye diseases. We will describe compounds currently in pre-clinical development stages as well as the results of clinical trials involving the use of these drugs as treatments for ocular neovascularization.
Collapse
Affiliation(s)
- Michael Dorrell
- Department of Cell Biology, The Scripps Research Institute, Department of Ophthalmology, Scripps Clinic, La Jolla, California 92014, USA
| | | | | | | |
Collapse
|
45
|
Abstract
Most diseases that cause catastrophic loss of vision do so as a result of abnormal angiogenesis and wound healing, often in response to tissue ischemia or inflammation. Disruption of the highly ordered tissue architecture in the eye caused by vascular leakage, hemorrhage, and concomitant fibrosis can lead to mechanical disruption of the visual axis and/or biological malfunctioning. An increased understanding of inflammation, wound healing, and angiogenesis has led to the development of drugs effective in modulating these biological processes and, in certain circumstances, the preservation of vision. Unfortunately, such pharmacological interventions often are too little, too late, and progression of vision loss frequently occurs. The recent development of progenitor and/or stem cell technologies holds promise for the treatment of currently incurable ocular diseases.
Collapse
Affiliation(s)
- Martin Friedlander
- Department of Cell Biology, The Scripps Research Institute, and Division of Ophthalmology, Scripps Clinic, La Jolla, CA 92037, USA.
| |
Collapse
|
46
|
Roldán-Pallarés M, Rollín R, Martínez-Montero JC, Fernández-Cruz A, Bravo-Llata C, Fernández-Durango R. IMMUNOREACTIVE ENDOTHELIN-1 IN THE VITREOUS HUMOR AND EPIRETINAL MEMBRANES OF PATIENTS WITH PROLIFERATIVE DIABETIC RETINOPATHY. Retina 2007; 27:222-35. [PMID: 17290206 DOI: 10.1097/01.iae.0000231376.76601.40] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To investigate the potential role of endothelin-1 (ET-1), a potent vasoconstrictor with mitogenic properties, in the pathogenesis of proliferative diabetic retinopathy (PDR). METHODS Plasma and vitreous samples were collected from normal patients (controls; n = 25), diabetic patients with PDR (n = 25), and diabetic patients with non-PDR (n = 25). The patients had to have epiretinal membranes (ERMs) or other ocular conditions that made them candidates for vitrectomy. Immunoreactive ET-1 (IR-ET-1) was assayed in plasma and vitreous samples by radioimmunoassay. IR-ET-1 was immunohistochemically localized in ERMs. Expression of endothelin receptors A (ETA) and B (ETB) was confirmed by reverse transcription-polymerase chain reaction analysis. RESULTS IR-ET-1 levels in plasma and vitreous samples from diabetic patients were higher (P < 0.0001) than those in samples from the control group. The levels for patients with PDR were even higher (P < 0.0001) than those for patients with non-PDR. Eyes with ERMs in the PDR group had the highest vitreous IR-ET-1 levels (14.67 +/- 0.67 pg/mL). IR-ET-1 was localized in the cellular and stromal components of ERMs in diabetic and nondiabetic patients. Furthermore, the ETA and ETB receptors were expressed in both diabetic and nondiabetic ERMs. CONCLUSIONS Diabetic patients with PDR and ERMs had the highest plasma and vitreous IR-ET-1 levels. ET-1 and its ETA and ETB receptors were present in ERMs. These data suggest that ET-1 is involved in diabetic vitreoretinal disease.
Collapse
Affiliation(s)
- Manuela Roldán-Pallarés
- Departamento de Oftalmología, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
47
|
Campochiaro PA. Seeing the light: New insights into the molecular pathogenesis of retinal diseases. J Cell Physiol 2007; 213:348-54. [PMID: 17654481 DOI: 10.1002/jcp.21213] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In the past, most treatments for retinal diseases have been empirical. Steroids and/or laser photocoagulation and/or surgery have been tried for almost every condition with little or no understanding of the underlying disease. Over the past several years vision researchers have uncovered molecular components of processes, such as visual transduction and the visual cycle, that are critical for visual function, and identified other molecules that lead to dysfunction and disease processes such as neovascularization and macular edema. It is becoming clear that dysregulation of certain molecules can have major effects on retinal structure and function. Studies in animal models have suggested that inhibiting or augmenting levels of a single molecule can have major effects in complex disease processes. Although several molecules probably contribute to neovascularization and excessive vascular permeability in the eye, blockade of vascular endothelial growth factor (VEGF) has remarkable beneficial effects in animal models that have now been proven to apply to human diseases in clinical trials. Intraocular injection of VEGF antagonists has revolutionized the treatment of choroidal neovascularization (CNV) and macular edema and serves as a model of targeted ocular pharmacotherapy. Significant progress elucidating the molecular pathogenesis of several disease processes in the eye may soon lead to new treatments following the lead of VEGF antagonists. Initial treatments that provide benefit from frequent intraocular injections are likely to be followed by sustained delivery of drugs and/or prolonged protein delivery by gene transfer. The eye has entered the era of molecular therapy.
Collapse
Affiliation(s)
- Peter A Campochiaro
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-9277, USA.
| |
Collapse
|
48
|
Ohneda O, Nagano M, Fujii‐Kuriyama Y. Role of Hypoxia‐Inducible Factor‐2α in Endothelial Development and Hematopoiesis. Methods Enzymol 2007; 435:199-218. [DOI: 10.1016/s0076-6879(07)35011-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
49
|
Yeoh J, Sims J, Guymer RH. A review of drug options in age-related macular degeneration therapy and potential new agents. Expert Opin Pharmacother 2006; 7:2355-68. [PMID: 17109611 DOI: 10.1517/14656566.7.17.2355] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Age-related macular degeneration (AMD) is the leading cause of legal blindness in people > 50 years of age in the developed world. AMD is both a debilitating and costly disease for the individual and the community. Greater understanding of the mechanisms and pathways involved in causing the visual loss in AMD has resulted in the advent of several newer and more effective treatment options, making it an exciting time in the management of AMD. This paper will examine the principles behind the existing drug therapies available, as well as those being developed in the management or prophylaxis of AMD and its vision-threatening complications.
Collapse
Affiliation(s)
- Jonathan Yeoh
- Centre for Eye Research Australia, The University of Melbourne, Australia
| | | | | |
Collapse
|
50
|
Lima e Silva R, Kachi S, Akiyama H, Shen J, Hatara MC, Aslam S, Gong YY, Khu NH, Lauer TW, Hackett SF, Marton LJ, Campochiaro PA. Trans-scleral delivery of polyamine analogs for ocular neovascularization. Exp Eye Res 2006; 83:1260-7. [PMID: 16950258 DOI: 10.1016/j.exer.2006.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2006] [Revised: 05/26/2006] [Accepted: 07/01/2006] [Indexed: 10/24/2022]
Abstract
Periocular injections of the polyamine analog CGC-11144 three times a week causes regression of choroidal neovascularization. This regimen was selected to maximize chances of success for proof of concept, but is not ideal for clinical application. In this study we explored other regimens for periocular delivery of CGC-11144, and 2 other polyamine analogs, CGC-11047 and CGC-11093. A single periocular injection of 200 microg of CGC-11144, 2 mg of CGC-11047, or 1.5 mg of CGC-11093 caused significant suppression and regression of laser-induced choroidal neovascularization. An injection of 2 mg of CGC-11047 or 1.5 mg of CGC-11093 one or two weeks before, but not 3 weeks before, rupture of Bruch's membrane also caused significant suppression. Periocular injection of polyamine analogs also caused strong inhibition of retinal or subretinal neovascularization in mice with oxygen-induced ischemic retinopathy or Rhodopsin promoter/VEGF transgenic mice, respectively. These data suggest that periocular injection of one of 3 different polyamine analogs inhibits retinal or choroidal neovascularization and a single injection provides inhibitory activity for at least 2 to 3 weeks, which could provide the basis for a feasible treatment regimen for clinical trials.
Collapse
Affiliation(s)
- Raquel Lima e Silva
- The Departments of Ophthalmology and Neuroscience, The Johns Hopkins University School of Medicine, Maumenee 719, 600 N. Wolfe Street, Baltimore, MD 21287-9277, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|