bannter.jpg
·
Clinical Research·

 

Effect of different lens status on intraocular pressure elevation in patients treated with anti-vascular endothelial growth factor injections

 

Amir Sternfeld1,2, Rita Ehrlich1,2, Dov Weinberger1,2, Assaf Dotan1,2

 

1Department of Ophthalmology, Rabin Medical Center Beilinson Hospital, Petach Tikva 4941402, Israel

2Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 6997801, Israel

Correspondence to: Assaf Dotan. Department of Ophthalmology, Rabin Medical Center-Beilinson Hospital, Petach Tikva 4941492, Israel. docdotan@gmail.com

Received: 2018-11-08        Accepted: 2019-07-11

 

Abstract

AIM: To assess the effect of lens status on sustained intraocular pressure (IOP) elevation in patients treated intravitreally with anti-vascular endothelial growth factor (VEGF) agents.

Methods: Data were retrospectively collected for all patients treated with intravitreal injections of anti-VEGF medication at a tertiary medical center in July 2015. Findings were analyzed by lens status during 6 months’ follow-up. The main outcome measure was a sustained increase in IOP (≥21 mm Hg or change of ≥6 mm Hg from baseline on ≥2 consecutive visits, or addition of a new IOP-lowering medication during follow-up).

Results: A total of 119 eyes of 100 patients met the study criteria: 40 phakic, 40 pseudophakic, and 39 pseudophakic after Nd:YAG capsulotomy. The rate of sustained IOP elevation was significantly higher in the post-capsulotomy group (23.1%) than in the phakic/pseudophakic groups (8.1%; p=0.032), with no statistically significant differences among the 3 groups in mean number of injections, either total (p=0.82) or by type of anti-VEGF mediation (bevacizumab: p=0.19; ranibizumab: p=0.13), or mean follow-up time (p=0.70).

Conclusion: Nd:YAG capsulotomy appears to be a risk factor for sustained IOP elevation in patients receiving intravitreal anti-VEGF injections. This finding has important implications given the growing use of anti-VEGF treatment and the irreversible effects of elevated IOP.

Keywords: anti-VEGF injections; cataract surgery; intraocular pressure; Nd:YAG capsulotomy

DOI:10.18240/ijo.2020.01.12

Citation: Sternfeld A, Ehrlich R, Weinberger D, Dotan A. Effect of different lens status on intraocular pressure elevation in patients treated with anti-vascular endothelial growth factor injections. Int J Ophthalmol 2020;13(1):79-84

 

Introduction

The overexpression of vascular endothelial growth factor (VEGF) has been identified as a cornerstone of the pathologic process leading to vascular leakage and neovascularization. Accordingly, intravitreal injection of anti-VEGF inhibitors is the main mode of treatment for exudative age-related macular degeneration and other retinal vascular diseases characterized by macular edema and neovascularization including diabetic macular edema and retinal vein occlusion[1-3]. Several anti-VEGF agents are currently available: bevacizumab (Avastin; Genentech, Inc., South San Francisco, CA, USA), a recombinant full-length humanized monoclonal antibody; ranibizumab (Lucentis; Genentech), a humanized monoclonal antibody fragment; and aflibercept (Eylea; Regeneron Pharmaceuticals, Tarrytown, NY, USA), a soluble decoy receptor fusion protein[4-6]. The drugs have a favorable safety profile, with only rare reports of adverse vision-threatening events such as endophthalmitis, vitreous hemorrhage, retinal detachment, and retinal tears, despite their widespread use[7].

A known immediate effect of any intravitreal injection is a transient elevation of intraocular pressure (IOP). In the majority of cases, the spikes normalize within 30min without further intervention[8-9]. However, the occurrence of sustained ocular hypertension in this setting is controversial, with only sparse reports in the recent literature. Both the MARINA trial (anti-VEGF antibody ranibizumab in the treatment of neovascular age-related macular degeneration) and the ANCHOR Trial (ranibizumab versus verteporfin for neovascular age-related macular degeneration), which investigated the effect of ranibizumab treatment in patients with minimal classic or occult neovascularization, reported an absence of long-term effects of monthly injections on mean IOP[10]. Similar findings were noted in a phase III study on the use of aflibercept for neovascular age-related macular degeneration[6]. Wehrli et al[11] conducted a large retrospective study of 302 eyes treated with bevacizumab and/or ranibizumab, with 226 untreated fellow eyes serving as the control group. No between-group difference was noted in the rate of delayed ocular hypertension.

By contrast, several reports in recent years have provided evidence that IOP elevation may be a long-term adverse effect of anti-VEGF drugs[12-15]. Among them is a post-hoc data analysis of the MARINA and ANCHOR trials comparing the anti-VEGF-treated eyes with eyes treated with sham injections or photodynamic therapy. The authors found that the study group had a statistically significant increase in the number of eyes with an elevation of 6 mm Hg or more in IOP from baseline or an IOP equal to or greater than 25 mm Hg on at least 2 consecutive visits[16].

The risk factors contributing to the sustained IOP elevation remain unclear owing to differences among the studies in the number of injections[11,13-15] and type of anti-VEGF agent used[13-14], and the inclusion of patients with pre-existing glaucoma[14-15,17] or prior use of topical or intravitreal steroids[12,15,17].

Besides the medication, there is evidence pointing to the contribution of the cataract extraction itself[18], alone or with the performance of neodymium doped yttrium aluminum garnet (Nd:YAG) capsulotomy[19], and Nd:YAG capsulotomy itself[20]. The present study sought to shed further light on this issue. The aim of the study was to assess the effect of lens status, in particular after Nd:YAG capsulotomy, on sustained IOP elevation.


SUBJECTS AND Methods

Ethical Approval  The study adhered to the tenets of the Declaration of Helsinki and was approved by the institutional review board of Rabin Medical Center. Informed consent was waived due to the retrospective nature of the study.

The database of the Retina Clinic of Rabin Medical Center, a tertiary university-affiliated hospital, was retrospectively reviewed for all patients with retinal vascular disease who were treated with an intravitreal injection of anti-VEGF agent in July 2015. It is our departmental policy in these cases to start with bevacizumab (1.25 mg/0.05 mL) and switch to ranibizumab (0.5 mg/0.05 mL) on the basis of the patient response. Therefore, none of the patients received only ranibizumab. We included only patients for whom the pre-injection (baseline) IOP value and all IOP values during a minimum of 6 months’ follow-up from the first injection were available. Exclusion criteria were a change in lens status during follow-up (cataract surgery or Nd:YAG capsulotomy), intraocular lens implantation outside the bag, receipt of intravitreal corticosteroid treatment during follow-up, and any intraocular surgery other than cataract surgery prior to or during follow-up, with the exception of  glaucoma surgery (trabeculectomy, glaucoma drainage devices or laser trabeculoplasty) during follow-up.

The following data were collected from the medical files: patient age and sex, reason for treatment, number of anti-VEGF injections, type of injection, follow-up time (defined as the time from the first injection until the last follow-up), lens status prior to and throughout treatment, IOP prior to treatment and at every follow-up visit, diagnosis of glaucoma, and medications for glaucoma before anti-VEGF injection and throughout follow-up.

IOP was measured by Goldmann applanation tonometry. Measurements were performed before every injection and at every office visit.

The main outcome measure was a sustained increase in IOP, defined as IOP≥21 mm Hg or a change from baseline of ≥6 mm Hg, recorded on at least 2 consecutive visits, or the addition of a new IOP-lowering medication after the start of anti-VEGF treatment for 45d or more, during follow-up. These criteria were selected to rule out normal IOP fluctuations and represent a clinically significant IOP elevation.

Continuous variables are presented as means and standard deviation, and nominal variables, as number and percentage. Continuous data were checked for normality (Shapiro-Wilk test). One-way analysis of variance (ANOVA) with Bonferroni post hoc test or the Kruskal-Wallis nonparametric test was used to compare variables by lens status. Unpaired Student’s t-test or the nonparametric Mann-Whitney test was used to compare variables by the presence or absence of sustained IOP elevation. A p value of <0.05 was considered statistically significant. All statistical analyses were performed using the SPSS v. 3.


Results

A total of 119 eyes of 100 patients met the study criteria. The patient characteristics and clinical data are shown in Table 1.

Table 1 Baseline demographic and clinical characteristics of patients treated with anti-VEGF agents, by lens status    n (%)

Characteristics

Phakic eyes

Pseudophakic eyes

Post-capsulotomy eyes

P

No. of eyes

40

40

39

 

Males

21 (52.5)

8 (20)

14 (35.9)

0.01a

Age (y)

73.2±14.5

82.6±7.9

82.9±8.5

<0.001a

Laterality (right eye)

18 (45)

15 (37.5)

19 (48.7)

0.59

Diagnosis

 

  Exudative AMD

25 (62.5)

31 (77.5)

27 (69.2)

0.13

  Diabetic macular edema

13 (32.5)

6 (15)

5 (12.8)

  Othersb

2 (5)

3 (7.5)

7 (18)

Number of injections

 

  Bevacizumab

10.2±3.5

9.9±4.0

8.7±4.3

0.19

  Ranibizumab

2.0±3.1

1.8±3.1

3.3±4.3

0.13

  Total

12.2±3.2

11.7±3.7

11.9±4.5

0.82

Follow-up (d)

701.3±80.1

710.4±100.0

735.7±301.9

0.70

Pretreatment glaucoma

3 (7.5)

1 (2.5)

5 (12.8)

0.22

AMD: Age-related macular degeneration. aStatistically significant (post-capsulotomy vs phakic/pseudophakic); bOthers indication for anti-VEGF treatment included myopic choroidal neovascularization, peripapillary choroidal neovascularization, central retinal vein occlusion and branch retinal vein occlusion.

 

Division by lens status during follow-up yielded 3 groups: 40 phakic eyes, 40 pseudophakic eyes, and 39 pseudophakic eyes following Nd:YAG capsulotomy. There was no significant difference among the groups in the rate of pre-treatment glaucoma (phakic, 7.5%; pseudophakic, 2.5%; post-capsulotomy, 12.8%; p=0.22) or mean pre-treatment IOP [14.6±2.3 mm Hg (range 10-20), 14.2±2.1 mm Hg (10-18) and 14.3±2.6 mm Hg (10-22), respectively; p=0.76]. All patients with pre-treatment glaucoma had primary open angle glaucoma, were followed by an ophthalmologist for their glaucoma prior to their retinal disease and were at their target pressures under topical treatment (2.11±1.27 medications). There was also no difference among the groups in the diseases for which they were treated (p=0.13). Patients with phakic eyes were significantly younger than patients with pseudophakic eyes (73.2±14.5, 82.6±7.9, and 82.9±8.5y, respectively; p<0.001).

The association of lens status with IOP elevation is shown in Table 2.

Table 2 Sustained IOP elevation criteria by study groups                                         n (%)

Criterion

Phakic eyes

Pseudophakic eyes

Post-capsulotomy eyes

p

6 mm Hg increase on 2 consecutive visits

3 (7.5)

3 (7.5)

5 (12.8)

0.64

 

6 (7.5)

5 (12.8)

0.35

21 mm Hg on 2 consecutive visits

0

0

0

 

New IOP lowering medication

0

2 (5)

4 (10.3)

0.11

 

2 (2.5)

4 (10.3)

0.09

Total

3 (7.5)

4 (10)

9 (23.1)

0.09

 

7 (8.8)

9 (23.1)

0.03a

IOP: Intraocular pressure. aP<0.05.

 

The overall rate of sustained IOP elevation was 13.4%. The rate was significantly higher in the post-capsulotomy group than in the phakic+pseudophakic groups (23.1% vs 8.8%; p=0.03). There were no statistically significant differences among the 3 groups (Table 1) in mean number of injections, either total (phakic: 12.2±3.2; pseudophakic: 11.7±3.7; post-capsulotomy: 11.9±4.5; p=0.82) or by type of anti-VEGF medication (bevacizumab, 10.2±3.5, 9.9±4.0 and 8.7±4.3, respectively, p=0.19; ranibizumab, 2.0±3.1, 1.8±3.1 and 3.3±4.3, respectively, p=0.13). Mean follow-up time was similar in all 3 groups (701.3±80.1, 710.4±100.0, and 735.7±301.9d, respectively; p=0.70) as was the interval between the beginning of treatment and IOP elevation (247.0±158.0, 251.0±11.7 and 445.1±259.1, P=0.23). IOP in the three groups ranged between 8-23 mm Hg in the phakic group, 10-30 mm Hg in the pseudophakic group and 10-28 mm Hg in the post-capsulotomy group.

Sustained IOP elevation rate remained significantly higher in the post-capsulotomy group following further analysis of the data using multivariable logistic regression that included total number on injections and pre-existing glaucoma (P=0.047). When separately analyzing bevacizumab and ranibizumab, significance remained with the former while there was a trend toward significance with the latter (P=0.045 and 0.073, respectively).

Two patients with pre-treatment glaucoma had sustained IOP elevation (12.5% of all patients with sustained IOP elevation and 22.2% of patients with pre-treatment glaucoma), both were in the post-capsulotomy group. Their initial IOP elevation occurred 10 and 13mo after beginning treatments and after receiving 9 and 10 injections, respectively. Their IOP was stable until that point and restabilized after adding topical treatments (two and one IOP lowering drops, respectively).

Comparison of the patients with and without a sustained IOP elevation (Table 3) revealed that the group with a consistently high IOP had a significantly lower baseline IOP (13.3 mm Hg vs 14.5 mm Hg, p=0.04). However, there were no between-group differences in the number of injections, either total (11.8±3.9 vs 12.9±3.4; p=0.3) or by type of medication (bevacizumab: 9.1±4.1 vs 9.7±4.0; p=0.56; ranibizumab: 3.8±4.5 vs 2.1±3.4; p=0.16), or in mean follow-up time (778.8±170.0 vs 705.8±188.4d; p=0.15).

Table 3 Comparison between patients with sustained IOP elevation and patients without sustained IOP elevation     n (%)

Characteristics

Sustained IOP elevation

No IOP elevation

p

Eyes

16

103

 

Males

6 (37.5)

37 (35.9)

0.90

Age (y)

81.3±7.6

79.3±12.1

0.53

No. of injections

 

  Bevacizumab

9.1±4.1

9.7±4.0

0.56

  Ranibizumab

3.8±4.5

2.1±3.4

0.16

  Total

11.8±3.9

12.9±3.4

0.30

Baseline IOP (mm Hg)

13.3±2.7

14.5±2.2

0.04a

Diagnosis

 

  Exudative AMD

10 (62.5)

73 (70.9)

0.46

  Diabetic macular edema

3 (18.8)

21 (20.4)

  Others

3 (18.8)

9 (8.7)

Follow-up (d)

778.8±170.0

705.8±188.4

0.15

Pretreatment glaucoma

2 (12.5)

7 (6.8)

0.42

AMD: Age-related macular degeneration; IOP: Intraocular pressure. aP<0.05.

 

Discussion

The present study suggests that Nd:YAG capsulotomy is a risk factor for sustained IOP elevation in patients treated with anti-VEGF injections. This finding in important given the increasing use of anti-VEGF injections and the potentially irreversible damage caused by elevated IOP.

Several leading theories have been proposed to explain the mechanism underlying long-term IOP elevation after anti-VEGF injection. Some authors suggested that microparticles from the medication’s packaging or delivery equipment may obstruct the trabecular meshwork. This assumption is based on reports of a different aggregate high-molecular-weight protein concentration in repackaged samples of bevacizumab and a higher prevalence of sustained IOP elevations in patients attending centers using repackaged bevacizumab than in patients treated in centers receiving bevacizumab in its original package[14,19].

It is also possible that the high-molecular-weight drugs themselves, especially bevacizumab (MW 150 kDa; ranibizumab, 48 kDa), obstruct the outflow channels. Support for this assumption was provided by findings of a higher prevalence of sustained elevated IOP in studies of patients receiving bevacizumab[12]. Moreover, in a recent experimental study, bevacizumab was found in the trabecular meshwork and Schlemm’s canal after injection into a rat model[21].

A third theory suggests that recurrent episodes of transient post-injection IOP elevation chronically damage the aqueous outflow channels, eventually causing sustained IOP elevation[22]. Alternatively, inflammation, whether recurrent inflammation, post- injection subclinical inflammation, or chronic drug-induced trabeculitis or uveitis, may induce scar formation and fibroblast proliferation which gradually obstruct aqueous outflow[9,23].

Although cataract surgery is generally thought to lower IOP to some degree[18], in the setting of intraocular anti-VEGF injections, lens extraction and, especially, opening of the posterior capsule during Nd:YAG capsulotomy, may promote the introduction of the injected proteins and molecules into the trabecular meshwork,  thereby increasing IOP[19]. Supporting evidence to this theory could be drawn from several animal studies showing increased clearance of bevacizumab and ranibizumab after lensectomy, vitrectomy or both. The improved clearance is attributed, at least in part and specifically in the aphakic eyes, to increased role of the trabecular meshwork[24-26]. It is possible that posterior capsulotomy increases the evacuation through the trabecular meshwork in a similar manner, which in turn increases the risk for sustained elevated IOP. Our finding of higher prevalence of increased IOP in patients after Nd:YAG capsulotomy supports this assumption. Interestingly, if increased clearance does exist after posterior capsulotomy, this may be an indication for more frequent anti-VEGF injections in these patients. Further studies are needed to shed light on this matter. Two previous studies focused on the effect of lens status on sustained IOP elevation. In the first, Hoang et al[15] reported no association between these factors. However, they included only the final lens status at the last injection in the analysis. In a subsequent study, the same group found, in accordance with our results, that phakic lenses are apparently protective against sustained IOP elevation. Both of the earlier studies, however, by contrast to ours, included only a minimal number of patients who had undergone Nd:YAG capsulotomy (3.9% and 6%, respectively).

The 13.4% total incidence of a sustained increase in IOP in the present study was higher than reported by Bakri et al[16] on post hoc analysis of the ANCHOR and MARINA trials (5.4% for 0.3 mg ranibizumab and 4.5% for 0.5 mg ranibizumab), and higher than in the studies of Hoang et al[15,17] (7.2% and 11.6%), and Good et al[14] (6%). The discrepancy may be attributable to our high percentage of post-capsulotomy patients. The high number of bevacizumab injections may also have played a role, as bevacizumab has been found to be a risk factor for sustained IOP elevation[14]. Bevacizumab was not used at all in the ANCHOR and MARINA trials, and was used in only a relatively low percentage of eyes in the studies of Hoang et al[15,17] (34.1% and 32.4%) and Good et al[14] (55.3%). The incidence of increased IOP in the post-capsulotomy group in our study is higher than attributed to the procedure in the literature (23.1% vs 1%-6%, respectively)[20,27-28], possibly related to the role of anti-VEGF injections in this pathology.

The present study did not identify other major risk factors for sustained IOP elevation. Hoang et al[15,17] found total number of anti-VEGF injections to be a risk factor, but their patients received a much higher mean number of injections (20.8 and 21.1, respectively) than ours (11.9). Good et al[14] noted an association of pre-existing glaucoma with a high frequency of sustained IOP elevation (33%). In our study, the number of patients with pre-existing glaucoma (n=9) may have been too small to achieve statistical significance, although a relatively high percentage of the patients with sustained IOP elevation had a pretreatment diagnosis of glaucoma (12.5%). This is certainly an important direction for future research. The lack of a difference in the rate of IOP elevation by type of anti-VEGF agent in our study may be explained by our departmental policy of starting all patients on bevacizumab such that none received only ranibizumab, making it difficult to differentiate the individual effect of the two drugs.

The fact that capsulotomy did not remain a significant risk factor following multivariable analysis using number of ranibizumab injections could result from the smaller size of the molecule. Potentially this enables easier diffusion of the molecule to the anterior chamber even without the opening in the posterior capsule and therefore eliminates its effect. 

The patients with a sustained IOP elevation had a lower baseline IOP (13.3 mm Hg) than the other patients (14.5 mm Hg; p=0.04). This finding, although statistically significant, does not seem to have any clinical importance owing to the small difference between the groups. Patients with phakic eyes were significantly younger than patients with pseudophakic eyes, in line with the progression of cataract over the years and the need for cataract-removal surgery at older age.

This study has several limitations. Our sample size was limited, future prospective multi-center studies are needed for the evaluation of this important subject.

Excluding patients whose baseline or follow-up IOP measurements were not available could have created a selection bias assuming these data would not be missing in patients at risk for increased IOP. This may explain the relatively high percentage of patients with pre-existing glaucoma found to have sustained IOP elevations. However, since it is our policy to always measure and document the IOPs, this phenomenon is probably marginal resulting from inattention and was spread evenly between the groups.

As a result of the retrospective nature of this study, the effect of the time frame between the cataract surgery, Nd:YAG capsulotomy and IOP elevation, or Nd:YAG capsulotomy specifications (energy, spot size, number of shots) and IOP elevation could not be assessed. This information is not detailed in most patients’ charts. Evaluating these data in future studies could add valuable information.

Another drawback was the higher prevalence of pretreatment glaucoma in the post capsulotomy group. This could have potentially confounded the outcomes of the study, especially since 12.5% of the sustained IOP elevation group did have pre-existing glaucoma. However, since these differences did not reach statistical significance, neither in the 3 study groups nor between the patients with and without sustained IOP elevation, and since the absolute numbers and overall prevalence of pretreatment glaucoma patients were not high, we believe their effect on the results was minimal at most. The effect of pre-existing glaucoma combined with lens status could also be an interesting direction for research.

In conclusion, this study suggests that Nd:YAG capsulotomy is a risk factor for a sustained elevation in IOP in patients treated with anti-VEGF injections. This finding may have clinical implications when capsulotomy is being considered in patients under anti-VEGF treatment. It supports the recommendation for routine monitoring of IOP levels in all patients treated with anti-VEGF drugs, and particularly, patients after capsulotomy.


ACKNOWLEDGEMENTS

Authors’ contributions: Sternfeld A, Dotan A: substantial contributions to conception and design, acquisition of data, analysis and interpretation of data. Drafting the article or revising it critically for important intellectual content. Final approval of the version to be published. Ehrlich R, Weinberger D: contributions to conception and design, acquisition of data, analysis and interpretation of data. Drafting the article or revising it critically for important intellectual content. Final approval of the version to be published.

Conflicts of Interest: Sternfeld A, None; Ehrlich R, None; Weinberger D, None; Dotan A, None.


References

1 Ciulla TA, Bracha P, Pollack J, Williams DF. Real-world outcomes of anti-vascular endothelial growth factor therapy in diabetic macular edema in the United States. Ophthalmol Retina 2018;2(12): 1179-1187.
https://doi.org/10.1016/j.oret.2018.06.004
PMid:31047187

 

2 Adamis AP, Shima DT. The role of vascular endothelial growth factor in ocular health and disease. Retina 2005;25(2):111-118.
https://doi.org/10.1097/00006982-200502000-00001
PMid:15689799

 

3 Bressler SB, Odia I, Glassman AR, Danis RP, Grover S, Hampton GR, Jampol LM, Maguire MG, Melia M. Changes in diabetic retinopathy severity when treating diabetic macular edema with ranibizumab: DRCR.net protocol I 5-year report. Retina 2018;38(10):1896-1904.
https://doi.org/10.1097/IAE.0000000000002302
PMid:30234859 PMCid:PMC6547828

 

4 Cai S, Bressler NM. Aflibercept, bevacizumab or ranibizumab for diabetic macular oedema: recent clinically relevant findings from DRCR.net Protocol T. Curr Opin Ophthalmol 2017;28(6):636-643.
https://doi.org/10.1097/ICU.0000000000000424
PMid:28837425

 

5 Pershing S, Talwar N, Armenti ST, Grubbs J Jr, Rosenthal JM, Dedania VS, Stein JD. Use of bevacizumab and ranibizumab for wet age-related macular degeneration: influence of CATT results and introduction of aflibercept. Am J Ophthalmol 2019;207:385-394.
https://doi.org/10.1016/j.ajo.2019.05.011
PMid:31100217

 

6 Khurana RN, Chang LK, Bansal AS, Palmer JD, Wu C, Wieland MR. Aflibercept for previously treated macular edema associated with central retinal vein occlusions: 1-year results of the NEWTON study. Ophthalmol Retina 2018;2(2):128-133.
https://doi.org/10.1016/j.oret.2017.05.017
PMid:31047339

 

7 Rich RM, Rosenfeld PJ, Puliafito CA, Dubovy SR, Davis JL, Flynn HW Jr, Gonzalez S, Feuer WJ, Lin RC, Lalwani GA, Nguyen JK, Kumar G. Short-term safety and efficacy of intravitreal bevacizumab (Avastin) for neovascular age-related macular degeneration. Retina 2006;26(5):495-511.
https://doi.org/10.1097/01.iae.0000225766.75009.3a
PMid:16770255

 

8 Farhood QK, Twfeeq SM. Short-term intraocular pressure changes after intravitreal injection of bevacizumab in diabetic retinopathy patients. Clin Ophthalmol 2014;8:599-604.
https://doi.org/10.2147/OPTH.S58413
PMid:24707164 PMCid:PMC3971946

 

9 Hollands H, Wong J, Bruen R, Campbell RJ, Sharma S, Gale J. Short-term intraocular pressure changes after intravitreal injection of bevacizumab. Can J Ophthalmol 2007;42(6):807-811.
https://doi.org/10.3129/i07-172
PMid:18026202

 

10 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.
https://doi.org/10.1056/NEJMoa062655
PMid:17021319

 

11 Wehrli SJ, Tawse K, Levin MH, Zaidi A, Pistilli M, Brucker AJ. A lack of delayed intraocular pressure elevation in patients treated with intravitreal injection of bevacizumab and ranibizumab. Retina 2012;32(7):1295-1301.
https://doi.org/10.1097/IAE.0b013e31823f0c95
PMid:22466465

 

12 Adelman RA, Zheng Q, Mayer HR. Persistent ocular hypertension following intravitreal bevacizumab and ranibizumab injections. J Ocul Pharmacol Ther 2010;26(1):105-110.
https://doi.org/10.1089/jop.2009.0076
PMid:20187807

 

13 Choi DY, Ortube MC, McCannel CA, Sarraf D, Hubschman JP, McCannel TA, Gorin MB. Sustained elevated intraocular pressures after intravitreal injection of bevacizumab, ranibizumab, and pegaptanib. Retina 2011;31(6):1028-1035.
https://doi.org/10.1097/IAE.0b013e318217ffde
PMid:21836409

 

14 Good TJ, Kimura AE, Mandava N, Kahook MY. Sustained elevation of intraocular pressure after intravitreal injections of anti-VEGF agents. Br J Ophthalmol 2011;95(8):1111-1114.
https://doi.org/10.1136/bjo.2010.180729
PMid:20702430

 

15 Hoang QV, Mendonca LS, Della Torre KE, Jung JJ, Tsuang AJ, Freund KB. Effect on intraocular pressure in patients receiving unilateral intravitreal anti-vascular endothelial growth factor injections. Ophthalmology 2012;119(2):321-326.
https://doi.org/10.1016/j.ophtha.2011.08.011
PMid:22054994

 

16 Bakri SJ, Moshfeghi DM, Francom S, Rundle AC, Reshef DS, Lee PP, Schaeffer C, Rubio RG, Lai P. Intraocular pressure in eyes receiving monthly ranibizumab in 2 pivotal age-related macular degeneration clinical trials. Ophthalmology 2014;121(5):1102-1108.
https://doi.org/10.1016/j.ophtha.2013.11.029
PMid:24393349

 

17 Hoang QV, Tsuang AJ, Gelman R, Mendonca LS, Della Torre KE, Jung JJ, Freund KB. Clinical predictors of sustained intraocular pressure elevation due to intravitreal anti-vascular endothelial growth factor therapy. Retina 2013;33(1):179-187.
https://doi.org/10.1097/IAE.0b013e318261a6f7
PMid:22990314

 

18 Melancia D, Abegão Pinto L, Marques-Neves C. Cataract surgery and intraocular pressure. Ophthalmic Res 2015;53(3):141-148.
https://doi.org/10.1159/000377635
PMid:25765255

 

19 Kahook MY, Liu L, Ruzycki P, Mandava N, Carpenter JF, Petrash JM, Ammar DA. High-molecular-weight aggregates in repackaged bevacizumab. Retina 2010;30(6):887-892.
https://doi.org/10.1097/IAE.0b013e3181d50cea
PMid:20458261

 

20 Ge J, Wand M, Chiang R, Paranhos A, Shields MB. Long-term effect of Nd: YAG laser posterior capsulotomy on intraocular pressure. Arch Ophthalmol 2000;118(10):1334-1337.
https://doi.org/10.1001/archopht.118.10.1334
PMid:11030814

 

21 Gal-Or O, Dotan A, Dachbash M, Tal K, Nisgav Y, Weinberger D, Ehrlich R, Livnat T. Bevacizumab clearance through the iridocorneal angle following intravitreal injection in a rat model. Exp Eye Res 2016;145:412-416.
https://doi.org/10.1016/j.exer.2016.02.006
PMid:26923799

 

22 Dedania VS, Bakri SJ. Sustained elevation of intraocular pressure after intravitreal anti-VEGF agents: what is the evidence? Retina 2015;35(5):841-858.
https://doi.org/10.1097/IAE.0000000000000520
PMid:25905784

 

23 Menke MN, Salam A, Framme C, Wolf S. Long-term intraocular pressure changes in patients with neovascular age-related macular degeneration treated with ranibizumab. Ophthalmologica 2013;229(3): 168-172.
https://doi.org/10.1159/000346397
PMid:23548723

 

24 Christoforidis JB, Williams MM, Wang J, Jiang A, Pratt C, Abdel-Rasoul M, Hinkle GH, Knopp MV. Anatomic and pharmacokinetic properties of intravitreal bevacizumab and ranibizumab after vitrectomy and lensectomy. Retina 2013;33(5):946-952.
https://doi.org/10.1097/IAE.0b013e3182753b12
PMid:23407351 PMCid:PMC4086838

 

25 Niwa Y, Kakinoki M, Sawada T, Wang X, Ohji M. Ranibizumab and aflibercept: intraocular pharmacokinetics and their effects on aqueous VEGF level in vitrectomized and nonvitrectomized macaque eyes. Invest Ophthalmol Vis Sci 2015;56(11):6501-6505.
https://doi.org/10.1167/iovs.15-17279
PMid:26447985

 

26 Edington M, Connolly J, Chong NV. Pharmacokinetics of intravitreal anti-VEGF drugs in vitrectomized versus non-vitrectomized eyes. Expert Opin Drug Metab Toxicol 2017;13(12):1217-1224.
https://doi.org/10.1080/17425255.2017.1404987
PMid:29134820

 

27 Jahn CE, Emke M. Long-term elevation of intraocular pressure after neodymium: YAG laser posterior capsulotomy. Ophthalmologica 1996;210(2):85-89.
https://doi.org/10.1159/000310679
PMid:9148259

 

28 Lin JC, Katz LJ, Spaeth GL, Klancnik JM Jr. Intraocular pressure control after Nd: YAG laser posterior capsulotomy in eyes with glaucoma. Br J Ophthalmol 2008;92(3):337-339.
https://doi.org/10.1136/bjo.2007.125310
PMid:18211944