Next Article in Journal
Beyond the Vestibulo-Ocular Reflex: Vestibular Input is Processed Centrally to Achieve Visual Stability
Next Article in Special Issue
Image Stabilization in Central Vision Loss: The Horizontal Vestibulo-Ocular Reflex
Previous Article in Journal
Can Contrast-Response Functions Indicate Visual Processing Levels?
Previous Article in Special Issue
A Retrospective Analysis of the Effect of Subretinal Hyper-Reflective Material and Other Morphological Features of Neovascular Age-Related Macular Degeneration on Visual Acuity Outcomes in Eyes Treated with Intravitreal Aflibercept over One Year
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Therapies of Neovascular AMD beyond Anti-VEGF Injections

by
Greggory M. Gahn
1 and
Arshad M. Khanani
2,*
1
University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
2
Sierra Eye Associates, Reno, NV 89502, USA
*
Author to whom correspondence should be addressed.
Vision 2018, 2(1), 15; https://doi.org/10.3390/vision2010015
Submission received: 22 January 2018 / Revised: 4 February 2018 / Accepted: 12 March 2018 / Published: 19 March 2018
(This article belongs to the Special Issue Age-Related Macular Degeneration)

Abstract

:
Neovascular age-related macular degeneration is a leading cause of vision loss among the aging population. The current standard of care to treat neovascular age-related macular degeneration is inhibiting vascular endothelial growth factor (VEGF) through intravitreal injections. Recent studies have demonstrated that the tyrosine kinase with immunoglobulin-like and epidermal growth factor-like domains 2 (Tie2) pathway also plays a critical role in angiogenesis and vascular stability. Additionally, newly developed treatment delivery systems have been designed to greatly reduce the frequency of injections. In targeting the Tie2 pathway and utilizing a sustained release delivery system, patients may experience improved visual outcomes and a reduced burden of treatment.

1. Introduction

With a predicted worldwide prevalence of 288 million adults by 2040, age-related macular degeneration (AMD) is one of the leading causes of vision loss among individuals over 50 years old [1,2]. Recent advances in anti-vascular endothelial growth factor (anti-VEGF) agents have revolutionized the management of neovascular (wet) AMD, allowing patients to improve short-term visual acuity while slowing the progression of vision loss in the long-term. Despite the tremendous success of anti-VEGF therapy, patients are experiencing an increased burden of treatment due to frequent injections and multiple office visits to assess treatment response. This can be demanding for patients and ultimately result in reduced patient adherence and an inconsistent level of treatment [3]. Additionally, numerous retrospective studies have reported sub-optimal responder rates for anti-VEGF agents to be as high as 10–15% [4,5]. It has been hypothesized that this high rate of sub-responders is largely due to angiogenic factors and pathways other than VEGF playing a role in the disease progression. Long-term studies evaluating the efficacy of anti-VEGF agents have demonstrated that initial improvements in vision over the first two years are not always sustained over longer periods of time [6]. This may be the result of under treatment in the real world setting or potentially the development of macular atrophy in these patients.
Emerging treatment modalities for neovascular AMD aim to overcome the shortcomings of current agents by providing a more predictable improvement of vision while reducing the frequency of injections that are needed. Among these modalities are emerging agents that target alternative angiogenic pathways, and delivery systems that allow a constant and sustained level of treatment.

2. The Tie2 Pathway

Located on vascular endothelial cells, tyrosine kinase with immunoglobulin-like and epidermal growth factor-like domains 2 (Tie2) is a trans membrane receptor that serves as a binding site for the angiopoietin family of ligands that includes angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2). Ang1 is an endogenous full agonist of Tie2 that is produced in pericytes surrounding the retinal vasculature. Once bound to Tie2, Ang1 functions to phosphorylate the receptor and, thereby, activate downstream pathways that ultimately suppress vascular permeability and maintain vascular stability [7]. Conversely, Ang2 is an endogenous weak, partial agonist of Tie2 that competes with Ang1 to suppress phosphorylation and the activation of Tie2 [8,9]. Preclinical studies have shown that embryonic mice that are deficient in the Tie2 receptor die by embryonic day 10.5 due to incorrect vessel organization and failure of vessels to mature [10,11]. Additionally, mice deficient in Ang1 result in a phenotype that is similar to that of Tie2-deficient mice, while overexpression of Ang2 in mice results in a phenotype reminiscent of the Tie2 and Ang1-deficient mice, demonstrating the opposing activities of Ang1 and Ang2 [12].
The interaction between the angiopoietin family and VEGF has been studied in the ocular setting using mice that co-express VEGF-A and ANG-1. When expressed simultaneously with VEGF-A, Ang1 prevented retinal detachment and blocked neovascularization [13]. Despite Ang1 effectively suppressing neovascularization, there was no observed impact on lesions that were previously established [13]. Additionally, early studies using mice have demonstrated that despite increased expression of VEGF in the inner surface of the retina, neovascularization and vascular leakage do not occur unless in the presence of elevated levels of Ang2 [14,15,16,17]. Ang2 has also been further studied for its role in upregulating inflammation via a mechanism of inducing endothelial cells to become more sensitive to the effect of tumor necrosis factor-alpha (TNFα) [18]. In fact, Ang2 is believed to be stored within endothelial cells in Weibel–Palade bodies where it is rapidly released under the influence of angiogenic, inflammatory, and other cytokines [18,19]. Further significance of Ang2 is established from studies that have shown elevated levels of Ang2 and VEGF-A in the vitreous of diabetic patients undergoing a vitrectomy [20]. The levels of Ang2 and VEGF-A were shown to correlate with each other as well as disease severity [20,21]. Thus, Ang2 appears to play a considerable role in both angiogenesis and inflammation, and has become an attractive target for therapeutics aimed at inhibiting angiogenesis [22].

3. Emerging Therapeutics Targeting the Tie2 Pathway

RG7716 (Roche/Genentech) is a bispecific monoclonal antibody that has been designed to bind VEGF-A with one arm and Ang2 with the other arm. Through this mechanism, RG7716 has the ability to target multiple pathways implicated in angiogenesis.
This antibody is currently being studied as an intravitreal therapy in the treatment of neovascular age-related macular degeneration. A phase I clinical study has demonstrated that the antibody is safe and well tolerated, with no unexpected adverse events [23]. This study consisted of two treatment groups: a single ascending dose group and a multiple ascending dose group. In the single ascending dose group, patients received 0.5 mg, 1.5 mg, 3 mg or 6 mg with a study duration of up to 16 weeks. In the multiple ascending group, patient received either 3 mg or 6 mg of the drug with a total of three administrations in monthly intervals for a study duration of 24 weeks. The single ascending dose group documented a median improvement of best-corrected visual acuity of 7 letters, while the multiple ascending group demonstrated a median improvement in visual acuity of 7.5 letters [24,25]. Two phase II studies are underway to assess the efficacy of RG7716 in treating neovascular AMD, including a 76-patient extended-dosing study (STAIRWAY) and a 273-patient randomized, double-masked study (AVENUE) [26,27]. STAIRWAY and AVENUE have an estimated completion time of early 2018 with the data becoming available in the middle of 2018 [26,27]. Together, these studies will provide further clarity into the efficacy and durability of RG7716 in the management of neovascular AMD.

4. Sustained Release: Ranibizumab Port Delivery System

The ranibizumab port delivery system is a refillable implant that is placed beneath the conjunctiva and is designed to provide sustained release of ranibizumab into the vitreous [28,29]. The port delivery system has the potential to greatly reduce the burden of frequent injections. For this reason, the ranibizumab port delivery system may provide physicians with an improved tool in the management of neovascular AMD. In a phase I study, the port delivery system was shown to be well tolerated and demonstrated an improvement in best-corrected visual acuity (BCVA) in treatment-naïve patients with neovascular age-related macular degeneration [28,29]. The ranibizumab port delivery system is currently being evaluated in a randomized, double blind phase II study (LADDER) with a 220 patient enrollment [30]. LADDER will assess the safety and efficacy of ranibizumab delivered via the port delivery system compared to the standard of care intravitreal injections of ranibizumab.

5. Conclusions

Anti-VEGF therapies will likely remain the treatment of choice for neovascular AMD for many years to come; however, emerging therapies that target alternative pathways and the development of new delivery systems show great promise for improving the frequency of injections and visual outcomes. Early clinical studies have shown that targeting the Tie2 pathway and utilizing a sustained release delivery system are both potential therapeutic strategies as we advance beyond traditional anti-VEGF intravitreal injection modalities. The data from ongoing large clinical trials to assess efficacy and safety will show if these treatments will become viable options to treat patients with neovascular AMD in the future.

Conflicts of Interest

Greggory M. Gahn—has no conflicts of interest to report; Arshad M. Khanani: consultant for Genentech/Roche, receives research funding from Genentech/Roche and is a speaker for Genentech.

References

  1. National Eye Institute. Facts About Age-Related Macular Degeneration. Available online: http://www.nei.nih.gov/health/maculardegen/armd_facts.asp (accessed on 9 March 2017).
  2. Wong, W.L.; Su, X.; Li, X.; Cheung, C.M.G.; Klein, R.; Cheng, C.-Y.; Wong, T.Y. Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: A systematic review and meta-analysis. Lancet Glob. Health 2014, 2, e106–e116. [Google Scholar] [CrossRef]
  3. Holz, F.G.; Tadayoni, R.; Beatty, S.; Berger, A.; Cereda, M.G.; Hykin, P.; Staurenghi, G.; Wittrup-Jensed, K.; Altemark, A.; Nilsson, J.; et al. Key drivers of visual acuity gains in neovascular age-related macular degeneration in real life: Findings from the AURA study. Br. J. Ophthalmol. 2016, 100, 1623–1628. [Google Scholar] [CrossRef] [PubMed]
  4. Otsuji, T.; Nagai, Y.; Sho, K.; Tsumura, A.; Koike, N.; Tsuda, M.; Nishimura, T.; Takahashi, K. Initial non-responders to ranibizumab in the treatment of age-related macular degeneration (AMD). Clin. Ophthalmol. 2013, 7, 1487–1490. [Google Scholar] [CrossRef] [PubMed]
  5. Suzuki, M.; Nagai, N.; Izumi-Nagai, K.; Shinoda, H.; Koto, T.; Uchida, A.; Mochimaru, H.; Yuki, K.; Sasaki, M.; Tsubota, K.; et al. Predictive factors for non-response to intravitreal ranibizumab treatment in age-related macular degeneration. Br. J. Ophthalmol. 2014, 98, 1186–1191. [Google Scholar] [CrossRef] [PubMed]
  6. Rofagha, S.; Bhisitkul, R.B.; Boyer, D.S.; Radda, S.R.; Zhang, K.; SEVEN-UP Study Group. Seven-Year Outcomes in Ranibizumab-Treated Patients in ANCHOR, MARINA, and HORIZON. Ophthalmology 2013, 120, 2292–2299. [Google Scholar] [CrossRef] [PubMed]
  7. Davis, S.; Aldrich, T.H.; Jones, P.F.; Acheson, A.; Compton, D.L.; Jain, V.; Ryan, T.E.; Bruno, J.; Radziejewski, C.; Maisonpierre, P.C.; et al. Isolation of angiopoietin-1, a ligand for the TIE2 receptor, by secretion-trap expression cloning. Cell 1996, 87, 1161–1169. [Google Scholar] [CrossRef]
  8. Maisonpierre, P.C.; Suri, C.; Jones, P.F.; Bartunkova, S.; Wiegan, S.T.; Radziejewski, C.; Compton, D.; McClain, J.; Aldrich, T.H.; Rapadopoulos, N.; et al. Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science 1997, 277, 55–60. [Google Scholar] [CrossRef] [PubMed]
  9. Yuan, H.T.; Khankin, E.V.; Karumanchi, S.A.; Parikh, S.M. Angio-poietin 2 is a partial agonist/antagonist of Tie2 signaling in the endothelium. Mol. Cell. Biol. 2009, 29, 2011–2022. [Google Scholar] [CrossRef] [PubMed]
  10. Dumont, D.J.; Gradwohl, G.; Fong, G.H.; Puri, M.C.; Gertsenstein, M.; Auerbach, A.; Breitman, M.L. Dominant-negative and targeted null mutations in the endothelial receptor tyrosine kinase, tek, reveal a critical role in vasculogenesis of the embryo. Genes Dev. 1994, 8, 1897–1909. [Google Scholar] [CrossRef] [PubMed]
  11. Sato, T.N.; Tozawa, Y.; Deutsch, U.; Wolburg-Buchholz, K.; Fujiwara, Y.; Gendron-Maguire, M.; Gridley, T.; Wolburg, H.; Risau, W.; Qin, Y. Distinct roles of the receptor tyrosine kinases Tie-1 and Tie-2 in blood vessel formation. Nature 1995, 376, 70–74. [Google Scholar] [CrossRef] [PubMed]
  12. Suri, C.; Jones, P.F.; Patan, S.; Bartunkova, S.; Maisonpierre, P.C.; Davis, S.; Sato, T.N.; Yancopoulos, G.D. Requisite role of angiopoietin-1, a ligand for the TIE2 receptor, during embryonic angiogenesis. Cell 1996, 87, 1171–1180. [Google Scholar] [CrossRef]
  13. Nambu, H.; Umeda, N.; Kachi, S.; Oshima, Y.; Akiyama, H.; Nambu, R.; Campochiaro, P.A. Angiopoietin 1 prevents retinal detachment in an aggressive model of proliferative retinopathy, but has no effect on established neovascularization. J. Cell. Physiol. 2005, 204, 227–235. [Google Scholar] [CrossRef] [PubMed]
  14. Hackett, S.F.; Ozaki, H.; Strauss, R.W.; Wahlin, K.; Suri, C.; Maisonpierre, P.; Yancopoulos, G.; Campochiaro, P.A. Angiopoietin 2 expression in the retina: Upregulation during physiologic and pathologic neovascularization. J. Cell. Physiol. 2000, 184, 275–284. [Google Scholar] [CrossRef]
  15. Hackett, S.F.; Wiegand, S.J.; Yancopoulos, G.; Campochiaro, P. Angiopoietin-2 plays an important role in retinal angiogenesis. J. Cell. Physiol. 2002, 192, 182–187. [Google Scholar] [CrossRef] [PubMed]
  16. Oshima, Y.; Deering, T.; Oshima, S.; Nambu, H.; Reddy, P.S.; Kaleko, M.; Connelly, S.; Hackett, S.F.; Campochiaro, P.A. Angiopoietin-2 enhances retinal vessel sensitivity to vascular endothelial growth factor. J. Cell. Physiol. 2004, 199, 412–417. [Google Scholar] [CrossRef] [PubMed]
  17. Oshima, Y.; Oshima, S.; Nambu, H.; Kachi, S.; Takahashi, K.; Umeda, N.; Shen, J.; Dong, A.; Apte, R.S.; Duh, E.; et al. Different effects of angiopoietin 2 in different vascular beds in the eye; new vessels are most sensitive. FASEB J. 2005, 19, 963–965. [Google Scholar] [CrossRef] [PubMed]
  18. Fiedler, U.; Reiss, Y.; Scharpfenecker, M.; Gruunow, V.; Koidl, S.; Thurston, G.; Gale, N.W.; Witzenrath, M.; Rosseau, S.; Suttorp, N.; et al. Angiopoietin-2 sensitizes endothelial cells to TNF-alpha and has a crucial role in the induction of inflammation. Nat. Med. 2006, 12, 235–239. [Google Scholar] [CrossRef] [PubMed]
  19. Fiedler, U.; Scharpfenecker, M.; Koidl, S.; Hegen, A.; Grunow, V.; Schmidt, J.M.; Kriz, W.; Thurston, G.; Augustin, H.G. The Tie-2 ligand angiopoietin-2 is stored in and rapidly released upon stimulation from endothelial cell Weibel-Palade bodies. Blood 2004, 103, 4150–4156. [Google Scholar] [CrossRef] [PubMed]
  20. Watanabe, D.; Suzuma, K.; Suzuma, I.; Ohashi, H.; Ojima, T.; Kurimoto, M.; Murakami, T.; Kimura, T.; Takagi, H. Vitreous levels of angiopoietin 2 and vascular endothelial growth factor in patients with proliferative diabetic retinopathy. Am. J. Ophthalmol. 2005, 139, 476–481. [Google Scholar] [CrossRef] [PubMed]
  21. Loukovaara, S.; Robciuc, A.; Holopainen, J.M.; Lehti, K.; Pessi, T.; Liinamaa, J.; Kukkonen, K.T.; Jauhiainen, M.; Koli, K.; Keski-Oja, J.; et al. Ang-2 upregulation correlates with increased levels of MMP-9, VEGF, EPO and TGFbeta1 in diabetic eyes undergoing vitrectomy. Acta Ophthalmol. 2013, 91, 531–539. [Google Scholar] [CrossRef] [PubMed]
  22. Gerald, D.; Chintharlapalli, S.; Augustin, H.G.; Benjamin, L.E. Angiopoietin-2: An attractive target for improved antiangiogenic tumor therapy. Cancer Res. 2013, 73, 1649–1657. [Google Scholar] [CrossRef] [PubMed]
  23. Dugel, P. A novel anti-VEGF/anti-angiopoietin2 bispecific monoclonal antibody for wet age-related macular degeneration and diabetic macular edema. In Proceedings of the American Society of Retina Specialists Annual Meeting, San Francisco, CA, USA, 9–14 August 2016. [Google Scholar]
  24. Chakravarthy, U.; Bailey, C.; Brown, D.; Campochiaro, P.; Chittum, M.; Csaky, K.; Tufail, A.; Yates, P.; Cech, P.; Giraudon, M.; et al. Phase I Trial of Anti–Vascular Endothelial Growth Factor/Anti-angiopoietin 2 Bispecific Antibody RG7716 for Neovascular Age-Related Macular Degeneration. Ophthalmol. Retin. 2017, 1, 474–485. [Google Scholar] [CrossRef]
  25. Regula, J.T.; von Leithner, P.L.; Foxton, R.; Barathi, V.A.; Cheung, C.M.G.; Tun, S.B.B.; Wey, Y.S.; Iwata, D.; Dostalek, M.; Moelleken, J.; et al. Targeting key angiogenic pathways with a bispecific CrossMAb optimized for neovascular eye diseases. EMBO Mol. Med. 2016, 8, 1265–1288. [Google Scholar] [CrossRef] [PubMed]
  26. Study to Evaluate RO6867461 (RG7716) for Extended Curability in the Treatment of Neovascular Age-Related Macular Degeneration (nAMD) (STAIRWAY). ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03038880 (accessed on 13 March 2018).
  27. AVENUE: A Proof-Of-Concept Study of RG7716 in Participants with Choroidal Neovascularization (CNV) Secondary to Age-Related Macular Degeneration (AMD). ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/record/NCT02484690 (accessed on 13 March 2018).
  28. Rubio, R. Long-Acting Anti-VEGF Delivery. Retina Today, 2014. Available online: http://www.retinatoday.com/2014/08/long-acting-anti-vegf-delivery(accessed on 18 March 2018).
  29. Helzner, J. Sustained-Release Delivery of Lucentis Studied. Retinal Physician, 2016. Available online: http://www.retinalphysician.com/issues/2016/jan-feb/subspecialty-news(accessed on 13 March 2018).
  30. Study of the Efficacy and Safety of the Ranibizumab Port Delivery System (RPDS) for Sustained Delivery of Ranibizumab in Participants With Subfoveal Neovascular Age-Related Macular Degeneration (AMD) (LADDER). Clinicaltrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT02510794 (accessed on 13 March 2018).

Share and Cite

MDPI and ACS Style

Gahn, G.M.; Khanani, A.M. New Therapies of Neovascular AMD beyond Anti-VEGF Injections. Vision 2018, 2, 15. https://doi.org/10.3390/vision2010015

AMA Style

Gahn GM, Khanani AM. New Therapies of Neovascular AMD beyond Anti-VEGF Injections. Vision. 2018; 2(1):15. https://doi.org/10.3390/vision2010015

Chicago/Turabian Style

Gahn, Greggory M., and Arshad M. Khanani. 2018. "New Therapies of Neovascular AMD beyond Anti-VEGF Injections" Vision 2, no. 1: 15. https://doi.org/10.3390/vision2010015

Article Metrics

Back to TopTop