·Basic Research· Current
Issue· ·Achieve· ·Search Articles· ·Online Submission· ·About IJO· PMC
Citation: Lee JE,
Kim JY, Jung JH, Shin DH, Park SW. Induction of vascular endothelial growth factor
receptor expression in human umbilical vein endothelial cells after repeated
bevacizumab treatment in vitro. Int J Ophthalmol
2017;10(7):1064-1068
Induction of vascular endothelial
growth factor receptor expression in human umbilical vein endothelial cells
after repeated bevacizumab treatment in vitro
Ji Eun Lee1,2, Jin Young Kim1,
Jae Ho Jung1,3 , Dong Hoon Shin3,4, Sung
Who Park1,2
1Department of Ophthalmology, School of Medicine, Pusan National
University, Yangsan 50612, Korea
2Medical Research Institute, Pusan National University Hospital,
Busan 49241, Korea
3Research Institute for Convergence of Biomedical
Science and Technology, Pusan National University Yangsan Hospital, Yangsan
50612, Korea
4Department of Pathology, School of Medicine, Pusan National
University, Yangsan 50612, Korea
Correspondence to: Sung Who Park. Department of Ophthalmology, School of Medicine,
Pusan National University & Medical Research Institute, Pusan National
University Hospital, 179 Gudeok-ro, Seo-gu, Pusan 49241, Korea. oph97@naver.com
Received:
2016-03-15
Accepted: 2017-01-09
AIM:
To investigate the mechanism underlying the loss of responsiveness to
anti-vascular endothelial growth factor (VEGF) treatment after repeated
injections for choroidal neovascularization, VEGF and VEGF receptor (VEGFR)
expressions were evaluated following repeated bevacizumab treatments in hypoxic
human umbilical vein endothelial cells (HUVECs) in vitro.
METHODS: HUVECs
were incubated under hypoxic conditions in two media of different bevacizumab
concentrations (1.0 or 2.5 mg/mL) for 17h, and then in a new medium without
bevacizumab for 7h. This procedure was repeated twice more. A culture with an
identical volume of excipients served as the control. Cytotoxicity and cell
proliferation were assessed using 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl
tetrazolium bromide and Ki-67 assays, respectively. Levels of VEGF and VEGFR
were assessed using enzyme-linked immunosorbent assay and Western blot
respectively.
RESULTS: Cytotoxic
effects were not reported for either bevacizumab concentration. Cell proliferation
was not reduced after anti-VEGF treatments. VEGF level after single treatment
was significantly higher than that of the control and after repeated treatments.
Phosphorylated VEGFR-2 expression increased significantly after single and
repeated bevacizumab treatments compared with the control. The 1.0 mg/mL
bevacizumab induced significantly higher expressions of VEGFR-2 than the 2.5
mg/mL in single and repeated treatment groups.
CONCLUSION: Bevacizumab
treatment of HUVECs elevated VEGFR expression in both single and repeated
treatments, indicating a mechanism for the reduced efficacy of anti-VEGF
therapy in ocular neovascular disorders.
KEYWORDS: vascular
endothelial growth factor; vascular endothelial growth factor receptor; choroidal
neovascularization; bevacizumab; repeated treatments
DOI:10.18240/ijo.2017.07.07
Citation: Lee JE, Kim JY, Jung JH, Shin DH, Park SW. Induction of vascular endothelial growth factor
receptor expression in human umbilical vein endothelial cells after repeated
bevacizumab treatment in vitro. Int J Ophthalmol
2017;10(7):1064-1068
Exudative
age-related macular degeneration (ARMD) complicated with choroidal
neovascularzation (CNV) is the leading cause of visual loss in developed countries[1-3]. CNV is thought to result from the
orchestrated interaction of various cellular components and a number of growth
factors[4], with vascular endothelial growth
factor (VEGF) being a key factor.
The
introduction of therapies targeting VEGF has changed the treatment paradigm of
neovascular ARMD[5-6]; however,
anti-VEGF treatment has some limitations, including the necessity of repeated
injections and non-responders in clinical practice[7].
Some patients who initially respond to anti-VEGF treatment lose responsiveness
following repeated injections[8-10].
Although the exact mechanism underlying this tolerance (also known as
tachyphylaxis) is still unknown, reduction of therapeutic effects is known to
develop in numerous ways and for different reasons including vascular
structural changes[9], systemic immune responses[11], up-regulation of other angiogenic signaling pathways
as a compensatory mechanism[4], and increased
expression of VEGF or VEGF receptors (VEGFRs)[12].
Repeated
anti-VEGF treatments under hypoxic conditions and comparison between single and
repeated anti-VEGF treatments would be essential to elucidate these mechanisms.
A previous study has shown that a single treatment of anti-VEGF eliminates VEGF
and reduces VEGFR expression in a non-hypoxic condition[13].
These laboratory observations cannot explain the changes in responsiveness to
repeated anti-VEGF treatments under hypoxia. This is because repeated
injections are performed in clinical settings where choroidal hypoxia is
suggested to be the mechanism for exudative ARMD, based on choroidal thinning
and presence of the watershed zone[14-15].
We
performed an in vitro study using human umbilical vein endothelial cells
(HUVECs) with single and repeated anti-VEGF treatments under hypoxic conditions
to observe changes in VEGF and VEGFR expression.
Cell
Culture Under Hypoxic Condition HUVECs
(Science Cell Research Labs, San Diego, CA, USA) between passages 4 and 9 were
cultured in M199 medium (Sigma-Aldrich, St. Louis, MO, USA) supplemented with
20% fetal bovine serum (Invitrogen Life Technologies, Scotland, UK), 1%
penicillin/streptomycin (Invitrogen Life Technologies), 0.1% heparin
(Sigma-Aldrich), and 0.1% endothelial cell growth supplement (Sigma-Aldrich), and
maintained at 37℃ in a humidified 5% CO2 condition.
HUVECs were subjected to a hypoxic condition when 70%-90% confluence was
achieved.
To
achieve the hypoxia condition, tissue-culture plates with HUVECs were placed in
a hypoxic chamber (Modular Incubator Chamber, Billups-Rothenberg, Del Mar, CA,
USA) and flushed with a gas mixture comprising 1% O2, 5% CO2,
and 94% N2 for 5min at 10 L/min. The chambers were then closed and
placed in a CO2 incubator at 37℃. The chamber gas was replaced daily. The
medium was pre-equilibrated with a 5% O2 gas mixture overnight
before the hypoxic experiments were performed.
Bevacizumab
Treatment First,
HUVECs were treated with 1.0 mg/mL and 2.5 mg/mL bevacizumab (Avastin®;
Genentech, Inc. San Francisco, CA, USA) for 17h under hypoxia with culture
medium, and then the dish was washed out five times with phosphate buffered
saline (PBS) and replaced with a new culture medium for 7h under the same
hypoxic condition. Second, this procedure was repeated twice more. Third, for
the control, the same volume of excipients[16]
was added to the HUVECs, and the cells incubated for 17h under hypoxia; the
dish was then washed out five times with PBS and incubated in new medium for 7h
under the same hypoxic condition.
Assays Each assay
was performed immediately to prevent exposure to normal O2 when the
testing condition was accomplished. The number of replicates varied from 6 to
14.
Potential
cell metabolic activity reduction after bevacizumab treatment on HUVECs were
evaluated using an MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium
bromide] assay. The cells were washed twice with PBS and then subjected to the
MTT assay as previously described[17]. Optical
density (OD) was measured at 540 nm.
Secreted
VEGF concentrations were quantified using a human VEGF enzyme-linked immunosorbent
assay kit (Quantikine, Human VEGF immunoassay, R&D System Inc.,
Minneapolis, MN, USA) in accordance with the manufacturer’s instructions. ODs
were measured at 450-570 nm, and the VEGF was calculated as the
protein-adjusted amount of VEGF (pg/mg protein).
Activated
VEGFR-2 expression was assessed using Western blot. Proteins were isolated from
HUVEC lysates using Tripure (Roche Diagnostics, Basel, Switzerland), quantified
using a spectrophotometer (Jenway, 6405 UV/vis, Essex, UK), and then equal
amounts of the protein was loaded on 8% sodium dodecyl sulfate polyacrylamide
gel electrophoresis with a 5% stacking gel. After electrophoresis, the proteins
were blotted onto a Hybond nitrocellulose membrane (Amersham Biosciences,
Piscataway, NJ, USA), using a mini-transblot electrophoretic transfer cell
(Amersham Biosciences). Immunodetection of phosphorylated (activated) VEGFR-2
and beta-actin were accomplished using enhanced chemiluminiscence (ECL kit,
Amersham Biosciences). The relative intensity of each protein on the blotting
analysis was measured using a computerized software program (Bio-Rad, Richmond,
CA, USA) and normalized with beta-actin bands to compare the expression of
proteins between different treatment conditions and concentrations.
Cell
proliferation in the different culture conditions was determined using a
monoclonal antibody that reacted with nuclear antigen Ki-67 of proliferating
human cells in the G1-M phase, but not in the G0 phase. A primary rabbit
anti-human Ki-67 antibody (Dako, Carpentiria, CA, USA) was utilized for
cytopsin preparation at a 1:40 dilution for 1h at room temperature in a moist
chamber. Excess antibodies were removed by washing the slides with
Trisaminomethane-buffered saline (TBS, Sigma-Aldrich). Negative controls with
TBS and an irrelevant antibody were established alongside the test slides.
Fluorescein isothiocyanate (FITC)-conjugated swine anti-rabbit antibodies
(Dako, Glostrup, Denmark) in a 1:20 dilution were utilized as the secondary
antibodies and incubated for 30min. The slides were mounted with glycerol/PBS.
The proportion of positively labeled cytopsin cells was determined by analyzing
200 cells per slide under a fluorescent microscope.
Statistical
Analysis We compared MTT, VEGF
secretion, VEGFR-2 expression, and Ki-67 between the control and each
bevacizumab treatment condition. We also compared VEGF secretion and VEGFR-2
expression between 1.0 mg/mL and 2.5 mg/mL bevacizumab in the single and
repeated bevacizumab treatment groups. Wilcoxon rank sum tests were used for
statistical analyses that were performed using the Statistical Package for
Social Sciences (SPSS 12.0 Inc., Chicago, IL, USA). P values <0.05
were considered statistically significant.
The
MTT assay revealed that cell metabolic activity was not affected by each
bevacizumab treatment condition compared with the control (all P>0.05).
The MTT assay was performed in highly confluent cell cultures with slow
replication rates. This ensured that the effects of bevacizumab on cell
proliferation, apoptosis, and cell morphology did not mask its potential toxic
effects.
The
amount of VEGF secretion with single 1.0 mg/mL and 2.5 mg/mL bevacizumab
treatment was higher than the control and each repeated bevacizumab treatment
conditions (all P<0.05, Figure 1), and there was no difference
between 1.0 mg/mL and 2.5 mg/mL bevacizumab treatments (P=0.5, Figure
1). VEGF secretion with 1.0 mg/mL and 2.5 mg/mL repeated bevacizumab treatments
was similar to the controls (all P>0.05, Figure 1) and similar between
both concentrations (P=0.3, Figure 1).
Figure
1 Results of secreted VEGF levels under hypoxic conditions The values were calculated from
separate experiments. 1:1.0 mg/mL bevacizumab single injection; 2.5:2.5 mg/mL
bevacizumab single injection; 1×3: 1.0 mg/mL bevacizumab repeated injection;
2.5×3: 2.5 mg/mL bevacizumab repeated injection. aP<0.05 vs
control, cP<0.05 vs 1.0 mg/mL in repeated
treatments, eP<0.05 vs 2.5 mg/mL in repeated
treatments.
The
VEGFR-2 expression level for each single bevacizumab treatment was
significantly higher than that the control (P=0.010 in 1.0 mg/mL and P=0.020
in 2.5 mg/mL, Figure 2), and the repeated bevacizumab treatment at each
concentration also resulted in a higher expression compared with the control (P<0.001
in 1.0 mg/mL and P=0.010 in 2.5 mg/mL, Figure 2). When comparing between
each bevacizumab concentration, 1.0 mg/mL significantly induced higher VEGFR-2
expression than 2.5 mg/mL bevacizumab in both single (P=0.03, Figure 2)
and also repeated bevacizumab treatment conditions (P=0.02, Figure 2).
Figure
2 Expression of VEGFR-2 under hypoxic conditions were evaluated by Western blot
(A) and statically compared to each groups using relative intensity of each
label from separate experiments (B) 1: 1.0 mg/mL bevacizumab single
injection; 2.5: 2.5 mg/mL bevacizumab single injection; 1×3: 1.0 mg/mL
bevacizumab repeated injection; 2.5×3: 2.5 mg/mL bevacizumab repeated
injection. aP<0.05 vs control, cP<0.05
vs 1.0 mg/mL in single treatment, eP<0.05 vs 2.5
mg/mL in single treatment, gP<0.05 vs 1.0 mg/mL in
repeated treatments, iP<0.05 vs 2.5 mg/mL in
repeated treatments.
Ki-67
expression, an indicator of the number of proliferating cells, was not
significantly different between each treatment group and the control (all P>0.05).
Exudative
ARMD is a chronic disease that requires long-term management[16-17]. Monthly injections of ranibizumab over a 2-year
period have been shown to be ineffective at preventing CNV recurrence and lead
to the development of a loss of responsiveness in some patients[8-10]. The reduction in anti-VEGF
treatment response challenges long-term treatment efficacy. Pathophysiological
mechanisms have yet to be elucidated with ongoing research into effective
treatments for the prevention of this progressive condition.
Angiogenesis
is a highly complex and coordinated process requiring multiple intercellular
mediators and receptors. VEGF is the most important activating factor of
angiogenesis. It mediates a wide range of responses primarily in the vascular
endothelial cells, from normal physiological functions to pathological disease
progression. VEGF is not only an angiogenic factor but also a trophic factor
for cell survival to prevent hypoxic damage[18-20]. Accordingly, depletion of VEGF by anti-VEGF
treatment may induce compensatory physiological responses. VEGF and VEGFR
levels following anti-VEGF treatment would be a key indicator of treatment
outcome for intraocular neovascular diseases.
In
our study, secretion of VEGF increased after a single treatment of bevacizumab;
however, VEGF secretion returned to control levels with repeated treatment. In contrast,
VEGFR expression was significantly elevated after a single and repeated
treatment, and 1.0 mg/mL bevacizumab treatment induced higher VEGFR-2
expression than 2.5 mg/mL concentration in both a single and repeated treatment
was. A previous study reported that VEGFR expression decreases in HUVECs after
treatment with various anti-VEGF agents under non-hypoxic conditions[13]. This discrepancy may be due to the different in
vitro HUVEC culture conditions including hypoxic condition versus normal
oxygen status between studies.
The
present findings may be considered a physiological mechanism for HUVEC survival
in order to compensate the depletion of VEGF under hypoxic conditions. We
suggest that increased secretion of VEGF after a single treatment accounts for
a prompt response to VEGF ablation, and increased expression of VEGFR accounts
for a delayed response for the maintenance of HUVEC cell survival. Although
these two stages of responses are postulated to compensate for anti-VEGF
treatment, the effects of anti-VEGF treatment may be different at each stage.
Increased
secretion of VEGF may be blocked by repeated treatments or higher doses of
anti-VEGF. However, up-regulated VEGFR will increase sensitivity to VEGF and
diminish responsiveness to anti-VEGF therapy. This may comprise a potential
mechanism for tolerance to anti-VEGF treatment, at least in some patients.
Our
experiments demonstrated there was no significant change in cell metabolic activity
based on MTT assay and cell proliferation based on Ki-67 expression after
treatment with 1.0 and 2.5 mg/mL bevacizumab. These findings suggest that our
experimental anti-VEGF treatment concentrations did not achieve
anti-proliferative effects on HUVECs. Anti-VEGF therapeutic effects would be
compensated by up-regulated VEGF secretion and VEGF receptor expression, as
observed in our study.
We
also recorded an increase in the expression of VEGFR with the lowest treatment
dose of bevacizumab. We propose that suboptimal treatment may result in a more
rapid loss of responsiveness to anti-VEGF treatment due to the up-regulation of
VEGFR under hypoxic conditions. Therefore, achieving optimal therapeutic
concentrations may be essential for improving treatment outcomes and preventing
tolerance in exudative ARMD.
Our
study has several limitations. First, the repeated treatment interval was much
shorter than in clinical practice. Expression changes of other cytokines
related to angiogenesis under the hypoxic condition were not investigated. As
mentioned earlier, neovascularization is the result of interactions among
numerous cells and cytokines. Finally, the pathway for the upregulated VEGF and
VEGFR-2 expression was not evaluated in the current experiment.
However,
we demonstrated that the response to anti-VEGF treatment may vary and be
occasionally paradoxical according to treatment dose and repetition. Further
studies are needed to find the feedback mechanism for VEGFR-2 expression
related to VEGF level under a hypoxic condition.
In
summary, bevacizumab treatment of hypoxic HUVECs potentially increased VEGFR
expression to maintain cellular metabolism under hypoxia. Our results may
partially explain reduced responsiveness after repeated anti-VEGF treatment for
neovascular ARMD in clinical settings.
Authors’
Contributions: Lee JE was responsible for the study conception and design, data
acquisition, data analysis, data interpretation and drafting of the article
manuscript. Lee JE also contributed substantially to the revising of the
article manuscript and approved its final version. Kim JY contributed to data
acquisition, data interpretation. Kim JY approved the final version of the
article manuscript. Jung JH contributed to the study design, data acquisition.
Jung JH approved the final version of the article manuscript. Shin DH
contributed to the study design and was responsible for revising the article
manuscript. Park SW was responsible for the study and design, and contributed
substantially to the data acquisition, data interpretation and revising of the
article manuscript. Park SW approved the final version of the article
manuscript. Guarantors for the article are Park SW and Lee JE.
Conflicts
of Interest: Lee JE, None; Kim JY, None; Jung JH, None;
Shin DH, None; Park SW, None.
1 Klein R, Klein BE, Linton KL. Prevalence of age-related maculopathy.
The Beaver Dam Eye Study. Ophthalmology
1992;99(6):933-943. [CrossRef]
2 Munoz B, West SK, Rubin GS, Schein OD, Quigley HA, Bressler SB,
Bandeen-Roche K. Causes of blindness and visual impairment in a population of
older Americans: The Salisbury Eye Evaluation Study. Arc Ophthalmol 2000;118(6):819-825. [CrossRef]
3 Prokofyeva E, Zrenner E. Epidemiology of major eye diseases leading to
blindness in Europe: a literature review. Ophthalmic
Res 2012;47(4):171-188. [CrossRef] [PubMed]
4 Spaide RF. Rationale for combination therapy in age-related macular
degeneration. Retina 2009;29(6
Suppl):S5-7. [CrossRef] [PubMed]
5 Rosenfeld PJ, Brown DM, Heier JS, Boyer DS, Kaiser PK, Chung CY, Kim
RY, MARINA Study Group. Ranibizumab for neovascular age-related macular
degeneration. N Engl J Med 2006;355(14):1419-1431.
[CrossRef] [PubMed]
6 Brown DM, Kaiser PK, Michels M, Soubrane G, Heier JS, Kim RY, Sy JP,
Schneider S, ANCHOR Study Group. Ranibizumab versus verteporfin for neovascular
age-related macular degeneration. N Engl
J Med 2006;355(14):1432-1444. [CrossRef] [PubMed]
7 Haller JA. Current anti-vascular endothelial growth factor dosing
regimens: benefits and burden. Ophthalmology
2013;120(5 Suppl):S3-7. [CrossRef] [PubMed]
8 Keane PA, Liakopoulos S, Ongchin SC, Heussen FM, Msutta S, Chang KT,
Waslsh AC, Sadda SR. Quantitative subanalysis of optical coherence tomography
after treatment with ranibizumab for neovascular age-related macular
degeneration. Invest Ophthalmol Vis Sci
2008;49(7):3115-3120. [CrossRef] [PMC free article] [PubMed]
9 Forooghian F, Cukras C, Meyerle CB, Chew EY, Wong WT. Tachyphylaxis
after intravitreal bevacizumab for exudative age-related macular degeneration. Retina 2009;29(6):723-731. [CrossRef] [PMC free article] [PubMed]
10 Gasperini JL, Fawzi AA, Khondkaryan A, Lam L, Chong LP, Elioot D, Walsh
AC, Hwang J, Sadda SR. Bevacizumab and ranibizumab tachyphylaxis in the
treatment of choroidal neovascularisation. Br
J Ophthalmol 2012;96(1):14-20. [CrossRef] [PubMed]
11 Forooghian F, Chew EY, Meyerle CB, Cukras C, Wong WT. Investigation
of the role of neutralizing antibodies against bevacizumab as mediators of
tachyphylaxis. Acta Ophthalmol
2011;89(2):e206-207. [CrossRef] [PubMed]
12 Binder S. Loss of reactivity in intravitreal anti-VEGF therapy:
tachyphylaxis or tolerance? Br J
Ophthalmol 2012;96(1):1-2. [CrossRef] [PubMed]
13 Carneiro A, Falcao M, Pirraco A, Milheiro-Oliveira P, Falcao-Reis F,
Soares R. Comparative effects of bevacizumab, ranibizumab and pegaptanib at
intravitreal dose range on endothelial cells. Exp Eye Res 2009;88(3):522-527. [CrossRef] [PubMed]
14 Chung SE, Kang SW, Lee JH, Kim YT. Choroidal thickness in polypoidal
choroidal vasculopathy and exudative age-related macular degeneration. Ophthalmology 2011;118(5):840-845. [CrossRef] [PubMed]
15 Xu W, Grunwald JE, Metelitsina TI, DuPont JC, Ying GS, Martin ER,
Dunaief JL, Brucker AJ. Association of risk factors for choroidal
neovascularization in age-related macular degeneration with decreased foveolar
choroidal circulation. Am J Ophthalmol 2010;150(1):40-47.e42.
[CrossRef] [PMC free article] [PubMed]
16 Singer MA, Awh CC, Sadda S, Freeman WR, Antoszyk AN, Wong P, Tuomi L.
HORIZON: an open-label extension trial of ranibizumab for choroidal
neovascularization secondary to age-related macular degeneration. Ophthalmology 2012;119(6):1175-1183. [CrossRef] [PubMed]
17 Rofagha S, Bhisitkul RB, Boyer DS, Sadda SR, Zhang K, Group S-US.
Seven-Year Outcomes in Ranibizumab-Treated Patients in ANCHOR, MARINA, and
HORIZON: A Multicenter Cohort Study (SEVEN-UP). Ophthalmology 2013;120(11):2292-2299. [CrossRef] [PubMed]
18 Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its
receptors. Nat Med 2003;9(6):669-676.
[CrossRef] [PubMed]
19 Bartolotii M, Franceschi E, Poggi R, Tosoni A, Di Battista M, Brandes
AA. Resistance to antiangiogenic therapies. Future
Oncol 2014;10(8):1517-1525. [CrossRef]
20 Soda Y, Myskiw C, Rommel A, Verma IM. Mechanisms of
neovascularization and resistance to anti-angiogenic therapies in glioblastoma
multiforme. J Mol Med 2013;91(4):439-448. [CrossRef] [PMC free article] [PubMed]