Subscribe to Newsletter
Subspecialties Glaucoma, Basic & Translational Research

A Glimpse Over the Horizon

When it comes to open-angle glaucoma, up to 10 percent of patients are not optimally responsive to current topical pressure-reducing medications; moreover, patient compliance is not an insignificant issue. In other words, there is room to improve.

Interestingly, the majority of medications in use today do not primarily target the major (conventional) aqueous outflow pathway – where aqueous humor drains through the trabecular meshwork (TM) into Schlemm’s canal (SC), and thus into the episclearal veins. Rather, they inhibit aqueous production by the ciliary body, or enhance its removal through the minor (uveoscleral) route. Much commercial interest therefore lies in the development of formulations that are more active on conventional outflow, and, indeed, such formulations have now emerged (Rhopressa) (1).

Glaucoma can be legitimately classified as a progressive retinopathy, elevated intraocular pressure-inducing degeneration at the optic nerve head with concomitant demise of the retinal ganglion cells. However, it is essentially unique among retinopathies in that the primary cause of the disease lies within the anterior portion of the eye, where aqueous drainage through the TM and SC is compromised, resulting in IOP elevation.

So far, no genetically-based therapeutics have been approved that target the primary outflow tissues (or indeed any aimed at protecting the viability of the ganglion cells), but there are signs on the horizon. The retina has been prominent as a target for gene therapy, principally because viral delivery systems (and especially, adeno-associated virus [AAV] vectors) can be introduced into the eye by the sub-retinal route. Proof of the point lies in the first FDA/EMA approval for a gene therapy targeting individuals with the inherited retinopathy, Leber congenital amaurosis (2).

Though tissues of the conventional outflow pathway have proven difficult to transfect with standard AAV vectors (1, 3), success has been seen with self-complementary AAV vectors (4, 5). In addition, AAV-Anc80L65, with its synthetically-designed capsid and a single-stranded genome, has also recently been shown to transfect tissues of the anterior chamber with high efficiency (6).

AAV9 has been similarly shown to transfect corneal endothelium in a highly efficient manner, and we recently reported a strategy for enhancement of outflow facility using this approach (7). Here, AAV9 expressing a matrix metalloproteinase (MMP3) transfects the corneal endothelium of mice (following a single intracameral inoculation), essentially rendering the endothelium a cellular factory for the expression of secretory proteins, which may then enter directly into the anterior chamber.

An entirely different approach recently reported by Tam and colleagues targeted SC itself with small-interfering siRNA (8). Aqueous humor funnels through the TM, converging on the endothelial cells lining SC; fluid then enters the canal either through the formation of vacuoles in endothelial membranes or through the spaces/clefts left between the tight junctions joining the endothelial cells together. By intracamerally inoculating rodents with siRNA targeting transcripts encoding selected tight-junction components, Tam and colleagues saw a subsequent widening of the clefts and an enhancement of canal permeability. Treatment of acute elevations in IOP could be one application of this approach.

It is of interest to note that siRNA delivery systems have recently been enhanced using nanoparticles that increase target specificity (9). In this study, researchers took advantage of the fact that TM and SC cells express the surface marker CD44. The polysaccharide hyaluronan can be used to target CD44, and so siRNA encapsulated in nanoparticles coated with hyaluronic acid allow for smaller amounts of siRNA to be efficiently delivered to these tissues.

Both siRNA and AAV vectors are accepted modes of ocular therapy. I’m personally excited that the advanced approaches shared here – and others like them – could hold significant potential in the future management of elevated IOP.

Receive content, products, events as well as relevant industry updates from The Ophthalmologist and its sponsors.

When you click “Subscribe” we will email you a link, which you must click to verify the email address above and activate your subscription. If you do not receive this email, please contact us at [email protected].
If you wish to unsubscribe, you can update your preferences at any point.

  1. T Borras et al., “Gene Therapy for Glaucoma: Treating a Multifaceted, Chronic Disease”, Invest. Ophthalmol. Vis. Sci., 43, 2513-2518 (2002). PMID: 12147578.
  2. U.S. Food & Drug Administration, “FDA approves novel gene therapy to treat patients with a rare form of inherited vision loss” (2017). Available at: tinyurl.com/ybrwkct4. Accessed January 17, 2019.
  3. T Borras, “Recent Developments in Ocular Gene Therapy”, Experimental Eye Research, 76, 643-652 (2003). DOI: 10.1016/S0014-4835(03)00030-7.
  4. T Borras et al., “Inducible scAAV2.GRE.MMP1 lowers IOP long-term in a large animal model for steroid-induced glaucoma gene therapy”, Gene Ther., 23, 438-449 (2016). PMID: 26855269.
  5. LK Buie et al., “Self-complementary AAV virus (scAAV) safe and long-term gene transfer in the trabecular meshwork of living rats and monkeys”, Invest. Ophthalmol. Vis. Sci., 51, 236-248 (2010). PMID: 19684004.
  6. L Wang et al., “Single stranded adeno- associated virus achieves efficient gene transfer to anterior segment in the mouse eye”, PLoS One., 12 (2017). PMID: 28763501.
  7. J O’Callaghan et al., “Therapeutic potential of AAV-mediated MMP3 secretion from corneal endothelium in treating glaucoma”. Hum Mol Genet, 26, 1230-1246 (2017). PMID: 28158775.
  8. LCS Tam et al., “Enhancement of Outflow Facility in the Murine Eye by Targeting Selected Tight-Junctions of Schlemm’s Canal Endothelia” Sci Rep., 16 (2017). PMID: 28091584.
  9. AE Dillinger et al., “Intracameral Delivery of Layer-by-Layer Coated siRNA Nanoparticles for Glaucoma Therapy”. Small, 14 (2018). PMID: 30353713.
About the Author
Jeffrey O'Callaghan

Jeffrey O'Callaghan is a Postdoctoral Researcher at the Institute of Genetics, Trinity College at the University of Dublin.

Related Case Studies
Business & Profession Glaucoma
TrabEx Pro: The Next Level in MIGS

| Contributed by MST

Finding Ocular Surface Inflammation

| Contributed by Quidel

Product Profiles

Access our product directory to see the latest products and services from our industry partners

Here
Most Popular
Register to The Ophthalmologist

Register to access our FREE online portfolio, request the magazine in print and manage your preferences.

You will benefit from:
  • Unlimited access to ALL articles
  • News, interviews & opinions from leading industry experts
  • Receive print (and PDF) copies of The Ophthalmologist magazine

Register

Disclaimer

The Ophthalmologist website is intended solely for the eyes of healthcare professionals. Please confirm below: