Skip to main content

Can intravitreal injections with higher volume cause higher intraocular eye pressure? Considerations for anti-complement injections in normal and glaucomatous eyes

Abstract

The approval of Syfovre® (pegcetacoplan) and Iverzay® (avacincaptad pegol) for the treatment of geographic atrophy (GA) marks a significant advancement in retinal disease therapy, offering both complement 3 and complement 5 inhibitors. With this breakthrough, an increase in intravitreal injections (IVI) is expected to treat GA, raising questions about potential effects on intraocular pressure (IOP). This concern is exacerbated by the larger injection volume required for GA treatment, potentially impacting IOP. Previous studies have shown that IVI can lead to a temporary increase in IOP with a 0.05 ml injection. This transient elevation is challenging to manage with glaucoma drops, and a preventive approach, such as paracentesis immediately before IVIs, may be more effective. Despite concerns, clinical significance and long-term effects of IOP changes with a 0.05 ml injection remain uncertain. To address these concerns, routine evaluations including macular optical coherence tomography (OCT), fundus autofluorescence, IOP measurements, and retinal nerve fiber layer OCT before the first IVI with avacincaptad pegol and pegcetacoplan are recommended to detect potential changes early. Further research is needed to determine the extent to which IOP changes impact GA patients and whether cumulative effects occur with repeated IVIs, especially in those with additional eye conditions.

Background

With the approval of Syfovre® (pegcetacoplan) by the U.S. Food and Drug Administration (FDA) in February 2023 and Iverzay® (avacincaptad pegol) in August 2023 for clinical use in patients with geographic atrophy (GA), it is now possible for the first time to treat the disease with either a complement 3 or a complement 5 inhibitor [1, 2].

Main text

Since GA is a widespread disease, an increasing number of intravitreal injections (IVI) is expected, which must be applied regularly at four to eight-week intervals according to the FDA label. However, the new form of therapy will not only expand our spectrum of treatable retinal diseases and significantly increase the number of injections administered annually, but it will also be administered with twice the volume, which could potentially be associated with a higher increase in intraocular pressure (IOP). In particular, older GA patients with inherently poorer perfusion may repeatedly develop short-term increases in IOP with additional retinal nerve fiber layer (RNFL) loss and glaucoma development/progression during larger volume injections [3]. While vascular disease is usually administered with anti-VEGF drugs with a volume of 0.05 mL, GA is treated with both drugs with a volume of 0.1 mL. Therefore, there are some questions in the community that we would like to explain and discuss from our first clinical experience with these drugs.

Millions of intravitreal injections are administered annually to patients with various retinal diseases. It is known from previous clinical studies that immediately after IVI of 0.05 ml, IOP can increase to 50 mmHg [4]. This temporary steep peek of IOP can only be inadequately cushioned by the administration of glaucoma drops, as their pharmacological effect starts slow and cannot prevent a rapid sudden increase. The quickest approach to prevent a postoperative ad hoc increase in IOP is to use paracentesis immediately before an IVI [5]. As early as 2018, a report by the American Academy of Ophthalmology evaluated the temporary increased IOP and possible consequences on the optic nerve. However, their clinical significance and long-term effects remained unknown for an applied volume of 0.05 ml [6]. After unilaterally repeated IVIs, a significant decrease in nerve fiber layer thickness after 2 years was documented in a bilateral comparison [7]. A systematic review showed a 2-fold increase in the risk of sustained IOP elevation in eyes undergoing repeated IVI. The authors also found that the longer the follow-up duration, the higher the risk ratio for this elevation [8].

Due to the recent doubling of the injection volume of several novel drugs, there may be a greater increase in IOP. No pathological changes directly attributed to IOP were observed in the pivotal OAKS and DERBY (pegcetacoplan), and GATHER 1 and GATHER 2 (avacincaptad pegol) studies, which monitored a period of up to two years, but optical coherence tomography (OCT) was not performed in these trials to monitor the RNFL [1, 2, 9]. However, experimental studies with similar higher volume showed a consecutively higher increase in IOP [10,11,12,13]. For example, Knip and Välimäki reported an increase in IOP from an average of 14.6 mmHg (SD, 2.4) to 47.1 mmHg (SD, 24.1) without paracentesis just 2 min after IVI of 0.09 ml pegaptanib (Macugen, Pfizer) [10]. Kotliar et al. also reported an increase in IOP of 40.6 mmHg immediately after IVI of 0.1 ml (SD, 12.1) and a reduction to 9.4 mmHg (SD, 4.9) after paracentesis. They also showed in a biomechanical model the exponential increase in IOP values with the reduced length of the globe [11]. Lorenz et al. required an additional paracentesis in 78 (33%) out of 234 consecutive cases after 0.1 ml bevacizumab IVIs to reduce the IOP [12]. Another recent publication showed an estimated IOP rise ranging from 32.3 (SD, 1.4) mmHg for 20-µL injection volume to 76.5 (SD, 1.0) mmHg for 80-µL injection volume in an experimental eye model [13]. For a 50-µL volume, the peak pressure was 50.7 (SD, 0.1) mmHg, and the pressure rise lasted for an average of 28 min (SD, 2).

The clinical benefit-risk ratio of paracentesis prior to intravitreal injection has been discussed repeatedly among experts [14]. However, there is a general consensus that eyes with glaucoma, increased IOP or shortened axial length are potentially more likely to develop higher IOP and chronic-cumulative RNFL damage after multiple consecutive IVI and that paracentesis can therefore be useful on an individual basis. Paracentesis is now generally a safe method of normalizing eye pressure in the event of a sharp increase in eye pressure or acute vision loss. Limited research has been done on the extent to which a temporarily moderate increase in IOP leads to subclinical occult changes in the optic nerve, and whether repeated IVIs individually lead to cumulative effects therefore requires systematic observation of all GA patients with OCT. Physicians should also be aware that patients with neovascular age-related macular degeneration (AMD) might have a reduced RNFL without any IVI [15]. Since some patients with GA may also present with neovascular AMD, some RNFL loss over time may also be found and should not be misconceived.

Finally, we would like to point out that a few cases of ischemic optic neuropathy were found in the pivotal trials OAKS and DERBY with pegcetacoplan [1, 16]. Besides some unanticipated drug-related predisposing factor, a reduced ocular blood flow secondary to an increased IOP should be regarded as a plausible cause. In agreement with this hypothesis, Chen et al. (2019) have shown that a greater number of IVI of antiangiogenic agents leads to increased likelihood of developing ischemic optic neuropathy, and they also attributed it to a higher IOP [17].

Conclusions

We recommend that in addition to macular OCT, fundus autofluorescence (FAF), IOP measurement and RNFL OCT be routinely carried out before the first IVI with either avacincaptad pegol or pegcetacoplan is performed in order to detect possible changes at an early stage after repeated injections. It remains to be seen to what extent these experimental results will play a role in all GA patients or only individual patients with additional diseases of the eye.

Data Availability

Not applicable.

Abbreviations

AMD:

Age-related macular degeneration

FA:

Fundus autofluorescence

FDA:

Food and Drug Administration

GA:

Geographic atrophy

IVI:

Intravitreal injection

IOP:

Intraocular pressure

OCT:

Optical coherence tomography

References

  1. Heier JS, Lad EM, Holz FG, et al. Pegcetacoplan for the treatment of geographic atrophy secondary to age-related macular degeneration (OAKS and DERBY): two multicentre, randomised, double-masked, sham-controlled, phase 3 trials. Lancet. 2023;402:1434–48. https://doi.org/10.1016/S0140-6736(23)01520-9.

    Article  CAS  PubMed  Google Scholar 

  2. Khanani AM, Patel SS, Staurenghi G, Tadayoni R, Danzig CJ, Eichenbaum DA, Hsu J, Wykoff CC, Heier JS, Lally DR, Monés J, Nielsen JS, Sheth VS, Kaiser PK, Clark J, Zhu L, Patel H, Tang J, Desai D, Jaffe GJ. ; GATHER2 trial investigators. Efficacy and safety of avacincaptad pegol in patients with geographic atrophy (GATHER2): 12-month results from a randomised, double-masked, phase 3 trial. Lancet. 2023 Sep 8:S0140-6736(23)01583-0. https://doi.org/10.1016/S0140-6736(23)01583-0.

  3. El Chehab H, Agard E, Russo A, Boujnah Y, Dot C. Intraocular pressure spikes after Aflibercept Intravitreal injections. Ophthalmologica. 2016;236:43–7.

    Article  PubMed  Google Scholar 

  4. Sharei V, Höhn F, Köhler T, Hattenbach LO, Mirshahi A. Course of intraocular pressure after intravitreal injection of 0.05 mL ranibizumab (Lucentis). Eur J Ophthalmol. 2010;20:174–9.

    Article  PubMed  Google Scholar 

  5. Bach A, Filipowicz A, Gold AS, Latiff A, Murray TG. Paracentesis following intravitreal drug injections in maintaining physiologic ocular perfusion pressure. Int J Ophthalmol. 2017;10:1925–7.

    PubMed  PubMed Central  Google Scholar 

  6. Hoguet A, Chen PP, Junk AK, Mruthyunjaya P, et al. The Effect of Anti-vascular endothelial growth factor agents on intraocular pressure and Glaucoma: a report by the American Academy of Ophthalmology. Ophthalmology. 2019;126(4):611–22. https://doi.org/10.1016/j.ophtha.2018.11.019.

    Article  PubMed  Google Scholar 

  7. Enders P, Sitnilska V, Altay L, Schaub F, Muether PS, Fauser S. Retinal nerve fiber loss in anti-VEGF therapy for age-related macular degeneration can be decreased by anterior chamber paracentesis. Ophthalmologica. 2017;237:111–8.

    Article  PubMed  Google Scholar 

  8. Zhou Y, Zhou M, Xia S, Jing Q, Gao L. Sustained Elevation of Intraocular Pressure Associated with Intravitreal Administration of anti-vascular endothelial growth factor: a systematic review and Meta-analysis. Sci Rep. 2016;6:39301. https://doi.org/10.1038/srep39301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Jaffe GJ, Westby K, Csaky KG, Monés J, Pearlman JA, Patel SS, Joondeph BC, Randolph J, Masonson H, Rezaei KA. C5 inhibitor Avacincaptad Pegol for Geographic Atrophy due to age-related Macular Degeneration: a Randomized Pivotal Phase 2/3 Trial. Ophthalmology. 2021;128(4):576–86. https://doi.org/10.1016/j.ophtha.2020.08.027.

    Article  PubMed  Google Scholar 

  10. Knip MM, Välimäki J. Effects of pegaptanib injections on intraocular pressure with and without anterior chamber paracentesis: a prospective study. Acta Ophthalmol. 2012;90:254–8.

    Article  CAS  PubMed  Google Scholar 

  11. Kotliar K, Maier M, Bauer S, Feucht N, Lohmann C, Lanzl I. Effect of intravitreal injections and volume changes on intraocular pressure: clinical results and biomechanical model. Acta Ophthalmol Scand. 2007;85:777–81.

    Article  PubMed  Google Scholar 

  12. Lorenz K, Zwiener I, Mirshahi A. Subconjunctival reflux and need for paracentesis after intravitreal injection of 0.1 ml bevacizumab: comparison between 27-gauge and 30-gauge needle. Graefes Arch Clin Exp Ophthalmol. 2010;248:1573–7.

    Article  PubMed  Google Scholar 

  13. Agra LLM, Sverstad A, Chagas TA, Araújo RH, Oliveira LG, Kristianslund O, Petrovski G, Maia M, Moe MC, Jørstad ØK, Melo GB, ACCURACY, PRECISION, AND RESIDUAL VOLUME OF COMMONLY USED SYRINGES FOR INTRAVITREAL INJECTIONS AND THE IMPACT ON INTRAOCULAR PRESSURE. Ophthalmol Retina. 2023;7:892–900. https://doi.org/10.1016/j.oret.2023.06.003.

    Article  PubMed  Google Scholar 

  14. Saxena S, Lai TY, Koizumi H, Farah ME, Ferrara D et al. For the international pharmacokinetic collaboration. Anterior chamber paracentesis during intravitreal injections in observational trials: effectiveness and safety and effects. Int J Retina Vitreous 2019:5:8. https://doi.org/10.1186/s40942-019-0157-z.

  15. Zucchiatti I, Parodi MB, Pierro L, Cicinelli MV, Gagliardi M, Castellino N, Bandello F. Macular ganglion cell complex and retinal nerve fiber layer comparison in different stages of age-related macular degeneration. Am J Ophthalmol. 2015;160(3):602–607e1. https://doi.org/10.1016/j.ajo.2015.05.030.

    Article  PubMed  Google Scholar 

  16. Antonio-Aguirre B, Arevalo JF. Treating patients with geographic atrophy: are we there yet? Int J Retina Vitreous. 2023;9(1):72. https://doi.org/10.1186/s40942-023-00493-6.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Chen YY, Chou P, Huang YF, Chien HJ, Wu YC, Lee CC, Huang LY, Chen HH. Repeated intravitreal injections of antivascular endothelial growth factor in patients with neovascular age-related macular degeneration may increase the risk of ischemic optic neuropathy. BMC Ophthalmol. 2019;19(1):268. https://doi.org/10.1186/s12886-019-1284-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

All authors wrote part of the manuscript and approved the final version.

Corresponding author

Correspondence to Gustavo Barreto Melo.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

CHM has received fees for advisory board for Apellis and lectures for Bayer. GBM is a scientific consultant to SJJ Solutions and Molecular Partners, and has been part of advisory boards of Bayer, Molecular Partners, and Roche. AMK is a consultant for Apellis and Iveric Bio and receives research grants from Apellis and Iveric Bio.

GBM is editor-in-chief of International Journal of Retina and Vitreous. This article was independently handled by a member of the Editorial Board.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Meyer, C.H., Melo, G.B. & Khanani, A.M. Can intravitreal injections with higher volume cause higher intraocular eye pressure? Considerations for anti-complement injections in normal and glaucomatous eyes. Int J Retin Vitr 9, 80 (2023). https://doi.org/10.1186/s40942-023-00517-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40942-023-00517-1

Keywords