glaucoma

Contents

Glaucoma

Glaucoma is an umbrella term for a group of serious eye conditions that include open-angle glaucoma and angle-closure glaucoma (closed-angle glaucoma) caused by increased pressure inside your eye (raised intraocular pressure [IOP]) that damages your eye’s optic nerve (cranial nerve number 2 [CN II]), potentially leading to irreversible vision loss and blindness, and early detection and treatment are essential for preserving your eyesight. Note a glaucoma called “normal tension glaucoma” or normal or low-pressure glaucoma can happen even with normal eye pressure 1, 2, 3. Glaucoma can happen at any age, but is more common in older adults over 60 years of age. Glaucoma is the second-leading cause of blindness worldwide with more than 80 million people have glaucoma worldwide and this number is expected to surpass 110 million by 2040 4, 5, 6, 7, 8. Your optic nerve (cranial nerve number 2 [CN II]) sends visual information from your retina at the back of the eyes to your brain and is vital for good vision. The main cause is of glaucoma is often due to fluid buildup from blocked drainage. This increased pressure inside your eye called increased intraocular pressure or IOP damages the optic nerve, which often progresses slowly, causing gradual vision loss, typically starting with peripheral (side) vision 8. This is why glaucoma is called the “silent thief of sight”. Because early stages of damage to your eye’s optic nerve often have no symptoms, regular eye exams, especially for those over 40 years of age or with a family history of glaucoma, are important for detecting glaucoma and preventing vision loss. Later stages of damage to your eye’s optic nerve can cause blurred vision, halos around lights, and tunnel vision. While vision loss caused by glaucoma is permanent and cannot be reversed, treatment can slow down or halt further damage by reducing your eye pressure. Glaucoma treatment may include eye drops, laser treatments, or surgery to lower your eye pressure and prevent further damage. If glaucoma is not treated then people with glaucoma will slowly lose their peripheral vision. Over time, central vision may decrease and eventually this can lead to blindness. Furthermore, glaucoma is an inherited condition, so be sure to tell your first-degree relatives (your parent, brother and sister and child) that you have glaucoma so they can have their eyes checked for glaucoma.

In its early stages, glaucoma may not cause any symptoms or have no warning signs. That’s why up to half of the people in the United States with glaucoma may not know they have it. And symptoms may not appear until glaucoma causes irreversible eyesight damage.

Some of the more common glaucoma symptoms include:

  • Eye pain or pressure
  • Headaches
  • Red or bloodshot eyes
  • Double vision (diplopia)
  • Blurred vision
  • Gradually developing low vision
  • Gradually developing blind spots (scotomas) or visual field defects like tunnel vision

Some types of glaucoma, particularly angle closure glaucoma, can cause sudden, severe symptoms that need immediate medical attention to prevent permanent vision loss. Emergency glaucoma symptoms include:

  • Blood gathering in front of your iris (hyphema)
  • Bulging or enlarged eyeballs (buphthalmos = “ox eye”)
  • Nausea and vomiting that happen with eye pain/pressure
  • Rainbow-colored halos around lights
  • Sudden appearance or increase in floaters (myodesopsias)
  • Sudden vision loss of any kind
  • Suddenly seeing flashing lights (photopsias) in your vision.

It’s important to have regular eye exams that include measurements of your eye pressure. If glaucoma is found early, vision loss can be slowed or prevented. If you have glaucoma, you’ll need treatment or monitoring for the rest of your life.

Figure 1. Eye anatomy

Eye anatomy

Figure 2. Retinal layers showing retinal ganglion cell

Retina layers of the eye

Footnotes: Schematic representation of the retina and the retinal cell layers. (A) Blood supply and (B) structure of the retina. The retina is a layered structure lining the back of the eye consisting of a pigmented layer called retinal pigment epithelium (RPE), and a multilayered neuroretina. The retinal pigment epithelium (RPE) is in close contact with the outer segments of the photosensitive rod and cone cells of the neuroretina. The connecting cilium connects the photoreceptor outer segments with the cell bodies, which constitute a layer known as the outer nuclear layer (ONL). The axons of the photoreceptors synapse with the neuronal (bipolar, amacrine, and horizontal) cells of the inner nuclear layer (INL) via the outer plexiform layer (OPL). The axons of the inner nuclear layer (INL) cells in turn synapse with the ganglion cell layer (GCL) via the inner plexiform layer (IPL). The axons of the ganglion cells converge to form the optic nerve. Approximately 1.2 million nerve fibers, or axons, make up each human optic nerve. A retinal ganglion cell (RGC) is a type of neuron located near the inner surface (ganglion cell layer [GCL]) of the retina of the eye. A retinal ganglion cell (RGC) receives visual information from photoreceptors via two intermediate neuron types: bipolar cells and retina amacrine cells. Retina amacrine cells, particularly narrow field cells, are important for creating functional subunits within the ganglion cell layer and making it so that ganglion cells can observe a small dot moving a small distance. Retinal ganglion cells collectively transmit image-forming and non-image forming visual information from the retina in the form of action potential to several regions in the thalamus, hypothalamus, and mesencephalon, or midbrain. Visual images from the retina travel through the optic nerve, optic tract, and eventually to the visual part of the brain (the occipital lobe). There the images are processed and interpreted by the brain. Any disease process which affects the optic nerve could disrupt this input, leading to visual loss.

Abbreviations: ILM: internal limiting membrane, NFL: nerve fiber layer, GCL: ganglion cell layer, IPL: inner plexiform layer, INL: inner nuclear layer, OPL: outer plexiform layer, ONL: outer nuclear layer, ELM: external limiting membrane, OS: photoreceptor outer segment, RPE: retinal pigment epithelium

[Source 9 ]

Can glaucoma be reversed?

Glaucoma eyesight damage is permanent and it cannot be reversed. But medicine and surgery can help to stop further damage. To treat glaucoma, your ophthalmologist may use one or more of the following treatments.

  • Medications. Glaucoma medications lower pressure inside your eye in different ways. Some of them cause your pupil to relax more, improving aqueous humor drainage. Others slow the production of aqueous humor.
  • Glaucoma surgery. This approach usually aims to improve fluid flow and drainage. Examples of glaucoma surgeries that do this include laser trabeculoplasty, laser iridotomy, trabeculectomy (glaucoma filtration surgery), drainage tubes and minimally invasive glaucoma surgery (MIGS).

Aqueous Humor Production and Physiology

The aqueous humor is a water-like fluid that is produced by the ciliary body that sits directly behind the iris (the colored part of your eye). Aqueous humor is produced at a rate of 2-3 microliters per minute (2-3 μL/minute) 10, 11. The aqueous humor is composed of organic and inorganic ions, carbon dioxide, amino acids, carbohydrates, glutathione, and water 1012. The aqueous humor fills the anterior chamber of your eye with continual production, secretion, and reabsorption 10. The cornea and the lens of your eye have no blood supply. They receive nourishment from nutrients in the aqueous fluid that fills your eye. The aqueous fluid flows between the iris and lens through the pupil and to the drainage angle at the junction of the iris and the cornea. Aqueous fluid exits the eye through a tissue called the trabecular meshwork in the drainage angle. As the aqueous fluid passes through the eye, it supplies the lens and cornea with nutrients and carries away waste products. The production, circulation and reabsorption of aqueous humor are vital processes maintaining homeostasis of the eye. The pressure of the fluid in your eye called the intraocular pressure (IOP) is determined by the amount of aqueous humor fluid entering the eye through the ciliary body and exiting the eye through the trabecular meshwork. In most people, the balance between the aqueous humor fluid coming in and going out of the eye results in an eye pressure between 10 and 21 mm of Hg. In patients with glaucoma, aqueous humor fluid drains from the eye through the trabecular meshwork at a slower rate causing the pressure in your eye to rise or increased intraocular pressure (ocular hypertension) resulting in optic nerve damage and glaucoma.

Aqueous humor functions as a physical component allowing clear optics and filling the anterior chamber of the eye 10, 11. The aqueous humor is responsible for providing nourishment to the avascular components of the anterior chamber including the cornea and lens 10, 11. In addition, aqueous humor is responsible for removing waste products, blood, macrophages and other debris from the anterior chamber, including the trabecular meshwork 10, 11. The structure and function of the trabecular meshwork may become compromised by chronic oxidative stress from reactive oxygen species and insufficient antioxidant defense in the aqueous humor 10, 11, 13, 14. Decreased levels of antioxidants in aqueous humor are present in glaucomatous eyes versus normal eyes, consistent with the presence of increased oxidative stress and low-grade inflammation 13, 14.

The primary anatomic structures vital to the homeostasis of aqueous humor include the ciliary body as the site of principle production, and the trabecular meshwork and uveoscleral pathway as the sites of primary outflow 10, 15. Aqueous humor is produced by the ciliary body via a multistep process closely correlating with systemic vascular blood flow 10, 16, 17. Initially, blood enters the ciliary processes, which propels ultrafiltrate from the blood into the ciliary interstitial space via a pressure gradient 10, 16, 17. Next, the ciliary epithelium transports plasma components from the basal to the apical surface in order to synthesize aqueous humor and transport it into the posterior chamber 10, 16, 17. Passive diffusion and ultrafiltration are key in initial synthesis, and active secretion across a blood-aqueous barrier via aquaporins, Na-K-ATPase and carbonic anhydrase enzymes are necessary for final synthesis 10, 16, 17, 18. These active transport enzymes necessary for final synthesis are common pharmacologic targets in decreasing aqueous humor production. Although systemic blood flow via the ciliary artery is required for the initial production of ultrafiltrate, the production of aqueous humor is independent from systemic blood pressure due to a fixed rate of 4% filtration of plasma 17. Therefore, there is minimal association between systemic high blood pressure (hypertension) and elevated intraocular pressure (IOP). The estimated rate of aqueous humor production is approximately 2.4 microliters per minute (2.4 μL/minute), with diurnal variations leading to higher aqueous humor flow in the morning and lower flow in the evening 10, 16.

While aqueous humor production is well documented, the mechanism of drainage is still poorly understood.

There are 2 main drainage pathways for aqueous humor 10, 19, 16:

  1. The conventional pathway via trabecular meshwork, Schlemm’s canal, collector channels, and the episcleral venous system), and
  2. The unconventional pathway via uveoscleral, uveovortex, uveolymphatic.

The conventional pathway drainage pathways for aqueous humor involves passive drainage throughout the trabecular meshwork although the Schlemm’s canal has been documented with paracellular and intracellular pores 10, 19, 16. The trabecular meshwork is a triangular porous structure composed of a layer of connective tissue and endothelium with sympathetic innervation from superior sympathetic ganglion, and parasympathetic innervation from the ciliary ganglion 10, 19, 16. The trabecular meshwork may be divided into the uveal meshwork (iris root, ciliary body, peripheral cornea), corneoscleral meshwork (scleral spur), and juxtacanalicular meshwork (transition into Schlemm’s canal) 10, 19, 16. Schlemm’s canal is a structure with composition similar to venous vasculature, with fenestrated thin endothelium surrounded by connective tissue 10, 19, 16. After drainage through the trabecular meshwork and the Schlemm’s canal, aqueous humor continues through collector channels into the episcleral venous system which deposits into the main venous system 10, 19, 16.

Resistance to outflow through the trabecular meshwork and Schlemm’s canal has been documented although it is poorly understood, yet resistance remains an important factor in regulating intraocular pressure and the pathogenesis of glaucomatous processes. In humans, up to 75% of aqueous outflow resistance is contributed by the trabecular meshwork while the remaining 25% is due to resistance beyond Schlemm’s canal 10. The rate of outflow is directly influenced by iris and ciliary muscles which contract and relax based on cholinergic innervation and pharmacodynamics 10, 19, 1615, 20. In ciliary contraction, the trabecular meshwork and Schlemm’s canal dilate, decreasing resistance and increasing outflow 10, 19, 1615, 20. The rate of outflow is also influenced by intraocular pressure, with higher intraocular pressure altering the structure of endothelial lining in Schlemm’s canal to increase the number of porous vacuoles allowing increased outflow 10, 19, 1615, 20. However, it is still debated if this finding substantially contributes to increasing outflow in glaucomatous eyes 10, 19, 1615, 20.

The unconventional pathway involves drainage into the orbital vasculature, vortex veins and ciliary lymphatics, contributing up to 25-40% of total aqueous outflow in cynomolgus and vervet monkey models. The uveoscleral pathway involves diffusion into the sclera and episcleral through the orbital vasculature. The uveovortex pathway involves osmotic absorption of fluid through the choroid, passing into the vortex veins 19. Lastly, the uveolymphatic pathway involves drainage into lymphatic vessels within the ciliary body, although the extent of drainage under normal physiological conditions remains controversial 19. In addition, the unconventional pathway also includes corneal, iridial and retinal routes, albeit less clinically significant 21. Regardless of downflow pathway, all unconventional paths require drainage through the interstitial spaces of the ciliary muscle 19, 21. Resistance also exists within the unconventional pathway likely due to ciliary muscle tone, as seen with changes in outflow in the setting of pilocarpine, increasing ciliary tone and decreasing flow, and atropine, decreasing ciliary tone and increasing flow 19, 21. Therefore, the unconventional pathways are also clinically important in moderating intraocular pressure, and serve as a potential target in glaucoma therapy.

Figure 3. Normal aqueous outflow

Normal aqueous outflow
aqueous humor formation and drainage
Trabecular Meshwork

Footnotes: The ciliary body is a structure that sits directly behind the iris (the colored part of your eye). One of ciliary body’s jobs is to create an important fluid called aqueous humor, a fluid that nourishes the cornea and lens. Aqueous humor flows through a specific route into the front of the eye (the anterior chamber). This route allows aqueous humor to send important nutrients and oxygen to other parts of the eye, such as the lens and cornea. The aqueous humor is produced behind the iris, flows into the anterior chamber through the pupil, and exits the eye between the iris and cornea via the trabecular meshwork, a specialized eye tissue located at the chamber angle of the eye next to the cornea 22. In a healthy eye, this is a constant process. The ciliary body is always producing aqueous humor, and 80%-90% aqueous humor is always draining through the trabecular meshwork. The trabecular meshwork is a specialized spongy tissue in the anterior chamber of the eye that regulates the outflow of aqueous humor 22. The trabecular meshwork acts as a filter, controlling how quickly aqueous humor drains out of the eye through a structure called Schlemm’s canal, ultimately maintaining intraocular pressure (IOP). The canal of Schlemm, also known as Schlemm’s canal or the scleral venous sinus, is a circular, lymphatic-like vessel in the eye that drains aqueous humor from the anterior chamber into the episcleral blood vessels. The canal of Schlemm and the trabecular meshwork (TM) play a crucial role in maintaining intraocular pressure (IOP) by facilitating the outflow of aqueous humor. Too much aqueous humor production or obstruction of its outflow causes a rise in intraocular pressure (IOP) that can lead to glaucoma.

[Source 23 ]

Glaucoma types

There are many different types of glaucoma. However, most glaucomas can be divided into 2 categories 24:

  • Open-angle glaucoma: “Open-angle” means that the drainage angle, where the inside of the sclera (the white of your eye) and the outer edge of your iris meet, is open wide. Aqueous humor flows into the drainage angle so it can drain out of the anterior chamber. But other parts of the drainage system don’t drain properly like a clogged drain. This may lead to a slow, gradual increase in eye pressure that starts to damage the optic nerve. Open-angle glaucoma is the most common form of glaucoma. In the United States, open-angle glaucoma is what most people mean when they talk about glaucoma 25. Open-angle glaucoma is painless and causes no vision changes at first.
  • Closed-angle glaucoma, also called angle-closure glaucoma or narrow-angle glaucoma, where the drain of the eye is partially or fully obstructed or “closed” by the bulging iris. The bulging iris partially or completely blocks the drainage angle. You can think of it like a piece of paper sliding over a sink drain. As a result, fluid (aqueous humor) can’t circulate through the eye and pressure increases. Angle-closure glaucoma is a uncommon type of glaucoma and usually affects one eye at a time. Angle-closure glaucoma may happen suddenly or gradually. There are 2 main types of closed-angle glaucoma:
    • Acute closed-angle glaucoma is a medical emergency. No fluid can drain out of your eye, so eye pressure increases suddenly. This causes eye pain, blurry vision, and other symptoms. Without prompt treatment, acute angle-closure glaucoma can cause blindness in a few days.
      • Here are the signs of an acute angle-closure glaucoma attack:
        • Your vision is suddenly blurry
        • You have severe eye pain
        • You have a headache
        • You feel sick to your stomach (nausea)
        • You throw up (vomit)
        • You see rainbow-colored rings or halos around lights
    • Chronic closed-angle glaucoma develops slowly over time. You may not have any symptoms at first. Or you may have symptoms such as eye discomfort, blurry vision, redness, or headaches. The symptoms often get better with sleep. Without treatment, vision loss develops slowly.

Within these categories there are different variants of open angle and angle-closure glaucoma which can be classified according to cause:

  • Primary glaucomas: No known cause, occurs in susceptible individuals.
  • Secondary glaucomas: Secondary glaucoma is when another condition or problem within the eye increases eye pressure such as eye injury, eye surgery or eye procedures, certain medications especially corticosteroids and cycloplegics or other eye diseases (e.g., pigmentary dispersion syndrome, uveitis) causing the glaucoma. This is when another condition or event increases eye pressure, which leads to glaucoma.
  • The two most common types of glaucoma are Primary Open Angle Glaucoma (POAG) and Primary Angle-Closure Glaucoma (PACG).

Childhood glaucoma also known as infantile glaucoma or congenital glaucoma. Childhood glaucoma is a rare glaucoma where high pressure builds up inside the eye during fetal development, potentially causing vision loss and even blindness if left untreated. Childhood glaucoma is typically diagnosed within the first few months of life commonly in children under 2 years of age and is caused by a developmental defect in the eye’s drainage system, preventing fluid from flowing out properly. Childhood glaucoma affects about 1 in 10,000 children under 2 years of age in the United States 26, 27. Childhood glaucoma can be caused by aniridia (a rare genetic eye disorder characterized by the complete or partial absence of the iris, the colored part of the eye), Axenfeld-Rieger syndrome, Marfan syndrome, congenital rubella syndrome and neurofibromatosis type 1. In most cases, childhood glaucoma is diagnosed by the age of six months, with 80% diagnosed in the first year of life.

Eye specialist (ophthalmologist) may also refer to someone as a “glaucoma suspect” if they think the person might be showing early signs of glaucoma but they are not yet sure. Many people suspected of having glaucoma at this stage turn out not to have it at all, but some do develop it in time and it is these people who can benefit the most from timely treatment.

Ocular hypertension is diagnosed in individuals with intraocular pressure (IOP) levels exceeding 21 mmHg without signs of glaucomatous optic neuropathy or functional visual field defects 28. Research indicates that around 20% of people with ocular hypertension may progress to glaucoma, highlighting the importance of regular testing, tonometry, and comprehensive eye examinations to initiate appropriate treatment aimed at reducing intraocular pressure (IOP) in the presence of initial glaucomatous damage 29

Figure 4. Glaucoma types

Glaucoma types
[Source 30 ]

Glaucoma Suspect

Eye specialist (ophthalmologist) will refer to someone as a “glaucoma suspect” if they think the person might be showing early signs of glaucoma such as higher than normal eye pressure called ocular hypertension but have no signs of optic nerve damage. Glaucoma suspects have no symptoms to suggest eye disease. They are usually identified as glaucoma suspects during routine checks by their optometrist. Many people suspected of having glaucoma at this stage turn out not to have it at all, but some do develop it in time and it is these people who can benefit the most from timely treatment. Their ophthalmologist (eye specialist) may notice something different about their optic nerve. Most “glaucoma suspects” have no symptoms. That is why you need to be carefully monitored by your ophthalmologist if you are a glaucoma suspect. An ophthalmologist can check for any changes over time and begin treatment if needed.

If someone has a very high intraocular pressure (high IOP) or very advanced optic nerve damage then the diagnosis of glaucoma is usually straightforward. However sometimes it is not entirely clear whether someone has glaucoma or not. The early signs of glaucoma can be subtle, and many glaucoma patients have a normal pressure.

There is no single test that is 100% effective in confirming the diagnosis of glaucoma all the time. Sometimes the only way to be sure that someone has glaucoma is to arrange follow up eye examinations every 4-6 months or so to work out whether progressive damage is occurring to the optic nerve in one or both eyes. Features in the examination which might lead to a patient being classified as a ‘glaucoma suspect’ include:

  • A high pressure within the eyeball (high IOP) but with no optic nerve damage yet this is also referred to as ocular hypertension.
  • A ‘suspicious’ optic disc appearance on examination such as ‘cupping’ of the disc or thinning of the neuro-retinal rim or nerve fiber layers.
  • Unusual or defective visual fields.

These are changes that can be seen with glaucoma, but can also be seen in other conditions such as farsightedness (myopia) where it may be a variation of normal.

Other risk factors for glaucoma such as a strong family history of glaucoma but without definite changes to the optic nerve as yet. Generally speaking, “glaucoma suspects” will not show any visual field defects on testing, or may show some field defects which are not yet entirely convincing as evidence of glaucoma. If you are a ‘glaucoma suspect’, the most important treatment is good follow-up care.

It is very important that someone suspected of experiencing the early onset of glaucoma has regular eye checks to make sure there is no continuing damage to the optic nerve. Even though a person is not yet receiving any treatment for glaucoma, she or he may still risk losing their vision if in fact they do turn out to have glaucoma. Thus it is very important to maintain follow-up care. Typically for a low-risk glaucoma suspect, this may require visits every 6 to 12 months. At each follow-up visit your eye doctor will check your vision and eye pressure, and examine the front and back of your eye, paying careful attention to the appearance of your optic nerves.

To examine the structure of the optic nerve, your doctor will perform a careful examination in the office, obtain optic nerve imaging, and obtain a baseline set of optic nerve photographs. To examine the function of the optic nerve, an automated visual field test we be implemented with the help of a technician, who will instruct you on the correct way to perform the test. All of these tests may be repeated at yearly intervals (or more or less frequently, as determined by your eye doctor) to assess if there are changes or “progression” over time. The follow-up visits are crucial to maintaining optimal eye health.

Sometimes eye doctors are on the fence about whether to start treatment, and it is only through repeat follow-up visits that they get a sense of whether or not someone has glaucoma. Usually a person thought to be a “glaucoma suspect” will not be treated for the condition until the diagnosis is confirmed. Typically, glaucoma advances slowly so its progress can be tracked safely without treatment until the diagnosis is confirmed.

If you’re a “glaucoma suspect” and needed treatment, initial treatment options may include topical eye drops or laser treatment of the drainage angle to increase the amount of fluid draining from the eye, both of which can lower the eye pressure. The decision to treat is often not a cut-and-dry one; your ophthalmologist will assess all of your risk factors, your examination findings, and seek your input as to whether to treat or continue to observe your eyes over time. Some patients prefer to “watch and wait” or are worried about the side effects of treatment, while others may be more risk-averse and would rather begin treatment and have peace of mind. There are some glaucoma risk calculators available but most eye doctors would agree that these may aid in diagnosis and assessment, but will not replace your doctor’s clinical judgment.

Normal Tension Glaucoma

Normal tension glaucoma (NTG), also known as normal or low-pressure glaucoma, is a type of primary open-angle glaucoma (POAG) where the optic nerve damage and vision loss characteristic of glaucoma occur despite eye pressure (intraocular pressure [IOP]) remaining within the normal range and an open, normal appearing anterior chamber angle 131, 2, 3, 32, 33. The definitions of normal tension glaucoma (NTG), however, may vary slightly amongst different countries. The European Glaucoma Society Guidelines, published in 2021 34, state that “normal tension glaucoma is a specific type of POAG characterized by glaucomatous optic nerve head damage and corresponding visual field defects in patients with IOP consistently less than 21 mmHg”. The Preferred Practice Pattern Guidelines published in 2021 by the American Academy of Ophthalmology 35 define the normal tension glaucoma as “a common form of POAG, i.e., a chronic, progressive optic neuropathy that results in a characteristic optic nerve head cupping, retinal nerve fiber layer thinning and functional visual field loss, in which there is no measured elevation of the IOP”. In 2015, the Canadian Ophthalmological Society Guidelines 36 reported normal tension glaucoma as “a subgroup of POAG with characteristic visual field defects and glaucomatous optic nerve head changes in patients having normal IOP levels less than 21 mmHg”. The Asia-Pacific Glaucoma Guidelines, published in 2016 37, reported that “the normal tension glaucoma is a condition in which the typical glaucomatous progressive optic nerve damage and visual field loss occur although the intraocular pressure remains normal”. The Japanese Glaucoma Society published guidelines in 2023 38, which defined normal tension glaucoma as “a subtype of POAG in which the IOP always remains within the statistically determined normal range during the developmental process of glaucomatous optic neuropathy (GON)”.

Normal eye pressure is 10 to 21 mmHg and approximately 95% of general population will have an intraocular pressure (IOP) between 11 and 21 mmHg 39. A cut-off of 21 mmHg intraocular pressure (IOP) is often applied to define normal tension glaucoma 1. One of the main risk factors for the development of glaucoma is the increased intraocular pressure (the pressure within your eyeball is higher than normal). The higher the intraocular pressure (IOP), the more likely glaucoma is to develop. However this is not the only risk factor for glaucoma. It is also widely recognized that in about 1/3rd of cases of glaucoma the characteristic optic nerve changes and visual field loss can develop in an eye with normal pressure – this is termed normal tension glaucoma. Unlike the typical glaucoma, where high intraocular pressure (increased IOP) is the primary cause, normal tension glaucoma is often linked to other factors, such as blood flow issues to the optic nerve. Normal-tension glaucoma can also lead to vision loss and blindness. Drance and colleagues 40 described two forms of normal tension glaucoma: 1) a non-progressive form typically associated with a transient episode of vascular compromise, and 2) a progressive form thought to result from a chronic vascular insufficiency at the optic nerve. While some try to delineate normal tension glaucoma and primary open angle glaucoma (POAG) as two completely unique disease processes, it has also been suggested that the diseases exist on a continuum with intraocular pressure (IOP) playing a larger role in primary open angle glaucoma (POAG), and vascular or mechanical factors at the etiologic root in normal tension glaucoma 41.

The histopathological changes in normal tension glaucoma are the same as those found in primary open angle glaucoma (POAG), namely loss of retinal ganglion cell axons (retinal nerve fiber layer – RNFL) and glial tissue at the optic nerve head, leading to optic disc excavation and cupping 42. Typically, retinal nerve fiber layer loss is more common in the supero- and inferotemporal neuroretinal bundles, leading to notches, but loss may also be global and concentric 1. Classically, the presence of small flame shaped optic disc hemorrhages also known as Drance hemorrhages, has been associated with normal tension glaucoma though this finding can be observed in any form of primary open angle glaucoma (POAG) (see Figure 2). Optic disc hemorrhages or Drance hemorrhages are splinter or flame-shaped hemorrhages oriented perpendicular to the optic disc margin 43, 44, 45, 46. Classically, Drance hemorrhages are located in the prelaminar optic disc, cross the peripapillary zone, and extend into the adjacent superficial retinal nerve fiber layer, although they may not occupy the entire length from disc to retina 43, 44, 45. Alternately, deeper disc hemorrhages may appear round and blotchy 43. Less commonly, a optic disc hemorrhage may be noted in the peripapillary retinal nerve fiber layer reaching within one disc diameter of the optic disc margin 46. Additional research is needed to determine exactly why optic disc hemorrhages or Drance hemorrhages occur and to clarify why only some glaucoma patients develop optic disc hemorrhages 47. Drance hemorrhages and temporal retinal nerve fiber layer defects with localized, deep paracentral scotomas, more often in the superior hemifield are more common in normal tension glaucoma than in primary open angle glaucoma (POAG) 48, 49. Anderson et al 50 reported the presence of optic disc hemorrhages at the time of diagnosis of normal tension glaucoma as an unfavorable prognostic marker for likely visual field progression.

A recent cross-sectional study aimed to further describe a potential link between the incidence of normal tension glaucoma in patients with dementia 51. The study demonstrated an association between normal tension glaucoma status and poor cognition (measured with the Telephone Version of the Montreal Cognitive Assessment, T-MoCA) thereby concluding that there exists a disease association and shared features between normal tension glaucoma and dementia when compared to those with high tension glaucoma (HTG) 51. Conversely, a recent study demonstrated that persons with normal tension glaucoma had increased risk for developing vascular dementia, with particular increased risk when diagnosed with glaucoma at ages >70 52.

While the exact prevalence of normal tension glaucoma can vary by geographic region, normal tension glaucoma is estimated to comprise a significant proportion of all glaucoma cases, with some studies suggesting it may account for up to 30-40% of all glaucoma patients amongst Caucasians or Africans 53, 31. Whereas, for unclear reasons, the proportion of normal tension glaucoma amongst primary open angle glaucoma (POAG) in Asian populations was reported as high as 92.3% in Japan (the Tajimi Study), 84.6% in Singapore (the Singapore Malay Eye Study), 83.58% in northern China (the Handan Eye Study), 82% in south India (the Chennai Glaucoma Study), 79.6% in southern China (the Liwan Eye Study), 77% in South Korea (the Namii Study), yet only 31.7% in the United states of America (the Beaver Dam Eye Study) demonstrating that there is a predilection for those of Asian descent 54, 55. Current research tackles the disparity demonstrated by the aforementioned data, suggesting that other factors/predispositions within specific ethnic groups may be playing a significant role in the pathogenesis of normal tension glaucoma.

The therapeutic approaches to normal tension glaucoma are still strongly debated. Intraocular pressure (IOP)-lowering treatment management should be tailored specifically to each patient. Treatment may need to be changed as the disease progresses or a patient’s response to medicine changes. Considering that large prospective, multicenter, randomized and controlled clinical trials such as the Collaborative Normal-Tension Glaucoma Study (CNTGS) and the Early Manifest Glaucoma Trial (EMGT) have demonstrated that an intraocular pressure (IOP) reduction of at least 25–30% from baseline values is effective in delaying the progression of the visual field damage in a high percentage of normal tension glaucoma patients 56, 57. It is thus important to note that despite the fact that individuals with normal tension glaucoma have by definition normal intraocular pressure (IOP) levels, lowering the intraocular pressure (IOP) remains the gold standard in the normal tension glaucoma treatment 58, 59, 60.

Many different ocular hypotensive drugs are available on the market: prostaglandin and prostamide analogs, beta-blockers, alpha agonists and carbonic anhydrase inhibitors are examples of topical glaucoma drugs that are frequently used to reduce intraocular pressure (IOP), taken both as topical single therapy or in combination. Prostaglandins and prostamide analogs are the most safe and effective intraocular pressure (IOP)-lowering medications and represent the first choice in normal tension glaucoma therapy 59, 61, 62, 63.

The use of local and systemic beta-blockers and oral calcium channel blockers, especially in the evening, is of particular concern in normal tension glaucoma patients, because they may induce severe nocturnal systemic hypotension, with subsequent ocular perfusion pressure drop 64, 61, 65, 66, which is considered one of the most important risk factors for normal tension glaucoma onset and progression 67, 68.

Besides providing an intraocular pressure (IOP)-lowering effect, some ocular hypotensive drugs have also shown the ability to increase the optic nerve head blood flow (latanoprost, bimatoprost, betaxolol, carteolol, levobunolol, carvedilol and nebivolol) or neuroprotective properties (brimonidine, betaxolol, carteolol, carvedilol, latanoprost, bimatoprost and tafluprost) 69, 70, 71, 72, 73, 74, 63. These adjunctive (add-on) properties could be particularly useful in treating normal tension glaucoma patients. Moreover, the confirmation of the ability of the novel beta-blockers, carvedilol and nebivolol, to reduce intraocular pressure (IOP) and increase the ocular blood flow in clinical trials in glaucomatous patients may lead to the development of new glaucoma therapies.

For optimal management of normal tension glaucoma, regular monitoring of intraocular pressure (IOP), visual field and optic nerve head by eye care specialists is important. Moreover, performing diurnal intraocular pressure (IOP) curves and addressing intraocular pressure (IOP) peaks are considered to be the most important therapeutic strategies in normal tension glaucoma patients with normal office intraocular pressure (IOP) values 67, 59.

Considering that normal tension glaucoma patients have, by definition, a baseline intraocular pressure (IOP) within the statistically normal range, it is often difficult to reduce the intraocular pressure (IOP) values with medications alone, so non-medical options are often used, including laser and surgical treatments 59. The Collaborative Normal-Tension Glaucoma Study (CNTGS) 56 showed that 57% of the patients achieved a 30% intraocular pressure (IOP) reduction with topical medications, laser trabeculoplasty or both; the remaining 43% required filtering surgery, which remains the most proven option in the treatment of normal tension glaucoma patients when medications or laser treatments are unable to stop the visual field and/or optic nerve head damage progression 59, 75.

Figure 5. Drance hemorrhage (optic disc hemorrhage)

optic disc hemorrhage

Footnotes: Optic disc hemorrhage or Drance hemorrhage indicating inadequate intraocular pressure control in a patient with normal tension glaucoma. Disc hemorrhages are more common in normal tension glaucoma than in primary open angle glaucoma (POAG). This patient also has peripapillary atrophy, visible as a pale ring around the optic nerve.

[Source 76 ]

Figure 6. Normal tension glaucoma optic disc photographs

Normal tension glaucoma optic disc photographs

Footnotes: Normal tension glaucoma optic disc photographs. (A) Right and (B) Left eye in a patient with normal tension glaucoma. Note the focal superotemporal thinning and associated dropout of the retinal nerve fiber layer. The corresponding visual field defects are seen in Figure 7 below.

[Source 41 ]

Figure 7. Normal tension glaucoma visual fields

Normal tension glaucoma visual fields

Footnotes: Standard automated perimetry of (A) Left and (B) Right eye in a patient with normal tension glaucoma. Note the dense inferior arcuate scotomas occurring near fixation with minimal involvement of periphery. The corresponding optic disc photographs are seen in Figure 6 above.

[Source 41 ]

Normal tension glaucoma causes

The cause and mechanism for the development of normal tension glaucoma is unknown and remains an area of active research and debate. Several theories have been proposed to explain the onset and progression of normal tension glaucoma (NTG). Whereas intraocular pressure (IOP) is the main driver of progressive visual loss in most patients with primary open angle glaucoma (POAG), normal tension glaucoma (NTG) likely represents a diverse and multifactorial group of causes with a common final pathway of retinal ganglion cell loss 1. Despite intraocular pressure (IOP) in the normal range of 10 to 21 mmHg, there is evidence that an intraocular pressure (IOP)-dependent mechanism plays a role in the cause in many eyes with normal tension glaucoma 77, 78. Proposed intraocular pressure (IOP)-independent mechanisms include vascular insufficiency (lower blood pressure or reduced ocular blood flow) at the optic nerve head, impaired cerebrospinal fluid (CSF) circulation resulting in low retrobulbar cerebrospinal fluid pressure causing stagnation and decreased optic nerve protection, failure of the glymphatic system in the optic nerve, metabolic and neurodegenerative disorders, oxidative stress, and structural anomalies including structural weakness of the lamina cribrosa 79, 80, 81, 82, 83, 54. All of these mechanisms need further research to better define the pathophysiology of the disease process 41.

It has been theorized that the disease process in normal tension glaucoma results from an enhanced sensitivity to what would otherwise be physiologic intraocular pressure (IOP), resulting in glaucomatous damage of the optic nerve. This enhanced sensitivity may be due to impaired optic nerve blood flow, a higher translaminar pressure gradient (intraocular pressure [IOP] minus intracranial pressure [ICP]) due to lower intracranial pressure (ICP), or a structurally abnormal lamina cribrosa, which cannot withstand a normal range of intraocular pressure (IOP) 41. This theory of enhanced sensitivity is useful, at least conceptually, to rationalize the impact of intraocular pressure (IOP) in a disease process that may have an intraocular pressure (IOP) independent underlying etiology 41. The evidence for a role of IOP contributing to normal tension glaucoma comes from the Collaborative Normal Tension Glaucoma Study, which showed a slowing of disease progression in patients achieving a 30% or more reduction of already normal intraocular pressure (IOP) 77, 84. While some try to delineate normal tension glaucoma and primary open angle glaucoma (POAG) as two completely unique disease processes, it has also been suggested that the diseases exist on a continuum with IOP playing a larger role in primary open angle glaucoma (POAG), and vascular or mechanical factors at the etiologic root in normal tension glaucoma 41.

Specific histological studies of eyes with normal tension glaucoma are scarse but in general mimic those changes seen in primary open angle glaucoma (POAG) 41. Histopathologic changes of the optic nerve head include disarrangement and posterior bowing of the lamina cribrosa along with loss of nerve fibers 85. Non-invasive imaging by OCT and scanning laser modalities have characterized thinning of the peripapillary choroid 86, as well as thinning of the ganglion cell layers in normal tension glaucoma patients compared to other primary open angle glaucoma (POAG) and normal patients 87. In Asian patients this thinning has been correlated with vascular narrowing in asymmetric normal tension glaucoma when compared to normal fellow eyes and primary open angle glaucoma (POAG) patients with elevated pressures 88, 89, 90.

Genetics is also known to play a role in normal tension glaucoma, because of the strong association with family history with 21% of patients reporting a family history of glaucoma and variation in prevalence in different ethnicities that persists after migration 1. Four major genes have been implicated in normal tension glaucoma: optineurin (OPTN), TANK binding kinase 1 (TBK1), methyltransferase-like 23 (METTL23), and myocilin (MYOC) 82. Optineurin (OPTN) gene mutations, particularly the E50K variant, have been strongly associated with normal tension glaucoma, causing early-onset disease, large cup-to-disc ratios, and retinal ganglion cell death 41. TANK binding kinase 1 (TBK1) copy number variations have also been linked to normal tension glaucoma, with duplications and triplications contributing to retinal ganglion cell (RGC) loss 41. Methyltransferase-like 23 (METTL23) gene mutations were recently identified in familial normal tension glaucoma cases, with evidence suggesting that these mutations impact histone arginine methylation, potentially leading to retinal ganglion cell (RGC) degeneration 41. Myocilin (MYOC) gene commonly associated with primary open angle glaucoma (POAG) has been implicated in some normal tension glaucoma cases, though its role remains less clear 41. Further genetic research is certainly needed to better understand the role of these genes in normal tension glaucoma 82.

Genes associated with normal tension glaucoma 1, 82:

  • Optineurin (OPTN)
  • TANK binding kinase (TBK1)
  • Methyltransferase-like 23 (METTL23)
  • Myocilin (MYOC)

Normal tension glaucoma is typically not considered to be a heritable disease, as approximately 2% of normal tension glaucoma cases are caused primarily by a mutation of a single gene and found to be transmitted by an autosomal dominant inheritance pattern 41. Nevertheless, individuals who carry one of the many autosomal dominant gene mutations may present with symptoms of normal tension glaucoma as early as 23 years old 91. Genetic and pedigree studies continue to further identify numerous new genes associated with the development of normal tension glaucoma, but further studies that demonstrate a higher incidence of disease are necessary before a clinical indication for genetic screening and counseling can be recommended 41.

Though the quantity of axons that compose optic nerves in humans remains a predictable constant between individuals, variability in surface area of optic discs is observed. It is unclear if certain optic nerve head parameters place an eye at increased risk of normal tension glaucoma 41. Optic nerves with a larger surface area and with thinner inferior/inferotemporal rims have been reported to be at an increased risk for developing normal tension glaucoma 92, 93. Other studies evaluating the optic nerve head by scanning laser ophthalmoscopy found no morphologic differences between high-tension and normal tension glaucoma patients 94.

Frequently an area of peripapillary atrophy in a crescent or halo configuration is observed in patients with normal tension glaucoma. While this pattern of atrophy can be a finding in eyes without normal tension glaucoma, in glaucomatous eyes, peripapillary atrophy often occurs adjacent to areas of greatest disc thinning and corresponding visual field loss 95. While thinning of the optic nerve rim is observed in all primary open angle glaucoma (POAG), focal thinning or ‘notching’ is more commonly observed in normal tension glaucoma 96.

Intraocular Pressure (IOP)

Although always residing within the normal range for intraocular pressure (IOP), patients with normal tension glaucoma have been suggested to have higher-normal intraocular pressure (IOP) levels 97. By contrast, prospective evaluation of patients in the Low-Pressure Glaucoma Treatment Study found no relation between intraocular pressure (IOP) asymmetry and visual field asymmetry 98. Wide diurnal fluctuations in intraocular pressure (IOP) and nocturnal intraocular pressure (IOP) spikes have also been correlated with normal tension glaucoma 97.

Systemic vascular disease

Patients with systemic conditions that result in ischemic vascular disease such as diabetes and patients with a history of stroke have been shown to be at increased risk for bilateral normal tension glaucoma compared to unilateral normal tension glaucoma 41. Patients with normal tension glaucoma may have increased diastolic blood pressure and display larger dips in blood pressure overnight compared to normal 41. Similarly, it has been suggested that obstructive sleep apnea (OSA) may lead to transient episodes of nocturnal hypoxemia and compromised optic nerve head perfusion 41. A higher prevalence of obstructive sleep apnea (OSA) among normal tension glaucoma patients has been noted in several studies 99, 100, 101. Moreover, one study demonstrated a correlation between moderate/severe obstructive sleep apnea (OSA) and higher progression of retinal nerve fiber layer (RNFL) loss 102.

Certain vasospastic conditions such as Raynaud disease (a condition where blood vessels in the fingers and toes narrow in response to cold or stress, causing temporary blood flow restriction) are thought to be associated with normal tension glaucoma, with the most reported associated condition being migraine 103. Recently, a disease entity termed primary vascular dysregulation (also known as vasospastic syndrome, refers to a condition where the regulation of blood flow isn’t adapted to the needs of tissues, despite anatomically healthy vessels and the absence of an underlying disease) has been described pointing to retinal and optic nerve vasculature dysregulation as a potential risk factor for normal tension glaucoma 104. As above, a recent study demonstrated a significantly increased risk of developing vascular dementia in individuals diagnosed with normal tension glaucoma, further supporting a vascular deregulatory element in the pathogenesis of normal tension glaucoma 52.

A recent cross-sectional study investigated the prevalence of normal tension glaucoma in patients with Conn’s syndrome also called primary aldosteronism. Of the 212 patients with primary hyperaldosteronism included in the study, the prevalence of normal tension glaucoma in primary hyperaldosteronism patients was 11.8%, significantly higher than in hypertensive patients without primary aldosteronism (5.2%) 105. The study found a fourfold increase in the odds of developing normal tension glaucoma in primary hyperaldosteronism patients compared to those without primary hyperaldosteronism 105. These findings suggest that aldosterone dysregulation may contribute to the development of normal tension glaucoma, independent of blood pressure, and highlight the need for further research on the potential neuroprotective effects of mineralocorticoid receptor antagonists in normal tension glaucoma patients with primary hyperaldosteronism 105.

Another recent study explored potential clinical links between normal tension glaucoma and Alzheimer’s disease, focusing on shared neurodegenerative mechanisms 106. Both normal tension glaucoma and Alzheimer’s disease are progressive conditions, sharing risk factors such as age, female sex, and vascular dysfunction. Neuroimaging studies reveal that normal tension glaucoma may have cerebral manifestations similar to Alzheimer’s disease, further suggesting common mechanisms 106. Moreover, biomarkers like amyloid beta (Aβ) and tau proteins, traditionally linked with Alzheimer’s disease, have been implicated in normal tension glaucoma, indicating overlapping pathological processes. While connections between the two diseases remain debated, understanding normal tension glaucoma as part of a broader neurodegenerative spectrum may enhance both diagnostics and treatments for normal tension glaucoma, potentially offering insights into Alzheimer’s disease pathogenesis. Further research is needed to elucidate the exact relationship between normal tension glaucoma and Alzheimer’s disease 106.

Risk factors for normal tension glaucoma

Risk factors for normal tension glaucoma include 1, 107, 2, 108, 109, 110, 78:

  • Over 40 years of age
  • Family history of glaucoma
  • Female gender
  • Asian
  • High Myopia
  • Above-average intraocular pressure (IOP)
  • Thin central corneal thickness
  • Systemic hypertension
  • Nocturnal hypotension
  • Migraine
  • Raynaud phenomenon
  • Primary vascular dysfunction also called Flammer syndrome where the body’s blood vessels react abnormally to stimuli like cold or emotional stress 111. It’s associated with a cluster of symptoms and signs that can be present in both healthy individuals and those with various diseases, particularly normal-tension glaucoma.
  • Frontotemporal dementia and Alzheimer disease
  • Obstructive sleep apnea (OSA).

Normal tension glaucoma prevention

Due to the irreversible loss of vision due to normal tension glaucoma, early detection and treatment is important. Screening for and treatment of risk factors associated with normal tension glaucoma, such as nocturnal hypotension, currently does not have a defined role in primary prevention of normal tension glaucoma, particularly in the United States 41. A recent study that took place in China aimed to evaluate the effectiveness of a glaucoma screening program in identifying early-stage glaucoma cases 112. They compared 76 patients identified through glaucoma screening program with 272 consecutive outpatient cases from the same hospital. The findings indicate that patients detected through the screening program had significantly lower intraocular pressure (IOP) and were more likely to have normal tension glaucoma 112. These screening-detected patients also had less visual impairment and better visual field test results compared to clinic patients 112. The study suggests that health examination center-based glaucoma screening is effective in detecting early-stage glaucoma, especially those with normal tension glaucoma, and can complement opportunistic glaucoma detection 112. This is important in a country like China, where glaucoma is a significant public health concern. Further studies must take place to further characterize the role of primary prevention/screening for normal tension glaucoma before it potentially develops into standard practice, particularly in the US where the incidence of normal tension glaucoma is significantly less than in China 41. Nevertheless, the Chinese study demonstrates a potential role for glaucoma screening in patients who are particularly high risk of developing glaucoma.

Normal tension glaucoma signs and symptoms

Most patients with normal tension glaucoma in the early stages have no symptoms of the condition 113. There is no pain and vision seems normal with suspicion of glaucoma raised only by an optometrist during a routine eye testing or an incidental finding with an ophthalmologist 113, 114.

Even with moderately advanced disease, patients may be unaware of field defects because of unilateral disease, negative scotoma, and gradual onset 1. Because the intraocular pressure (IOP) is normal, suspicion is usually roused by optic disc appearance or a visual field defect on automated perimetry 1. If the presentation is advanced, patients may have symptoms of reduced vision, difficulty with low-contrast situations, and awareness of visual field defects. They may experience glare and difficulty adjusting to extreme lighting conditions 1. A family history of glaucoma and blindness should be obtained. Past medical history should include assessing risk factors for glaucoma, such as the history of steroid use, ocular trauma or surgery, and contraindications to treatments, including allergies. Medication usage should be reviewed.

A relative afferent pupillary defect is typical, though it may not be present in the early or symmetrical disease 1. Color vision is usually preserved, except in advanced disease. By definition, the intraocular pressure (IOP) is in the normal range 115. Slit-lamp examination and gonioscopy are essential to determine an open iridocorneal angle status and exclude secondary glaucoma causes. In particular, evidence of angle closure, uveitis, pigment dispersion, and pseudoexfoliation syndrome should be sought, as these are common causes of glaucoma presenting with an intraocular pressure (IOP) in the normal range.

A dilated fundus examination revealed changes in the glaucomatous optic disc. There is a progressive loss of ganglion cell neurons, leading to enlargement of the cup-to-disc ratio. This may be a focal (notch, retinal nerve fiber layer (RNFL) defect) or concentric defect (excavation, senile sclerotic disc) 1. Disc pallor occurs in advanced disease. Measurement of the optic disc size can help identify hypoplasia, physiological disc cupping, and disc asymmetry 1. Optic disc hemorrhages are more common in normal tension glaucoma than in primary open angle glaucoma (POAG) 108. So-called Drance hemorrhages are typically small flame hemorrhages at the disc margin in superior or inferior quadrants. Peripapillary atrophy may be seen but is non-specific. Glaucomatous disc abnormalities typically precede visual field defects in early (preperimetric) disease 116.

Normal tension glaucoma diagnosis

The only sure way to diagnose glaucoma is with a complete eye exam. An eye specialist can diagnose glaucoma using an eye exam, including several tests that are part of routine eye exams. A comprehensive eye exam can detect glaucoma long before you have eye damage and the symptoms that follow. Many of these tests involve pupil dilation (mydriasis), so your eye doctor can get a better look inside your eye. Your eye care specialist examines your eyes using a special magnifying lens. This provides a clear view of important tissues at the back of your eye to check for glaucoma or other eye problems. For a few hours after the exam your vision may be blurry and sensitive to light, so you will need someone to take you home.

Some of the most helpful glaucoma tests include:

  • Visual acuity testing. A visual acuity test assesses how clearly someone can see at a distance, typically using a Snellen chart or other standardized chart. The test is performed by an optometrist or ophthalmologist and involves reading progressively smaller letters or identifying shapes, with the results expressed as a fraction like 20/20 or 6/6, indicating the distance at which the person can see the letters or shapes
  • Visual field testing also called perimetry. This check of your peripheral (side) vision allows your eye care provider to find out how well you can see objects off to the side of your vision without moving your eyes. This test measures the entire area the forward-looking eye sees to document straight-ahead (central) and side (peripheral) vision. It measures the dimmest light seen at each spot tested. Each time patients perceive a flash of light, they respond by pressing a button.
  • Depth perception testing. A depth perception test assesses your ability to see the world in three dimensions (3D) and judge distances accurately. It checks if your eyes work together and if your brain processes the visual information correctly. These tests use 3D images or patterns like the Randot Stereo test to gauge how well your eyes coordinate to perceive depth. Some tests involve holding a finger in front of your eyes and focusing on a distant object, checking for double vision of the finger.
  • Tonometry. This measures the pressure inside your eye. Increased eye pressure is the most important risk factor for glaucoma. There are several methods of measuring eye pressure. The most common method is known as applanation, in which a tiny instrument contacts the eye’s surface after it is numbed with an eye drop.
    • Air-puff test. You’ll rest your chin on a machine and your eye specialist will blow a puff of air into your eye. This quick and painless test is used as part of a routine glaucoma screening. If the results show that your eye pressure is high, your eye specialist will do other eye-pressure tests to get a more accurate measurement.
    • Applanation tonometry. Your eye specialist will numb your eyes with drops before measuring your eye pressure using one of these methods:
      • You’ll rest your chin on a special magnifying device called a slit lamp. Your eye care specialist will examine your eye through the slit lamp while gently pressing a special tool on your eye to test the pressure.
      • Your eye care specialist will gently press a handheld device against your eye. The device measures your eye pressure.
  • Pachymetry. Pachymetry is a simple, painless test that measures the thickness of the cornea, the clear front part of the eye. The eye doctor uses an ultrasonic wave instrument to help determine the thickness of the cornea and better evaluate eye pressure.
  • Ophthalmoscopy. Your eye care specialist will do a dilated eye exam to look for damage to your optic nerve. This exam is part of a routine glaucoma check-up. You’ll be given eye drops that widen (dilate) your pupils (the openings that let light into your eyes). You’ll look straight ahead while your eye care specialist looks into your eye using a device with a light and magnifying lens.
  • Slit lamp exam. A slit lamp exam is a common eye test that uses a microscope with a focused beam of light to examine the front of your eye and the back of your eye with the aid of special lenses.
  • Gonioscopy. Gonioscopy is a specialized eye examination that allows an ophthalmologist to visualize the anterior chamber drainage angle, the space between the iris and the cornea where fluid drains out of the eye. Gonioscopy is a crucial part of diagnosing and monitoring glaucoma and other eye conditions. Eye doctors regularly examine the drainage angle to see if there is any visible obstruction to fluid leaving the eye through the trabecular meshwork. A special lens (gonioscopy lens) is needed to examine the trabecular meshwork. The gonioscopy lens is gently placed against the surface of the cornea and allows eye doctors to see the trabecular meshwork in the drainage angle.

If your eye specialist has a reason to suspect damage to your retina and/or optic nerve, they may also use additional types of eye imaging. These include:

  • Optical coherence tomography (OCT). Optical Coherence Tomography (OCT) measures the reflection of laser light similar to the way that ultrasound measures the reflection of sound. Using this device, a 3D reconstruction of the optic nerve can be created. Optical coherence tomography (OCT) is valuable for monitoring morphological changes in the optic nerve and retinal nerve fiber layer, especially in patients with ocular hypertension and early-to-moderate glaucoma 117. The most recent advances of OCT include OCT-A, or OCT-Angiography, whereby the blood flow to vessels surrounding the optic nerve and in the macula can be measured. This is still an active area of research, but scientists do know that some patients’ optic nerves are very vulnerable to changes in optic nerve blood flow, and this new measurement may be useful in evaluating these patients.
  • Heidelberg Retina Tomograph (HRT): Heidelberg Retina Tomograph (HRT) is also a laser that can produce a 3D representation of the optic nerve.
  • Nerve Fiber Analyzer (GDx): Nerve Fiber Analyzer (GDx) uses laser light to measure the thickness of the nerve fiber layer.
  • Fluorescein angiography. Fluorescein angiography is a diagnostic test used to examine the blood vessels in the retina and choroid of the eye. Fluorescein angiography involves injecting a fluorescent dye into the bloodstream and taking photographs of your retina and its blood vessels as the dye circulates, revealing potential blockages, leaks, or other abnormalities in the blood vessels. Fluorescein angiography is often recommended to find and diagnose eye disease including 118:
    • macular edema (swelling in the retina that distorts vision)
    • diabetic retinopathy (damaged or abnormal blood vessels in the eye caused by diabetes)
    • macular degeneration
    • blockage of veins inside the eye, called branch retinal vein occlusion (BRVO) or central retinal vein occlusion (CRVO)
    • macular pucker (a wrinkle in the retina caused by a buildup of fluid behind it)
    • ocular melanoma (a type of cancer affecting the eye)
    • rack changes in eye disease over time
    • target treatment areas
  • Less commonly, ultrasound, computed tomography (CT) or magnetic resonance imaging (MRI).

Normal tension glaucoma clinical diagnosis

Normal tension glaucoma is a diagnosis made based on similar criteria to primary open angle glaucoma (POAG) but with important clinical features that include:

  • Progressive excavation or ‘cupping’ of the optic nerve head from retinal nerve fiber layer loss resulting in corresponding visual field deficits.
  • Gonioscopic confirmation of open anterior chamber angle and absence of findings consistent with pigment dispersion or pseudoexfoliation syndrome.
  • Pre-treatment intraocular pressure (IOP) must always be less than 22 mm Hg. (diurnal measurement of intraocular pressure (IOP) to ensure there is not a circadian elevation in pressure that is missed by single period clinical measurement)

The diagnosis of normal tension glaucoma is only reached once other forms of optic neuropathy have been ruled out (e.g. ischemic, traumatic, toxic inflammatory, infectious, congenital, and compressive).

Careful history taking should be undertaken to elucidate any prior events that can mimic normal tension glaucoma such as:

  • Traumatic injuries
  • Inflammation
  • Severe blood-loss or hypotensive events
  • Medications that may precipitate a transient pathologic elevation in intraocular pressure (IOP)

After reasonable exclusion of all other causes, the demonstration of visual field loss on static, or less often kinetic, perimetry in conjunction with characteristic optic nerve central cupping with no elevation in intraocular pressure (IOP) above 21 mm Hg cement the diagnosis of normal tension glaucoma.

Other classically associated examination findings with normal tension glaucoma that can be helpful clues in raising suspicion for pursuing a diagnosis include optic nerve or “Drance” hemorrhages (also called optic disc hemorrhage) and peripapillary atrophy. While these findings are not specific, patients with normal tension glaucoma have a higher propensity for optic nerve hemorrhages compared to patients with primary open angle glaucoma (POAG). Focal defects in the retinal nerve fiber layer may be more commonly observed as well.

Visual field testing

Automated static perimetry is the most common modality used to detect and monitor for progression of the field loss associated with normal tension glaucoma 41. Visual field defects may include those common to primary open angle glaucoma (POAG) including nasal step and arcuate scotoma. However, defects noted in normal tension glaucoma tend to be more focal and occur closer to fixation early in the disease (Figure 4A and B). Dense paracentral scotomas may characteristically be noted at initial diagnosis.

Optic disc imaging

Visual field testing is useful in early detection but may miss early, pre-perimetric disease, as substantial retinal nerve fiber layer may be lost before functional field defects are noted 41. Therefore, optic disc imaging is an important and objective structural assessment of the optic nerve health 41. For several decades, the gold standard for detecting disease and monitoring changes in the optic nerve head has been stereo disc photography (Figure 3 A and B). In recent years, scanning laser ophthalmoscopy and optical coherence tomography (OCT) is gaining popularity as another means of detecting pathologic thinning of neural tissue and monitoring progression 41. Furthermore, with the introduction of Artificial Intelligence (AI), OCT interpretation continues to become more prevalent when classifying an eye as either a glaucoma suspect or early normal tension glaucoma 41. This was demonstrated in a recent study where a deep-learning algorithm was developed to discriminate between the normal tension glaucoma by using the parameter of Bruch’s membrane opening — minimum rim width, peripapillary retinal nerve fiber layer (RNFL) thickness and color classification of retinal nerve fiber layer (RNFL) — achieving an area under the curve of 0.966 119. Ultimately, these advances continue to improve the diagnostic specificity associated with either normal tension glaucoma or primary open angle glaucoma (POAG) 119.

A recent meta-analysis investigated differences in peripapillary choroidal thickness (PPCT) between primary open angle glaucoma (POAG), normal tension glaucoma, and healthy eyes 120. A systematic review of 18 studies, including 935 healthy control eyes, 446 normal tension glaucoma eyes, and 934 POAG eyes, was performed 120. OCT revealed significant reductions in peripapillary choroidal thickness (PPCT) in both POAG and normal tension glaucoma eyes compared with healthy eyes. POAG eyes demonstrated a mean reduction in peripapillary choroidal thickness (PPCT) of −16.32 µm compared to healthy controls, while normal tension glaucoma eyes showed a larger reduction of −34.96 µm compared to controls 120. Additionally, normal tension glaucoma eyes exhibited significantly thinner peripapillary choroidal thickness (PPCT) compared with POAG eyes, with a mean difference of −26.64 µm 120. These findings suggest that glaucomatous eyes, especially normal tension glaucoma eyes, have significantly reduced peripapillary choroidal thickness (PPCT), highlighting the potential role of peripapillary choroidal thickness (PPCT) as a diagnostic and monitoring tool in glaucoma management 120.

Pachymetry

Assessment of central corneal thickness (CCT) through pachymetry is essential in the work up of normal tension glaucoma 41. The measured intraocular pressure (IOP) by applanation may be artifactually low in eyes with low central corneal thickness (CCT) 41. Many patients with a diagnosis of normal tension glaucoma will demonstrate a low central corneal thickness (CCT) 121, 122. In some cases, correction of this under measurement may reveal an actual intraocular pressure (IOP) more consistent with primary open angle glaucoma (POAG) 121, 122.

Neurological Evaluation

At times, the diagnosis of normal tension glaucoma may simulate other neurological conditions. Most concerning to the clinician is an intracranial tumor masquerading as normal tension glaucoma. While these diagnoses are rare, clinicians should maintain a low threshold for neuroimaging with CT or MRI and a full neurological evaluation whenever the following exist 41:

  • Marked asymmetry or unilateral optic nerve involvement
  • Unexplained visual acuity loss
  • Color vision deficits in the absence of visual field deficits
  • Visual field defects not corresponding or out of proportion to optic nerve damage
  • Vertically aligned visual field defects
  • Atypical neurologic symptoms for glaucoma
  • Optic nerve pallor in excess of cupping
  • Age less than 50 years

Normal tension glaucoma treatment

Management of normal tension glaucoma mirrors the medical and surgical management of the other forms of glaucoma and hinges on reduction of intraocular pressure (IOP) from baseline 41. Identification of patients with clinical evidence of progression is important in the decision to initiate treatment for normal tension glaucoma. The natural history of normal tension glaucoma does not always include progression without treatment. As initially described by Drance 40, a significant portion of patients with normal tension glaucoma may not demonstrate clinical progression regardless of treatment.These patients typically had a history of systemic vascular compromise resulting in a one-time insult to the optic nerve. Nonetheless, for the majority of patients with normal tension glaucoma, reduction of intraocular pressure (IOP) remains the focus of treatment.

The Collaborative Normal-Tension Glaucoma Study (CNTGS) 7784 demonstrated the benefit of intraocular pressure (IOP) reduction for the treatment of patients with normal tension glaucoma. The Collaborative Normal-Tension Glaucoma Study (CNTGS) concluded that a 30 percent reduction in baseline intraocular pressure (IOP) resulted in a reduced risk of disease progression 77, 84. Criteria for initiation of treatment of the normal tension glaucoma patients in this study were defined as: documented visual field or optic nerve progression, visual field loss threatening fixation, or presence of disc hemorrhage 77, 84. The treatment group had a 12% risk of progression at 5 years compared to 35% progressing in the non-treatment group 84. The Collaborative Normal-Tension Glaucoma Study (CNTGS) was therefore instrumental in demonstrating the role of IOP in the pathogenesis of normal tension glaucoma and the benefit of treatment to lower it. The Collaborative Normal-Tension Glaucoma Study (CNTGS) also presents a reasonable goal for treatment in 30% intraocular pressure (IOP) reduction from patient’s baseline. Treatment intraocular pressure (IOP) goals may then be modified over the course of treatment to a level that sufficiently prevents or slows progression of disease.

Outside of intraocular pressure (IOP) lowering therapy, other aspects should be considered in the management of normal tension glaucoma patients. This may include cardiovascular problems such as systemic hypotension, nocturnal hypotension, anemia, and cardiac arrhythmias that can compromise optic nerve head perfusion 41. Consultation with primary care physicians can be helpful in addressing these concerns, but limited evidence is available to confirm a treatment benefit for normal tension glaucoma 41.

Medications

Topical intraocular pressure (IOP) lowering medications including prostaglandin analogues, alpha-2 agonists, beta-blockers, carbonic anhydrase inhibitors, and more recently Rho-kinase inhibitors are the mainstays of normal tension glaucoma medical therapy 41. Medications should be chosen on an individual basis to provide treatment that achieves a sufficient intraocular pressure (IOP) reduction with minimal side effects and ease of administration. Medication choice should also be cost-effective for the patient based on their resources.

Particularly with normal tension glaucoma, the effect of medications on systemic blood pressure, heart rate, and optic nerve perfusion should be considered. Furthermore, medications that have neuroprotective or intraocular pressure (IOP) independent effects would be extremely beneficial and remain an ongoing search. The Low-Pressure Glaucoma Study (LoGTS) demonstrated the importance of intraocular pressure (IOP) independent factors when choosing medical therapy for normal tension glaucoma 123. In the Low-Pressure Glaucoma Study (LoGTS), patients with low-tension glaucoma were randomized to treatment with either brimonidine tartrate 0.2% or timolol maleate 0.5% 123. While intraocular pressure (IOP) reduction was similar between the two treatment groups, patients treated with brimonidine were less likely to have visual field progression compared to patients treated with timolol 123. It is unclear whether this difference is due to an additional neuroprotective effect of brimonidine or a detrimental vascular effect from timolol 123. Moreover, Rho-kinase inhibitors are thought to be neuroprotective and increase vascular flow at the optic nerve head via the nitric oxide pathway 124, 125. The newer class of Rho-kinase inhibitor showed efficacy in both intraocular pressure (IOP) reduction in normal tension glaucoma and as add-on treatment in normal tension glaucoma patients with inadequate baseline intraocular pressure (IOP). The Rho-kinase (ROCK) inhibitor class of medication blocks the contraction of trabecular meshwork cells and increases the outflow of aqueous humor, thereby reducing intraocular pressure (IOP) 126, 127.

In patients with evidence of vasospasm, calcium channel blockers have been proposed to stabilize vascular tone, particularly in patients with concurrent hypertension, though the benefit has not been evaluated in large clinical trials 41.

Medical follow up

Once medical treatment is initiated, patients should be followed up 6-8 weeks later to ensure good adherence, minimal side effects, and adequate intraocular pressure (IOP) lowering efficacy 41. Different medication classes, laser treatment or surgical therapy may be trialed until an appropriate treatment is found 41. Once treatment goals have been met, periodic measurement of IOP during medical therapy is recommended every 3-4 months to ensure maintenance of goal intraocular pressure (IOP) and absence of progression 41. New technology has allowed for patients to partake in home tonometry. Patients can frequently report their findings to their supervising ophthalmologists allowing for a more complete representation of intraocular pressure (IOP) mean, peak and range (numerous measurements can be taken throughout the day) and closer follow-up 41. In addition to intraocular pressure (IOP), patients should be monitored for signs of progression by periodic assessment of the optic nerve head (disc photos, HRT, OCT, etc.) and visual field testing every 6-12 months, with more frequent intervals in advanced or actively progressing disease 41. If progression is detected despite goal intraocular pressure (IOP), treatment goals should be lowered with advance of therapy to achieve them 41.

Surgery

Laser and surgical treatment options for normal tension glaucoma mirror those for primary open angle glaucoma (POAG) 41. These include laser trabeculoplasty, minimally invasive glaucoma surgery (MIGS), trabeculectomy, and glaucoma drainage devices.

Selective Laser trabeculoplasty (SLT) may be a useful moderately invasive treatment with or without medical therapy 41. There is some literature that supports an IOP lowering, and decreased IOP variability, effect of Selective Laser trabeculoplasty (SLT) in normal tension glaucoma patients 128. For patients with IOP targets that are not achievable with medical/laser therapy, filtration surgery with or without a drainage device has traditionally been the mainstay of surgically lowering IOP 41. However, recent trends and practice patterns according to the American Academy of Ophthalmology Intelligent Research Insight Registry (IRIS) reveal a significant increase in the use of minimally invasive glaucoma surgery (MIGS) procedures from 2013-2018, and normal tension glaucoma is no exception 129.

Minimally invasive glaucoma procedures such as goniotomy with Kahook Dual Blade (KDB), the iStent trabecular bypass device, and the XEN gel stent have demonstrated an important although limited role in the surgical management of normal tension glaucoma 41. In theory, angle-based MIGS procedures can only lower IOP to a level equal to or above episcleral venous pressure of approximately 8-11 mm Hg 41. Goal IOP for normal tension glaucoma patients may be below this level making it difficult to achieve treatment goals by surgical means alone 41. However, maintaining a goal IOP with fewer medications is a reasonable indication for minimally invasive glaucoma surgery (MIGS) in normal tension glaucoma. This rationale also applies to laser trabeculoplasty, which augments aqueous outflow to the downstream episcleral venous system as well. According to American Academy of Ophthalmology Intelligent Research Insight Registry (IRIS) data regarding initial surgery for normal tension glaucoma, iStent has been the most common performed surgery and minimally invasive glaucoma surgery (MIGS) procedures in general are performed at a higher rate than filtration surgeries for normal tension glaucoma 129.

In the Collaborative Normal-Tension Glaucoma Study (CNTGS), the IOP reduction of 30% was only achieved in 57% of patients by topical medication and/or laser trabeculoplasty, while the remaining 43% required filtering surgery 41. While IOP lowering with filtration surgery has been shown to be effective in decreasing visual field progression a continued, slowed progression has been reported in postoperative patients followed for up to 6 years 41.

A lower starting IOP with normal tension glaucoma patients and a 30% reduction target may result in a narrower margin between therapeutic IOP reduction and hypotony in these patients 41. Increase risk of filtering surgery complications has been reported in this subset of primary open angle glaucoma (POAG) patients 41.

Selection of anti-metabolite drugs and means of application in filtering surgery is an important consideration and should be guided by specific treatment goals and surgeon specific experience with these agents 41. Mitomycin C (MMC) has been associated with achievement of lower IOPs post operatively compared to 5-fluorouracil (5-FU) in some studies, though literature also suggests equivalence of efficacy of these two agents in primary trabeculectomy for primary open angle glaucoma (POAG) 41. Mitomycin C use in normal tension glaucoma glaucoma has an associated increased risk of over-filtration complications that may play a role in the risk of visual field progression. Therefore, meticulous use of mitomycin C (e.g. 0.2-0.4 mg/ml for 1-3 minutes), careful flap suturing, and judicious use of viscoelastic with frequent postoperative follow up have been proposed as methods to mitigate the risks of hypotony early in the post operative phase while achieving target IOP 41. Early suture lysis may be required to achieve low target IOPs but should be weighed against the risk of resultant over-filtration. Given the context of a higher risk of hypotony following filtering surgery in patients with normal tension glaucoma, the XEN gel stent has recently been deployed to achieve a lower IOP goal while maintaining a lower rate of hypotony 41. One recent study demonstrated a mean IOP decrease of 5.6 +/- 2.7 mmHg in normal tension glaucoma patients, which represented an IOP reduction of 29% 130. Also, the use of the EX-PRESS glaucoma mini shunt has been employed as a means of preventing complications, while still achieving similar IOP goals compared to standard trabeculectomy 41. In one experience, the smaller, consistent outflow opening of the EX-PRESS shunt allows for earlier suture lysis to achieve low IOP targets with less risk of hypotony 41. The utility of this device remains an area of debate considering the additional cost of the device and mixed outcomes in the literature 41.

Cyclodestructive procedures provide the only surgical means of suppressing aqueous production to lower IOP. Due to their potentially vision threatening side effects, these procedures are typically reserved for eyes refractory to treatment or with poor visual potential 41. Ablation of the ciliary processes may be accomplished by transscleral cyclophotocoagulation (CPC) or by endoscopic cyclophotocoagulation (ECP) 41. Endoscopic cyclophotocoagulation (ECP) offers the unique advantage of direct visualization of the target tissue allowing a more targeted approach to achieve less inflammation and side effects 41.

A recent systematic review and meta-analysis evaluated the efficacy of angle-based minimally invasive glaucoma surgery (MIGS) in patients with normal tension glaucoma 131. The study analyzed outcomes from 15 studies, totaling 367 normal tension glaucoma eyes, with procedures including the iStent, iStent inject, Hydrus Microstent, Kahook Dual Blade, and Trabectome 131. The review found significant reductions in IOP and glaucoma medication usage postoperatively 131. Specifically, combined phacoemulsification and angle-based MIGS showed a mean IOP reduction of 2.44 mmHg at 6 months, 2.28 mmHg at 12 months, and sustained reductions up to 36 months 131. Glaucoma medication usage was also significantly reduced by 1.21 medications at 6 months and 1.18 at 12 months postoperatively. These findings suggest that angle-based MIGS, particularly in combination with cataract surgery, can be effective in reducing IOP and medication burden in normal tension glaucoma patients while maintaining a favorable safety profile 131.

Normal tension glaucoma prognosis

Like any form of glaucoma, normal tension glaucoma may progress to irreversible blindness, but is dependent on factors that include the disease severity at diagnosis, effectiveness of treatment, individual risk factors, overall ocular health and your general health 132, 133, 134, 135, 136, 1, 41. Similar to other types of glaucoma, normal tension glaucoma can progress to irreversible unilateral or bilateral blindness in the worst cases, even despite therapy 133. The prognosis for visual preservation is good in patients who undergo adequate treatment through intraocular pressure (IOP) reduction 41. The main risk factors associated with the normal tension glaucoma disease progression in both treated and untreated patients have been demonstrated to be female gender, greater variation in diurnal IOP and diastolic blood pressure, presence of disk hemorrhage, greater vertical cup/disc ratio and migraine at baseline 31. Age, mean IOP and baseline IOP were not shown to be risk factors for progression 31. Epidemiology studies have shown that Asians tend to show a slower rate of disease progression 132, 134, 135, 136. On average, the visual field damage progression has been reported to be slower in normal tension glaucoma than in primary open angle glaucoma (POAG), but with higher inter-patient variability 135.

The cumulative risk to develop legal unilateral blindness in treated normal tension glaucoma patients under standard ophthalmic care has been calculated to be 5.8% and 9.9% at 10 years and 20 years, respectively 31. The risk for bilateral blindness at 10 and 20 years was 0.3% and 1.4%, respectively 133. Patients presenting advanced damage at the diagnosis or rapidly progressing visual field loss, the so-called “rapid progressors”, are most likely to become blind due to the disease 31. It is fundamental that these patients need to be identified and managed with more aggressive treatment to avoid irreversible visual loss 133.

In the Collaborative Normal-Tension Glaucoma Study (CNTGS), 65% of patients in the control group with normal tension glaucoma did not progress even without treatment 84, 77. However, an intraocular pressure (IOP) reduction of 30% with treatment further lowered the likelihood of progression to only 12% 84, 77. The Early Manifest Glaucoma Trial (EMGT), a randomized double-masked clinical trial conducted on 255 open-angle glaucoma patients with early visual field defects, including 53% of normal tension glaucoma cases, showed that a 25% reduction in IOP can reduce the risk of disease progression to 45% in the study group, compared to 62% in the control group, after a 6-year follow-up 57. These studies have suggested that, to be significantly effective in decreasing the risk of normal tension glaucoma progression, the reduction in IOP should be at least 30% or more 137, 56. Moreover, studies with long follow-ups have shown that the amount of IOP reduction seems to be directly related to the reduction in the visual field progression rate in normal tension glaucoma patients 59. Additionally, the benefits of the IOP reduction in normal tension glaucoma patients seem to be significantly higher in females with migraine and a family history of glaucoma, without disk hemorrhages, family history of stroke and personal history of cardiovascular disease 134.

Patients with normal tension glaucoma that previously suffered an acute vascular compromise have also been shown to not progress over time as well 40. Given this relatively high rate of non-progression, some clinicians have suggested a conservative “wait and see” approach to initiating treatment 41. This recommendation should be cautioned, as it is often difficult to determine which patients will progress, and other studies have shown variable rates of progression in this disease 41. Risk factors for progression of visual field defects in normal tension glaucoma include migraine, disc hemorrhage, and female gender. Asians have been shown to have a slower rate of progression 110.

Pigmentary glaucoma

Pigmentary glaucoma is a type of secondary open-angle glaucoma where small pigment granules from the back of the iris breaks off and clogs the eye’s drainage system at the angle where the iris and cornea meet, leading to increased intraocular pressure (IOP) and potential optic nerve damage 138, 139, 140, 141, 142, 143. Pigment gives your iris its color. Pigment dispersion syndrome (PDS) is characterized by the presence of melanin pigment granules that circulate within the aqueous humor and accumulate on different structures found in the anterior chamber of the eye, including corneal endothelium deposits also known as the Krukenberg spindle, lens surface, zonules, iris, iridocorneal angle, and trabecular meshwork 144. The pigment granules that accumulate in the angular structures and trabecular meshwork can give rise to reduced aqueous outflow, leading to elevated intraocular pressure (IOP). Pigment dispersion syndrome (PDS) can cause ocular hypertension and can lead to secondary rise in intraocular pressure (IOP) that can damage the optic nerve resulting in pigmentary glaucoma development, which is a secondary type of glaucoma. Pigment dispersion syndrome (PDS) and pigmentary glaucoma in reality reflect different levels of severity on a continuum disease spectrum 142. Pigment dispersion syndrome (PDS) is used to classify individuals who exhibit these features but who have not progressed to optic nerve damage and/or visual field loss (signifiers of pigmentary glaucoma), even if the intraocular pressure (IOP) is elevated. Pigment dispersion syndrome (PDS) is an important risk factor in the development of ocular hypertension and pigmentary glaucoma 143.

Pigmentary glaucoma is a result of pigment dispersion syndrome (PDS), where pigment granules detach from the iris and accumulate in the trabecular meshwork, which is the main drainage channel of the eye. Pigment dispersion syndrome (PDS) happens when the pigment rubs off the back of your iris. The pigment then floats around to other parts of your eye. The tiny bits of pigment can clog your eye’s drainage angle. This pigment can raise eye pressure and lead to pigmentary glaucoma. Features of pigment dispersion syndrome (PDS) include anterior chamber pigment dispersion, spoke-like iris defects on trans-illumination, central corneal endothelial deposits also known as the Krukenberg spindle, and increased pigmentation in the iridocorneal angle 142. However, not everyone who has pigment dispersion syndrome (PDS) will develop pigmentary glaucoma. Approximately 15% of patients with pigment dispersion syndrome (PDS) will convert to pigmentary glaucoma after 15 years 138. Some people with pigment dispersion syndrome (PDS) or pigmentary glaucoma may see halos or have blurry vision after activities like jogging or playing basketball that stir up the pigment granules. See your ophthalmologist if you have these or other symptoms.

The iris of your eye is a flat ring of muscle that contains melanin, the pigment that gives your eyes their color. In front of and behind your iris are spaces filled with a fluid called aqueous humor. Pressure from the aqueous humor helps your eye hold its globe-like shape. When you have pigment dispersion syndrome (PDS), your iris can’t hold its shape and dips back too far 145, 146. That makes the back of your iris press against the muscle fibers that control your lens shape. As your iris widens or narrows, the iris rubs against those fibers and pigment granules in the iris wear away, like flakes of paint chipping away from a piece of wood 145, 146. Patients with pigment dispersion syndrome or pigmentary glaucoma have a 15-fold higher concentration of aqueous pigment granules in their anterior chamber compared to normal controls 147.

The release of pigment showers in the anterior chamber is mainly due to the friction and rubbing between the iris pigment epithelium and posterior surface and zonules of the lens, which is favored by the backward posterior bowing of the iris that can be found in moderate myopic eyes that have more space 148 and by reverse pupillary block mechanisms due to increased iridolenticular touch 149. Ultrasonographical studies have shown that events leading to increased friction and contact between anterior chamber structures that favor reverse pupillary block include physiological events such as accommodation, blinking, eye movements, head positions, and exercise 150. The increased contact between the iris and lens structures in eyes at risk of having a deep anterior chamber and/or a large iris can create a ball-valve mechanism in certain conditions in which the aqueous humor moves from the posterior chamber to the anterior chamber in a unidirectional mode, thus creating a high pressure in the anterior chamber that favors further apposition between the iris and lens surface 151. The aqueous humor trapped in the anterior chamber can cause posterior bowing and further friction between the peripheral posterior iris and zonules and lens structures leading to pigment dispersion showers.

Once those pigment granules are loose and floating in the aqueous humor, the flow of fluid carries them to other places inside your eye. The aqueous humor has a drainage system, the trabecular meshwork, and granules can accumulate in that meshwork and damage it 152, 147, 153. When that happens, aqueous humor fluid can’t drain out of your eye properly, causing high pressure inside your eye (ocular hypertension) and eventually causing glaucoma 146, 154. Without treatment, glaucoma causes irreversible, severe vision loss and blindness.

It was originally thought that pigment dispersion syndrome (PDS) and pigmentary glaucoma had a congenital cause or may be inherited (passed from parent to child), due to pigment loss from the iris from congenital mesodermal dysgenesis 155 or atrophy or degeneration of the iris pigment epithelium (IPE) 156, 157. Possible genetic factors have been hypothesized to explain the familial presence of Krukenberg spindle 158. Although the low incidence of familial pigment dispersion syndrome and pigmentary glaucoma, studies have reported a possible autosomal dominant inheritance for pigment dispersion syndrome 159 and multifactorial pattern of inheritance 160, which may play a role in the clinical expression of factors related to iris color, gender, and refractive error. Anderson et al. 161 reported a possible gene responsible for pigment dispersion syndrome located on chromosome 7Q35-q36 based on an autosomal dominant pattern observed in patients from 4 Irish families with pigment dispersion syndrome. Studies have reported several genetic locus associated with pigment dispersion syndrome, which include Glycoprotein nmb (GpnmbR150x), Gene Gpigment dispersion syndrome1 (glaucoma-related pigment dispersion syndrome 1), and (OMIM ID 600510) 162.

There are other reasons why pigment granules might break free from your iris. When there’s another cause for pigment displacement, that’s called “secondary pigment dispersion syndrome”.

Some causes of secondary pigment dispersion syndrome include:

  • Eye injuries
  • Tumors or growths inside your eye
  • An artificial intraocular lens (IOL) that moves out of position

There are are several possible contributing factors for developing pigmentary glaucoma:

  • Genetics. Research connects several DNA mutations with pigment dispersion syndrome and pigmentary glaucoma. That’s why pigment dispersion syndrome and pigmentary glaucoma can run in families but it’s usually in an unpredictable way. Direct examination of a small set of family members of patients with pigment dispersion syndrome showed that the disease was present in 2 out of 19 related individuals (12%) 163. In another family, signs of pigment dispersion syndrome were present in 36% of subjects’ parents and 50% of their siblings (but in no children), suggesting a possible autosomal dominant inheritance pattern with incomplete penetrance 164, 165, 166. Pigmentary glaucoma or pigment dispersion syndrome has also been described in families across multiple generations, with roughly 50% of family members described as having either condition, reinforcing the idea of an autosomal dominant inheritance pattern 167, 168.
  • Male sex. Pigment dispersion syndrome and pigmentary glaucoma disproportionately affects males with case series showing a male to female ratio of between 2:1 and 5:1. Much less of a male predominance is noted for pigment dispersion syndrome, with case series describing male to female ratios between 1:1 and 2:1 169, 165, 170, 171, 172.
  • Age. Diagnosis of pigment dispersion syndrome generally happens sometime between ages 20 and 50. Male patients with pigmentary glaucoma or pigment dispersion syndrome most often present in their 30s, whereas female patients typically present roughly a decade later in life 165, 170, 171, 173, 174. However, cases of pigment dispersion syndrome have been identified in patients as young as 12–15 years of age 175, 176, 177. Pigment dispersion syndrome and pigmentary glaucoma may be most common in middle age once the lens has enlarged and the iris is flexible enough to form a concave position 145.
  • Race. People who are of Black or Asian descent have a lower risk for pigment dispersion syndrome and pigmentary glaucoma, while the risk for white people is higher 169, 170.
  • Being nearsighted. People with myopia (nearsightedness) have a higher risk of pigment dispersion syndrome and pigmentary glaucoma. The more nearsighted you are (typically in the range of -3 to -4 D), the higher the risk of having pigment dispersion syndrome or having it turn into pigmentary glaucoma.
  • Eye structure. Pigment dispersion syndrome and pigmentary glaucoma are more likely to happen when you have a deep anterior chamber. A deeper chamber means it can hold more fluid, which can make the iris “bow” backward toward the lens. Having flatter corneas can also be a contributing factor.
  • Concave iris and posterior iris insertion. Concave iris and more posterior iris insertion are more common in patients with pigment dispersion syndrome or pigmentary glaucoma than in the normal population and result in greater iridolenticular contact in these individuals 178, 179, 180.
  • Flat corneas. Patients with pigment dispersion syndrome and pigmentary glaucoma have significantly flatter corneas than control subjects of similar age and refractive error 181, 182. A flat cornea might be more likely to result in burping of aqueous humor from the posterior chamber to the anterior chamber with blinking, resulting in increased irido-zonular contact 182.
  • Excessive activity level. Research links pigment dispersion syndrome and pigmentary glaucoma to excessive exercise or physical activity. If you have pigment dispersion syndrome or pigmentary glaucoma, your eye specialist will likely ask about how physically active you are. If your activity level could contribute to your pigment dispersion syndrome or pigmentary glaucoma — either now or in the future — your eye specialist will likely recommend reducing your activity level.

The treatments for pigment dispersion syndrome and pigmentary glaucoma are very similar to treatments for other forms of glaucoma. The treatments can involve one or more of the following:

  • Medications. Glaucoma medications lower pressure inside your eye in different ways. Some of them cause your pupil to relax more, improving aqueous humor drainage. Others slow the production of aqueous humor.
  • Glaucoma surgery. This approach usually aims to improve fluid flow and drainage. Examples of glaucoma surgeries that do this include laser trabeculoplasty, and incisional surgery with either trabeculectomy or glaucoma drainage implant.

In many cases, a combination approach may offer the best results for people with pigment dispersion syndrome and pigmentary glaucoma. An example would be combining medications with laser surgery. This works well when medication alone doesn’t lower pressure inside your eye as much as you need.

Your eye specialist is your best source of information about treatment approaches. Your eye specialist can advise you on how treatments are likely to affect you, what your alternatives are and what kinds of side effects you can expect.

Figure 8. Krukenberg spindle

pigmentary glaucoma
Krukenberg spindle

Footnote: Slit lamp photograph showing a Krukenberg spindle visible on the corneal endothelium.

[Source 139 ]

Figure 9. Scheie stripe (pigment deposition along the insertion of the zonular fibers to the lens)

Scheie stripe
Scheie stripe
[Source 183 ]
When to see a doctor

Some eye symptoms mean you need medical attention quickly. Not getting treatment can lead to irreversible eye damage. You should talk to your eye specialist after your diagnosis to learn about the symptoms that are most relevant to your specific case and what you should do if you have any of these symptoms.

Symptoms that mean you need emergency medical attention include:

  • Sudden vision changes like blurring or low vision
  • Noticeable new or worsening halos or glare around lights
  • Sudden, severe eye pain and/or headaches
  • Sudden vision loss (either partial or total)

Pigmentary glaucoma causes

The iris of your eye is a flat ring of muscle that contains melanin, the pigment that gives your eyes their color. In front of and behind your iris are spaces filled with a fluid called aqueous humor. Pressure from the aqueous humor helps your eye hold its globe-like shape. When you have pigment dispersion syndrome (PDS), your iris can’t hold its shape and dips back too far 145, 146. That makes the back of your iris press against the muscle fibers that control your lens shape. As your iris widens or narrows, the iris rubs against those fibers and pigment granules in the iris wear away, like flakes of paint chipping away from a piece of wood 145, 146. Patients with pigment dispersion syndrome or pigmentary glaucoma have a 15-fold higher concentration of aqueous pigment granules in their anterior chamber compared to normal controls 147.

The release of pigment showers in the anterior chamber is mainly due to the friction and rubbing between the iris pigment epithelium and posterior surface and zonules of the lens, which is favored by the backward posterior bowing of the iris that can be found in moderate myopic eyes that have more space 148 and by reverse pupillary block mechanisms due to increased iridolenticular touch 149. Ultrasonographical studies have shown that events leading to increased friction and contact between anterior chamber structures that favor reverse pupillary block include physiological events such as accommodation, blinking, eye movements, head positions, and exercise 150. The increased contact between the iris and lens structures in eyes at risk of having a deep anterior chamber and/or a large iris can create a ball-valve mechanism in certain conditions in which the aqueous humor moves from the posterior chamber to the anterior chamber in a unidirectional mode, thus creating a high pressure in the anterior chamber that favors further apposition between the iris and lens surface 151. The aqueous humor trapped in the anterior chamber can cause posterior bowing and further friction between the peripheral posterior iris and zonules and lens structures leading to pigment dispersion showers.

Once those pigment granules are loose and floating in the aqueous humor, the flow of fluid carries them to other places inside your eye. The aqueous humor has a drainage system, the trabecular meshwork, and granules can accumulate in that meshwork and damage it 152, 147, 153. When that happens, aqueous humor fluid can’t drain out of your eye properly, causing high pressure inside your eye (ocular hypertension) and eventually causing glaucoma 146, 154. Without treatment, glaucoma causes irreversible, severe vision loss and blindness.

It was originally thought that pigment dispersion syndrome (PDS) and pigmentary glaucoma had a congenital cause or may be inherited (passed from parent to child), due to pigment loss from the iris from congenital mesodermal dysgenesis 155 or atrophy or degeneration of the iris pigment epithelium (IPE) 156, 157. Possible genetic factors have been hypothesized to explain the familial presence of Krukenberg spindle 158. Although the low incidence of familial pigment dispersion syndrome and pigmentary glaucoma, studies have reported a possible autosomal dominant inheritance for pigment dispersion syndrome 159 and multifactorial pattern of inheritance 160, which may play a role in the clinical expression of factors related to iris color, gender, and refractive error. Anderson et al. 161 reported a possible gene responsible for pigment dispersion syndrome located on chromosome 7Q35-q36 based on an autosomal dominant pattern observed in patients from 4 Irish families with pigment dispersion syndrome. Studies have reported several genetic locus associated with pigment dispersion syndrome, which include Glycoprotein nmb (GpnmbR150x), Gene Gpigment dispersion syndrome1 (glaucoma-related pigment dispersion syndrome 1), and (OMIM ID 600510) 162.

There are other reasons why pigment granules might break free from your iris. When there’s another cause for pigment displacement, that’s called “secondary pigment dispersion syndrome”.

Some causes of secondary pigment dispersion syndrome include:

  • Eye injuries
  • Tumors or growths inside your eye
  • An artificial intraocular lens (IOL) that moves out of position

Risk factors for developing pigmentary glaucoma

There are are several possible contributing factors for developing pigmentary glaucoma:

  • Genetics. Research connects several DNA mutations with pigment dispersion syndrome and pigmentary glaucoma. That’s why pigment dispersion syndrome and pigmentary glaucoma can run in families but it’s usually in an unpredictable way. Direct examination of a small set of family members of patients with pigment dispersion syndrome showed that the disease was present in 2 out of 19 related individuals (12%) 163. In another family, signs of pigment dispersion syndrome were present in 36% of subjects’ parents and 50% of their siblings (but in no children), suggesting a possible autosomal dominant inheritance pattern with incomplete penetrance 164, 165, 166. Pigmentary glaucoma or pigment dispersion syndrome has also been described in families across multiple generations, with roughly 50% of family members described as having either condition, reinforcing the idea of an autosomal dominant inheritance pattern 167, 168.
  • Male sex. Pigment dispersion syndrome and pigmentary glaucoma disproportionately affects males with case series showing a male to female ratio of between 2:1 and 5:1. Much less of a male predominance is noted for pigment dispersion syndrome, with case series describing male to female ratios between 1:1 and 2:1 169, 165, 170, 171, 172.
  • Age. Diagnosis of pigment dispersion syndrome generally happens sometime between ages 20 and 50. Male patients with pigmentary glaucoma or pigment dispersion syndrome most often present in their 30s, whereas female patients typically present roughly a decade later in life 165, 170, 171, 173, 174. However, cases of pigment dispersion syndrome have been identified in patients as young as 12–15 years of age 175, 176, 177. Pigment dispersion syndrome and pigmentary glaucoma may be most common in middle age once the lens has enlarged and the iris is flexible enough to form a concave position 145.
  • Race. People who are of Black or Asian descent have a lower risk for pigment dispersion syndrome and pigmentary glaucoma, while the risk for white people is higher 169, 170.
  • Being nearsighted. People with myopia (nearsightedness) have a higher risk of pigment dispersion syndrome and pigmentary glaucoma. The more nearsighted you are (typically in the range of -3 to -4 D), the higher the risk of having pigment dispersion syndrome or having it turn into pigmentary glaucoma.
  • Eye structure. Pigment dispersion syndrome and pigmentary glaucoma are more likely to happen when you have a deep anterior chamber. A deeper chamber means it can hold more fluid, which can make the iris “bow” backward toward the lens. Having flatter corneas can also be a contributing factor.
  • Concave iris and posterior iris insertion. Concave iris and more posterior iris insertion are more common in patients with pigment dispersion syndrome or pigmentary glaucoma than in the normal population and result in greater iridolenticular contact in these individuals 178, 179, 180.
  • Flat corneas. Patients with pigment dispersion syndrome and pigmentary glaucoma have significantly flatter corneas than control subjects of similar age and refractive error 181, 182. A flat cornea might be more likely to result in burping of aqueous humor from the posterior chamber to the anterior chamber with blinking, resulting in increased irido-zonular contact 182.
  • Excessive activity level. Research links pigment dispersion syndrome and pigmentary glaucoma to excessive exercise or physical activity. If you have pigment dispersion syndrome or pigmentary glaucoma, your eye specialist will likely ask about how physically active you are. If your activity level could contribute to your pigment dispersion syndrome or pigmentary glaucoma — either now or in the future — your eye specialist will likely recommend reducing your activity level.

Disease Progression

  • Intraocular pressure (IOP). A retrospective study from Olmstead County Minnesota found IOP > 21 mm Hg to be the only risk factor for progression from pigment dispersion syndrome to pigmentary glaucoma 165.
  • Degree of iridolenticular contact in patients with asymmetric disease. More iris–lens contact in one eye vs the other may be associated with greater risk of disease progression 180.
  • Greater trabecular meshwork pigmentation. In eyes with bilateral pigment dispersion syndrome, worse disease is typically found in the eye with more severe trabecular meshwork pigmentation.

Pigmentary glaucoma prevention

There’s nothing you can do to prevent pigment dispersion syndrome and pigmentary glaucoma. They happen unpredictably and people often don’t know they have these conditions until an eye specialist sees the changes during a routine eye exam or they start developing symptoms.

Pigmentary glaucoma signs and symptoms

Pigment dispersion syndrome (PDS) often doesn’t cause symptoms, so many people don’t know they have it. When it does, they’re usually similar to the most common glaucoma symptoms. Those include:

  • Eye pain
  • Eye redness
  • Light sensitivity (photophobia)
  • Blurred vision
  • Seeing a glare or halos around lights after exercise.

In time, as the optic nerve becomes more damaged, you may notice that blank spots begin to appear in your field of vision. You usually won’t notice these blank spots in your day-to-day activities until the optic nerve is significantly damaged and these spots become large. If all of the optic nerve fibers die, blindness results. That is why it is so important to have an eye exam with your eye doctor.

Pigmentary glaucoma diagnosis

The most useful test for detecting pigment dispersion syndrome before it causes symptoms is a routine eye exam.

Some of the changes that pigment dispersion syndrome or pigmentary glaucoma can cause that show up on an eye exam include:

  • Krukenberg spindle. This is a faint, vertical line in front of your pupil that’s the same color as your iris (see Figure 4 above). It happens when higher pressure inside your eye pushes fluid forward and keeps loose pigment granules pressed against the inside surface of your cornea.
  • Radial iris defects. These are thin lines on your iris with worn-away pigment. They look like the spokes on a bicycle wheel. An eye specialist can see them using special lighting techniques.
  • Pigment accumulation near the outer rim of your iris. Your eye specialist can use a technique called gonioscopy to see pigment granules deposited there.

Several glaucoma tests can help with diagnosing pigment dispersion syndrome and pigmentary glaucoma. Some of these are common features of a routine eye exam, while others are a little more specific. Your eye specialist can tell you which tests they think will be the most helpful for diagnosing or ruling out pigment dispersion syndrome and pigmentary glaucoma.

These tests often include one or more of the following:

  • Visual acuity testing. A visual acuity test assesses how clearly someone can see at a distance, typically using a Snellen chart or other standardized chart. The test is performed by an optometrist or ophthalmologist and involves reading progressively smaller letters or identifying shapes, with the results expressed as a fraction like 20/20 or 6/6, indicating the distance at which the person can see the letters or shapes
  • Visual field testing also called perimetry. This check of your peripheral (side) vision allows your eye care provider to find out how well you can see objects off to the side of your vision without moving your eyes. This test measures the entire area the forward-looking eye sees to document straight-ahead (central) and side (peripheral) vision. It measures the dimmest light seen at each spot tested. Each time patients perceive a flash of light, they respond by pressing a button.
  • Depth perception testing. A depth perception test assesses your ability to see the world in three dimensions (3D) and judge distances accurately. It checks if your eyes work together and if your brain processes the visual information correctly. These tests use 3D images or patterns like the Randot Stereo test to gauge how well your eyes coordinate to perceive depth. Some tests involve holding a finger in front of your eyes and focusing on a distant object, checking for double vision of the finger.
  • Tonometry. This measures the pressure inside your eye. Increased eye pressure is the most important risk factor for glaucoma. There are several methods of measuring eye pressure. The most common method is known as applanation, in which a tiny instrument contacts the eye’s surface after it is numbed with an eye drop.
    • Air-puff test. You’ll rest your chin on a machine and your eye specialist will blow a puff of air into your eye. This quick and painless test is used as part of a routine glaucoma screening. If the results show that your eye pressure is high, your eye specialist will do other eye-pressure tests to get a more accurate measurement.
    • Applanation tonometry. Your eye specialist will numb your eyes with drops before measuring your eye pressure using one of these methods:
      • You’ll rest your chin on a special magnifying device called a slit lamp. Your eye care specialist will examine your eye through the slit lamp while gently pressing a special tool on your eye to test the pressure.
      • Your eye care specialist will gently press a handheld device against your eye. The device measures your eye pressure.
  • Pachymetry. Pachymetry is a simple, painless test that measures the thickness of the cornea, the clear front part of the eye. The eye doctor uses an ultrasonic wave instrument to help determine the thickness of the cornea and better evaluate eye pressure.
  • Ophthalmoscopy. Your eye care specialist will do a dilated eye exam to look for damage to your optic nerve. This exam is part of a routine glaucoma check-up. You’ll be given eye drops that widen (dilate) your pupils (the openings that let light into your eyes). You’ll look straight ahead while your eye care specialist looks into your eye using a device with a light and magnifying lens.
  • Slit lamp exam. A slit lamp exam is a common eye test that uses a microscope with a focused beam of light to examine the front of your eye and the back of your eye with the aid of special lenses.
  • Gonioscopy. Gonioscopy is a specialized eye examination that allows an ophthalmologist to visualize the anterior chamber angle, the space between the iris and the cornea where fluid drains out of the eye. Gonioscopy is a crucial part of diagnosing and monitoring glaucoma and other eye conditions

If your eye specialist has a reason to suspect damage to your retina and/or optic nerve, they may also use additional types of eye imaging. These include:

  • Optical coherence tomography (OCT). Optical Coherence Tomography (OCT) measures the reflection of laser light similar to the way that ultrasound measures the reflection of sound. Using this device, a 3D reconstruction of the optic nerve can be created. Optical coherence tomography (OCT) is valuable for monitoring morphological changes in the optic nerve and retinal nerve fiber layer, especially in patients with ocular hypertension and early-to-moderate glaucoma 117. The most recent advances of OCT include OCT-A, or OCT-Angiography, whereby the blood flow to vessels surrounding the optic nerve and in the macula can be measured. This is still an active area of research, but scientists do know that some patients’ optic nerves are very vulnerable to changes in optic nerve blood flow, and this new measurement may be useful in evaluating these patients.
  • Heidelberg Retina Tomograph (HRT): Heidelberg Retina Tomograph (HRT) is also a laser that can produce a 3D representation of the optic nerve.
  • Nerve Fiber Analyzer (GDx): Nerve Fiber Analyzer (GDx) uses laser light to measure the thickness of the nerve fiber layer.
  • Fluorescein angiography. Fluorescein angiography is a diagnostic test used to examine the blood vessels in the retina and choroid of the eye. Fluorescein angiography involves injecting a fluorescent dye into the bloodstream and taking photographs of your retina and its blood vessels as the dye circulates, revealing potential blockages, leaks, or other abnormalities in the blood vessels. Fluorescein angiography is often recommended to find and diagnose eye disease including 118:
    • macular edema (swelling in the retina that distorts vision)
    • diabetic retinopathy (damaged or abnormal blood vessels in the eye caused by diabetes)
    • macular degeneration
    • blockage of veins inside the eye, called branch retinal vein occlusion (BRVO) or central retinal vein occlusion (CRVO)
    • macular pucker (a wrinkle in the retina caused by a buildup of fluid behind it)
    • ocular melanoma (a type of cancer affecting the eye)
    • rack changes in eye disease over time
    • target treatment areas
  • Less commonly, ultrasound, computed tomography (CT) or magnetic resonance imaging (MRI).

Pigmentary glaucoma treatment

Pigment dispersion syndrome treatment varies depending on how it is affecting your eye pressure (IOP):

  • For pigment dispersion syndrome with normal or only slightly elevated intraocular pressure (IOP), there is a low risk of damage to the optic nerve. No treatment is needed other than seeing your ophthalmologist one time each year. They will monitor your condition by checking your IOP and looking for any changes in your vision.
  • For pigment dispersion syndrome with elevated intraocular pressure (IOP), there is a greater risk of damage to the optic nerve. To lower IOP, you may be treated with medicated eye drops or laser therapy.
  • When intraocular pressure (IOP) from pigment dispersion syndrome is so high that it damages the optic nerve, this is then called “pigmentary glaucoma”. In this case, treatment is needed and it may be medicated eye drops, laser therapy, or surgery.

Eye drops

Glaucoma treatment often starts with prescription eye drops. Some may decrease eye pressure by improving how fluid drains from your eye. Others decrease the amount of fluid your eye makes. Depending on how low your eye pressure needs to be, more than one eye drop may be prescribed.

Prescription eye drop medicines include:

  • Prostaglandins. These increase the outflow of the fluid in your eye, helping to reduce eye pressure (IOP). Medicines in this category include latanoprost (Xalatan), travoprost (Travatan Z), tafluprost (Zioptan), bimatoprost (Lumigan) and latanoprostene bunod (Vyzulta). Possible side effects include mild reddening and stinging of the eyes, darkening of the iris, darkening of the pigment of the eyelashes or eyelid skin, and blurred vision. This class of medicine is prescribed for once-a-day use.
  • Beta blockers. These reduce the production of fluid in your eye, helping to lower eye pressure. Examples include timolol (Betimol, Istalol, Timoptic) and betaxolol (Betoptic S). Possible side effects include difficulty breathing, slowed heart rate, lower blood pressure, impotence and fatigue. This class of medicine can be prescribed for once- or twice-daily use depending on your condition.
  • Alpha-adrenergic agonists. These reduce the production of the fluid that flows throughout the inside of your eye. They also increase the outflow of fluid in the eye. Examples include apraclonidine (Iopidine) and brimonidine (Alphagan P, Qoliana). Possible side effects include irregular heart rate; high blood pressure; fatigue; red, itchy or swollen eyes; and dry mouth. This class of medicine is usually prescribed for twice-daily use but sometimes can be prescribed for use three times a day.
  • Carbonic anhydrase inhibitors. These medicines reduce the production of fluid in your eye. Examples include dorzolamide and brinzolamide (Azopt). Possible side effects include a metallic taste, frequent urination, and tingling in the fingers and toes. This class of medicine is usually prescribed for twice-daily use but sometimes can be prescribed for use three times a day.
  • Rho kinase inhibitor. This medicine lowers eye pressure by suppressing the rho kinase enzymes responsible for fluid increase. It is available as netarsudil (Rhopressa) and is prescribed for once-a-day use. Possible side effects include eye redness and eye discomfort.
  • Miotic or cholinergic agents. These increase the outflow of fluid from your eye. An example is pilocarpine (Isopto Carpine). Side effects include headache, eye pain, smaller pupils, possible blurred or dim vision, and nearsightedness. This class of medicine is usually prescribed to be used up to four times a day. Because of potential side effects and the need for frequent daily use, these medicines are not prescribed very often anymore.

Combination drugs:

  • Timolol/Brinzolamide (Azarga-not available in the US)
  • Timolol/Dorzolamide (Cosopt)
  • Timolol/Latanoprost (Xalacom-not available in the US)
  • Timolol/Bimatoprost (Ganfort-not available in the US)
  • Timolol/Brimonidine (Combigan)
  • Brinzolamide/Brimonidine (Simbrinza)

Because some of the eye drop medicine is absorbed into the bloodstream, you may experience some systemic side effects unrelated to your eyes. To minimize this absorption, close your eyes for 1 to 2 minutes after putting the drops in. You also may press lightly at the corner of your eyes near your nose to close the tear duct for 1 to 2 minutes. Wipe off any unused drops from your eyelid.

You may be prescribed multiple eye drops or need to use artificial tears. Make sure you wait at least five minutes in between using different drops.

Never change or stop taking your glaucoma medications without talking to your ophthalmologist. If you are about to run out of your medication, ask your ophthalmologist if you should have your prescription refilled.

Oral medicines

Eye drops alone may not bring eye pressure down to the desired level. So an eye doctor also may prescribe oral medicine. This medicine is usually a carbonic anhydrase inhibitor. Possible side effects include frequent urination, tingling in the fingers and toes, depression, stomach upset, and kidney stones.

Surgery, laser and other therapies

Other treatment options include laser therapy and surgery. The following techniques may help to drain fluid within the eye and lower eye pressure:

  • Laser trabeculoplasty is an option if eye drops can’t be tolerated. Laser trabeculoplasty also may be used if medicine hasn’t slowed the progression of the glaucoma. An eye doctor also may recommend laser surgery before using eye drops. It’s done in the eye doctor’s office. An eye doctor uses a small laser to improve the drainage of the tissue located at the angle where the iris and cornea meet. It may take a few weeks before the full effect of this procedure becomes apparent.
  • Laser iridotomy. Laser iridotomy is for people who have angle-closure glaucoma. The ophthalmologist uses a laser to create a tiny hole in the iris. This hole helps fluid flow to the drainage angle.
  • Glaucoma filtration surgery also called a trabeculectomy. The eye doctor (ophthalmologist) creates an opening in the white of your eye, which also is known as the sclera. This is where your eye surgeon creates a tiny flap in the sclera. They will also create a bubble like a pocket in the conjunctiva called a filtration bleb. It is usually hidden under the upper eyelid and cannot be seen. Aqueous humor will be able to drain out of the eye through the flap and into the bleb, lowering eye pressure. In the bleb, the fluid is absorbed by tissue around your eye.
  • Drainage tubes. In this procedure, the eye surgeon inserts a small drainage tube in your eye to drain excess fluid to lower eye pressure. The glaucoma drainage implant sends the fluid to a collection area called a reservoir. Your eye surgeon creates this reservoir beneath the conjunctiva. The fluid is then absorbed into nearby blood vessels.
  • Minimally invasive glaucoma surgery (MIGS). An eye doctor may suggest a minimally invasive glaucoma surgery (MIGS) procedure to lower eye pressure. This procedure generally require less immediate postoperative care and have less risk than trabeculectomy or using a drainage device. A minimally invasive glaucoma surgery (MIGS) procedure is often combined with cataract surgery. There are a number of minimally invasive glaucoma surgery (MIGS) techniques available.
  • Cataract surgery. For some people with narrow angles, removing the eye’s natural lens can lower eye pressure. With narrow angles, the iris and the cornea are too close together. This can cover (block) the eye’s drainage channel. Removing the eye’s lens with cataract surgery creates more space for fluid to leave the eye. This can lower eye pressure.

After your procedure, you’ll need to see your eye doctor for follow-up exams. You can expect to visit your ophthalmologist about every 3 to 6 months. However, this can vary depending on your treatment needs. And you may eventually need to undergo additional procedures if your eye pressure begins to rise or other changes happen in your eye.

Laser iridotomy

Some reports have demonstrated that laser iridotomy can eliminate iris concavity and reduce iridolenticular contact in eyes with pigment dispersion syndrome 184, 185, 186, 151. However, some eyes may retain a concave iris configuration even after laser treatment 187. In addition, laser iridotomy may not always prevent exercise-induced pigment release and intraocular pressure (IOP) elevation 188, 189. There is limited data on whether laser iridotomy is effective in controlling intraocular pressure (IOP) in patients with pigment dispersion syndrome or pigmentary glaucoma. While one small randomized controlled trial of 21 patients demonstrated a lower rate of IOP elevation over 2 years of follow-up in eyes treated with laser iridotomy as compared to eyes that were not 190, a retrospective study of 60 patients did not suggest any laser iridotomy benefit 191. A 2016 Cochrane review based on 5 randomized controlled trials showed that there was inadequate evidence to use peripheral iridotomy to treat pigmentary glaucoma, and further studies are needed to evaluate the clinical use of this laser treatment in pigment dispersion syndrome and pigmentary glaucoma patients 192.

Follow-up after laser iridotomy is similar to the follow-up for iridotomy performed for angle closure glaucoma.

Rise in IOP after laser iridotomy is greater in pigment dispersion syndrome and pigmentary glaucoma patients than in patients with occludable angles. Ways to mitigate this concern include the use of lower energy levels for surgery, the administration of alpha-adrenergic agonists before and after the laser treatment, and the use of argon laser instead of YAG laser, as it is less disruptive in terms of pigment liberation and inflammation 193.

Pigmentary glaucoma prognosis

Not everyone who has pigment dispersion syndrome will have it lead to pigmentary glaucoma. But the risk of it doing so goes up over time. About 10% of people with pigment dispersion syndrome will develop pigmentary glaucoma within 10 years. That number rises to 15% after 15 years  138. The lifetime risk is between 35% and 50%.

Because pigment dispersion syndrome and pigmentary glaucoma usually start much earlier in life than other forms of glaucoma, it’s important to manage this condition as best you can. Part of that is seeing your eye specialist for regular follow-ups. Those visits can happen every three to six months or yearly, depending on the details of your case. Follow-up visits allow your eye doctor to detect pressure increases and recommend treatment to prevent damage or at least stop it from getting worse.

Ongoing monitoring and managing of your pigment dispersion syndrome or pigmentary glaucoma are key to keeping your eyesight. It’s rare for people who prioritize follow-up visits and manage their condition based on their specialist’s recommendations to have permanent blindness 194.

In a community-based study of 113 patients with pigment dispersion syndrome or pigmentary glaucoma who were followed for a median of 6 years, 1 patient experienced unilateral blindness and another became bilaterally blind 165. In the same study, 10% of patients with pigment dispersion syndrome progressed to pigmentary glaucoma at 5 years, while 15% progressed at 10 years; 23% of patients were noted to have pigmentary glaucoma at diagnosis  165. Visual fields worsened in 44% of patients with pigmentary glaucoma over a mean follow-up period of 6 years. A group of patients followed from a glaucoma clinic showed higher rates of progression (35% over a median follow-up at 15 years), and roughly 40% of patients with pigmentary glaucoma experienced worsening of optic nerve damage 174. In some cases, trabecular meshwork pigmentation and iris transillumination defects have been observed to normalize over time, as has elevated IOP, suggesting return of normal trabecular meshwork function 195, 172, 177, 196. Some older patients with a diagnosis of normal tension glaucoma have been identified with iris transillumination defects and dense trabecular meshwork pigmentation, suggesting they may have had pigmentary glaucoma at some point with subsequent IOP normalization due to cessation of pigment release 197. In such patients, the presence of “pigment reversal sign” helps to distinguish between different types of glaucoma.

Childhood glaucoma

Childhood glaucoma also known as congenital glaucoma, pediatric glaucoma, infantile glaucoma is a rare childhood eye condition where high pressure builds up inside the eye during fetal development that is either present at birth or develops during very early childhood, potentially causing vision loss and even blindness if left untreated 198, 199, 200, 201, 202, 203, 204. Glaucoma can affect one eye or both. Children with childhood glaucoma have ocular hypertension (high pressure inside their eyes). The fluids in their eyes (aqueous humor) fail to drain normally, so they build up, raising the pressure inside. This puts stress on their optic nerves and can eventually cause structural changes to their eyes. Childhood glaucoma is typically diagnosed within the first few months of life commonly in children under 2 years of age caused by a developmental defect in the eye’s drainage system, preventing fluid from flowing out properly and is  often suspected when there is eye enlargement at birth 201. Congenital glaucoma affects about 1 in 10,000 children under 2 years of age in the United States 26, 27.

Eye doctors often classify childhood glaucoma by the age when it first appears 203:

  1. Newborn (neonatal) onset (0-1 month)
  2. Infantile onset (>1-24 months)
  3. Late onset or late-recognized (>24 months)
  4. Spontaneously arrested primary congenital glaucoma (very rare), classic findings of eye stretching including Haab striae with normal intraocular pressure (IOP); must follow as glaucoma suspects.

Primary congenital glaucoma commonly presents between the ages of 3-9 months, but the most severe form is the newborn onset 203. Infantile glaucoma affects individuals between the ages of 1 and 36 months 205, while juvenile glaucoma is used to indicate individuals diagnosed with glaucoma between the ages of 3 and 40 years 206. In most cases, childhood glaucoma is diagnosed by the age of six months, with 80% diagnosed in the first year of life.

The elevated intraocular pressure (IOP) is associated with the classic “triad” of symptoms such as eyes are sensitive to light (photophobia), excessive tearing of the eye (epiphora) and uncontrollable muscle twitching that forces eyes closed (blepharospasm), which occurs due to rapid expansion of the child’s eye causing buphthalmos (“ox-eyed” in Greek), corneal enlargement, horizontal or oblique breaks in Descemet membrane (Haab striae) and subsequent corneal edema and opacification (see Figure 3 and 4 below). If Haab striae and buphthalmos are seen without elevated intraocular pressure (IOP), optic nerve cupping or corneal edema, then the patient has spontaneously arrested primary congenital glaucoma 199. It’s very important to recognize and treat childhood glaucoma as soon as possible to minimize the damage and vision loss it can cause in your child.

Due to the elasticity of the eye in young children, the 2013 International Classification System for Childhood Glaucoma defined childhood glaucoma as irreversible or reversible damage to the whole eye and not just the optic nerve as glaucoma is defined for adults 199. Therefore, additional important clinical signs in primary congenital glaucoma, besides elevated intraocular pressure (IOP) and optic nerve cupping, are corneal enlargement and clouding, Haab striae, and buphthalmos. Not all signs are always present, however, and other parts of the eye also stretch with elevated intraocular pressure (IOP). Diagnosis of childhood glaucoma can be delayed if corneas remain clear, despite being enlarged, and bilateral primary congenital glaucoma can be missed if signs and symptoms are mild in one eye. Irreversible vision loss results if elevated intraocular pressure (IOP) is untreated or uncontrolled in primary congenital glaucoma. Optic nerve damage occurs, and focal corneal edema overlying Haab striae, which can be single or multiple, can lead to permanent corneal scarring and opacification. This corneal scarring can obscure the visual axis or cause astigmatism (a common eye condition where the cornea or sometimes the lens doesn’t have a perfectly round spherical shape leading to blurry or distorted vision at all distances, this irregular shape causes light rays to focus at multiple points on the retina instead of a single point, resulting in a fuzzy or wavy image) with or without refractive amblyopia. Amblyopia may also develop due to optic nerve damage, anisometropia (a condition of asymmetric refraction between the two eyes), strabismus or a combination.

There are many causes of childhood glaucoma. It can be hereditary or it can be associated with other eye disorders.

  • If childhood glaucoma cannot be attributed to any other cause, other noticeable eye defects or systemic problem, it is classified as primary congenital glaucoma. The cause of primary congenital glaucoma is not completely understood, though there is significant research to suggest that the trabecular meshwork is immature and compressed. Studies suggest that the normal posterior migration of embryonic neural crest cells destined to become the trabecular meshwork is abnormally halted 207. The drainage angle where the inside of the sclera (the white of your eye) and the outer edge of your iris meet of children with primary congenital glaucoma is described as immature, thick, and compressed. High intraocular pressures (IOP) are believed to be a consequence of increased resistance to aqueous outflow in this abnormal trabecular meshwork. Researchers have identified several gene mutations that can lead to primary congenital glaucoma. Mutations in the CYP1B1 (cytochrome P450 family 1 subfamily B member 1) gene are the predominant genetic anomalies linked to primary congenital glaucoma 208.
  • If childhood glaucoma is a result of another eye disorder, eye injury, or other disease, it is classified as secondary childhood glaucoma.
    • Associated with eye abnormalities e.g., Axenfeld-Rieger syndrome, aniridia (a rare genetic eye disorder characterized by the complete or partial absence of the iris, the colored part of the eye), iridotrabecular dysgenesis, Peter’s anomaly, sclerocornea (a rare, non-progressive, congenital condition where the cornea, normally transparent, becomes opaque and blends with the sclera, the white part of the eye), microcornea (a congenital condition where the cornea, the transparent front part of the eye, is smaller than normal, with a horizontal diameter of less than 10-11 mm), microphthalmos (a developmental disorder of the eye where one or both eyes are abnormally small and have anatomical malformations), ectopia lentis, persistent fetal vasculature, oculodermal melanocytosis, posterior polymorphous dystrophy,
    • Associated with systemic abnormalities e.g., chromosomal disorders like trisomy 21 (Down syndrome), connective tissue disorders such as Marfan syndrome, Stickler syndrome (a group of genetic disorders that primarily affect connective tissues, particularly in the face, eyes, ears, and joints), phakomatoses (a group of genetic disorders also known as neurocutaneous syndromes or neuro-oculo-cutaneous syndromes characterized by systemic hamartomas, primarily affecting the central nervous system, eyes, and skin) common examples include neurofibromatosis (types 1 and 2), Sturge-Weber syndrome, tuberous sclerosis, Lowe syndrome and von Hippel-Lindau disease
    • Glaucoma secondary to acquired causes e.g., retinopathy of prematurity, eye trauma, intraocular tumors, uveitis, eye inflammation, lens‑induced (with/without pupillary block), steroid-induced, intraocular infections, maternal rubella (congenital rubella syndrome), raised episcleral venous pressure
    • Glaucoma after surgery for congenital cataract.

Childhood glaucoma commonly starts with a defect in the way your child’s eye develops. The most common defect is in the trabecular meshwork, the tissue that the eye fluids (aqueous humor) drain through. When the trabecular meshwork doesn’t develop right, the aqueous humor fluids don’t drain properly. The buildup of fluids (aqueous humor) causes pressure in your child’s eye, which damages their optic nerve. It can also cause their cornea to enlarge, stretch, tear and scar. This process is progressive. How fast it progresses depends on how severe the defect in your child’s eye is, how much fluid (aqueous humor) is building up and how high the pressure is inside the eye (intraocular pressure [IOP]). When glaucoma appears in young infants, it’s because these conditions were already progressing during fetal development. When symptoms appear later, it’s because these conditions were less severe at birth, so they took longer to build up.

To diagnose childhood glaucoma, your child’s eye doctor will ask about your child’s medical history and do a complete eye examination of your child.

Furthermore, your child’s eye doctor may perform diagnostic procedures such as:

  • Visual acuity test – the common eye chart test (with letters and images), which measures vision ability at various distances.
  • Pupil dilation – the pupil is widened with eyedrops to allow a close-up examination of the eye’s retina and optic nerve.
  • Visual field – a test to measure a child’s side (peripheral) vision. Lost peripheral vision may be an indication of glaucoma.
  • Tonometry – a standard test to determine the fluid pressure inside the eye.

Younger children may be examined with hand-held instruments, whereas older children are often examined with standard equipment that is used with adults. An eye examination can be difficult for a child. It is important that parents encourage cooperation. At times, the child may have to be examined under anesthesia, especially young children, in order to examine the eye and the fluid drainage system, and to determine the appropriate treatment.

Specific treatment for glaucoma will be determined by your child’s eye doctor based on:

  • your child’s age, overall health, and medical history
  • extent of the disease
  • your child’s tolerance for specific medications, procedures, or therapies
  • expectations for the course of the disease
  • your opinion or preference

It is important for treatment of childhood glaucoma to start as early as possible. Treatment may include:

  • Medications. Some medications cause the eye to produce less fluid, while others lower pressure by helping fluid drain from the eye.
  • Surgery. The purpose of surgery is to create a new opening for fluid to leave the eye. Surgical procedures are performed by using microsurgery or lasers.

Both medications and surgery have been successfully used to treat childhood glaucoma. However, surgery is the primary treatment modality for primary congenital glaucoma. In managing secondary childhood glaucoma, medications is the first-line treatment 209.

Surgical procedures used to treat glaucoma in children include the following:

  • Trabeculotomy and goniotomy.
    • Trabeculotomy is a surgical procedure, primarily used in the treatment of childhood glaucoma, that creates a new drainage opening in the eye’s trabecular meshwork, improving the outflow of aqueous humor and reducing the intraocular pressure (IOP).
    • Goniotomy is a microinvasive glaucoma surgery (MIGS) technique that improves fluid flow in the eye to lower intraocular pressure (IOP). A goniotomy involves making a small incision within the trabecular meshwork, the eye’s natural drainage system, to create a more efficient pathway for fluid outflow. This procedure can be used to treat conditions like childhood glaucoma.
  • Trabeculectomy. Trabeculectomy is a surgical procedure that involves the removal of part of the trabecular meshwork drainage system, allowing the fluid to drain from the eye. Trabeculectomy works by creating a new drainage pathway for the fluid (aqueous humor) within the eye, allowing it to drain into a space beneath the outer layer of the eye (conjunctiva). This new pathway, called a bleb, helps reduce eye pressure and can slow or prevent further vision loss.
  • Iridotomy. Iridotomy is a surgical procedure to treat or prevent angle-closure glaucoma, a condition where the iris (colored part of the eye) blocks the drainage angle, leading to increased eye pressure. The eye surgeon may use a laser to create this hole. Laser iridotomy involves using a laser to create a small hole in the iris, allowing fluid to flow freely and preventing or relieving pressure build-up.
  • Cyclophotocoagulation. Cyclophotocoagulation is a laser procedure that uses a laser beam to freeze selected areas of the ciliary body – the part of the eye that produces aqueous humor – to reduce the production of fluid and reduce intraocular pressure (IOP). Cyclophotocoagulation is a type of cyclodestruction procedure, meaning it aims to reduce intraocular pressure (IOP) by damaging the ciliary body, a key part of the eye that produces aqueous humor. This type of surgery may be performed with severe cases of childhood glaucoma.

The primary treatment of primary congenital glaucoma is angle surgery, either goniotomy or trabeculotomy, to lower intraocular pressure (IOP) by improving aqueous outflow. If angle surgery is not successful, trabeculectomy enhanced with mitomycin C or glaucoma implant surgery with a Molteno, Baerveldt, or Ahmed implant can be performed. In refractory cases, cycloablation can be performed using an Nd:YAG laser, diode laser, or cryotherapy, with diode laser being the most widely used device. Medications, either topically or orally, is typically used as a temporizing measure prior to surgery and to help decrease corneal clouding to facilitate goniotomy, and to supplement intraocular pressure (IOP) control after surgery.

Figure 10. Congenital glaucoma

Congenital glaucoma

Footnote: Primary congenital glaucoma with cloudy corneas.

[Source 203 ]

Figure 11. Buphthalmos in primary congenital glaucoma

Buphthalmos in primary congenital glaucoma

Footnotes: Buphthalmos is derived from “ox-eyed” in Greek. Buphthalmos describes the visible enlargement of the eyeball at birth or soon after due to increased intraocular pressure (IOP) 210. Buphthalmic eyes typically have corneal diameters exceeding 12 mm in newborns or 13 mm in children older than 1 year 210. The corneal diameters sometimes exceed 16 mm, with the globe appearing noticeably enlarged. Normal corneal diameters are 9.5 to 10.5 mm at birth and 11 to 12 mm by age 1 211. Primary congenital glaucoma (onset at birth) and primary infantile glaucoma (onset after birth to 3 years) are the most frequent causes of buphthalmos 212, 213. Corneal edema, increased corneal diameter, and optic disc cupping are the classical manifestations in patients with buphthalmos 214.

[Source 215 ]

Figure 12. Haab striae in primary congenital glaucoma

Haab striae in primary congenital glaucoma

Footnotes: Haab striae are curvilinear breaks in Descemet’s membrane, resulting acutely from stretching of the cornea in primary congenital glaucoma. Haab striae are typically oriented horizontally or concentric to the limbus in contrast to Descemet’s tears, resulting from birth trauma, that are usually vertical or obliquely oriented.

[Source 216 ]

Childhood Glaucoma types

According to the Childhood Glaucoma Research Network (CGRN) classification, childhood glaucoma is classified into primary glaucoma, secondary glaucoma and glaucoma suspect (Figure 6) 199.

  1. Primary childhood glaucoma encompasses primary congenital glaucoma (PCG) and juvenile open-angle glaucoma (JOAG)
    • Primary congenital glaucoma (PCG) is further classified as:
      • Neonatal onset (0–1 month of age),
      • Infantile onset (1–24 months of age),
      • Late onset or late recognition of disease (>2 years of age),
      • Spontaneously arrested primary congenital glaucoma (PCG). Spontaneously arrested primary congenital glaucoma (PCG) was diagnosed in the presence of buphthalmos and Haab striae, with normal intraocular pressure (IOP), normal-appearing optic discs, and no corneal edema 217.
    • Juvenile open-angle glaucoma (JOAG) is defined as a diagnosis of open-angle glaucoma between age 4 to less than 40 years of age, not exhibiting features of primary congenital glaucoma (PCG) (i.e., buphthalmos, Haab striae). Individuals were further reported to have normal-tension glaucoma (NTG, described as maximum recorded IOP ≤ 21 mmHg) or high-tension glaucoma (HTG, maximum recorded IOP > 21 mmHg) in the affected eye/s, where possible.
  2. Secondary childhood glaucoma is classified based on the underlying pathology. Secondary childhood glaucoma includes glaucoma associated with nonacquired ocular anomalies (e.g., Axenfeld-Rieger spectrum, iris hypoplasia, aniridia), glaucoma associated with nonacquired systemic disease (e.g., phacomatoses, Juvenile Idiopathic Arthritis [JIA]), and glaucoma associated with acquired conditions (e.g., uveitis, trauma, or intraocular surgery). Glaucoma following cataract surgery is classified separately 218.
    • Glaucoma associated with acquired conditions in which glaucoma is secondary to a condition that is not present at birth.
    • Glaucoma associated with nonacquired ocular anomalies in which glaucoma is secondary to a nonacquired condition that is predominantly ocular.
    • Glaucoma associated with nonacquired systemic disease in which glaucoma develops in the presence of a disease that is predominantly systemic, with or without ocular manifestations.
    • Glaucoma following cataract surgery in which cataract surgery precedes glaucoma onset regardless of any coexisting ocular or systemic abnormality.

As per the Childhood Glaucoma Research Network (CGRN) classification, individuals were classified as having glaucoma associated with nonacquired ocular anomalies, even in the presence of systemic disease, if the disorder was predominantly ocular 199. This includes individuals with Peters’ anomaly or Axenfeld-Rieger spectrum (ARS) 199. Peters anomaly is a rare congenital disorder characterized by central corneal opacity with a relatively clear peripheral cornea, often with iris and lens adhesions 219. Peters anomaly can have associated systemic abnormalities like cleft lip, cleft palate, short stature, abnormal ears, and intellectual disability 219. Individuals with only posterior embryotoxon and no systemic features were not considered to have Axenfeld-Rieger spectrum (ARS) as per the 9th Consensus Report of the World Glaucoma Association 199. When an individual had anterior segment dysgenesis (ASD) that did not fit a specific phenotype, experts used the term “unclassified ASD” as recommended by Idrees et al 220. Individuals with primary angle-closure glaucoma were classified as having glaucoma associated with nonacquired ocular anomalies because this entity is caused by anatomic disorders of the iris, lens, and retrolenticular structures 221.

Figure 13. Childhood Glaucoma types

Childhood Glaucoma types

Footnotes: Childhood Glaucoma Research Network and World Glaucoma Association algorithm for the classification of childhood glaucoma.

Abbreviations: AL = axial length; C/D = cup-disc; JOAG = juvenile open-angle glaucoma; ROP = retinopathy of prematurity; VF = visual field

[Source 199 ]

Congenital glaucoma causes

There are many causes of childhood glaucoma. It can be hereditary or it can be associated with other eye disorders.

  • If childhood glaucoma cannot be attributed to any other cause, other noticeable eye defects or systemic problem, it is classified as primary congenital glaucoma. The cause of primary congenital glaucoma is not completely understood, though there is significant research to suggest that the trabecular meshwork is immature and compressed. Studies suggest that the normal posterior migration of embryonic neural crest cells destined to become the trabecular meshwork is abnormally halted 207. The drainage angle where the inside of the sclera (the white of your eye) and the outer edge of your iris meet of children with primary congenital glaucoma is described as immature, thick, and compressed. High intraocular pressures (IOP) are believed to be a consequence of increased resistance to aqueous outflow in this abnormal trabecular meshwork. Researchers have identified several gene mutations that can lead to primary congenital glaucoma. Mutations in the CYP1B1 (cytochrome P450 family 1 subfamily B member 1) gene are the predominant genetic anomalies linked to primary congenital glaucoma 208.
  • If childhood glaucoma is a result of another eye disorder, eye injury, or other disease, it is classified as secondary childhood glaucoma.
    • Associated with eye abnormalities e.g., Axenfeld-Rieger syndrome, aniridia (a rare genetic eye disorder characterized by the complete or partial absence of the iris, the colored part of the eye), iridotrabecular dysgenesis, Peter’s anomaly, sclerocornea (a rare, non-progressive, congenital condition where the cornea, normally transparent, becomes opaque and blends with the sclera, the white part of the eye), microcornea (a congenital condition where the cornea, the transparent front part of the eye, is smaller than normal, with a horizontal diameter of less than 10-11 mm), microphthalmos (a developmental disorder of the eye where one or both eyes are abnormally small and have anatomical malformations), ectopia lentis, persistent fetal vasculature, oculodermal melanocytosis, posterior polymorphous dystrophy,
    • Associated with systemic abnormalities e.g., chromosomal disorders like trisomy 21 (Down syndrome), connective tissue disorders such as Marfan syndrome, Stickler syndrome (a group of genetic disorders that primarily affect connective tissues, particularly in the face, eyes, ears, and joints), phakomatoses (a group of genetic disorders also known as neurocutaneous syndromes or neuro-oculo-cutaneous syndromes characterized by systemic hamartomas, primarily affecting the central nervous system, eyes, and skin) common examples include neurofibromatosis (types 1 and 2), Sturge-Weber syndrome, tuberous sclerosis, Lowe syndrome and von Hippel-Lindau disease
    • Glaucoma secondary to acquired causes e.g., retinopathy of prematurity, eye trauma, intraocular tumors, uveitis, eye inflammation, lens‑induced (with/without pupillary block), steroid-induced, intraocular infections, maternal rubella (congenital rubella syndrome), raised episcleral venous pressure
    • Glaucoma after surgery for congenital cataract.

Childhood glaucoma commonly starts with a defect in the way your child’s eye develops. The most common defect is in the trabecular meshwork, the tissue that the eye fluids (aqueous humor) drain through. When the trabecular meshwork doesn’t develop right, the aqueous humor fluids don’t drain properly. The buildup of fluids (aqueous humor) causes pressure in your child’s eye, which damages their optic nerve. It can also cause their cornea to enlarge, stretch, tear and scar. This process is progressive. How fast it progresses depends on how severe the defect in your child’s eye is, how much fluid (aqueous humor) is building up and how high the pressure is inside the eye (intraocular pressure [IOP]). When glaucoma appears in young infants, it’s because these conditions were already progressing during fetal development. When symptoms appear later, it’s because these conditions were less severe at birth, so they took longer to build up.

Primary congenital glaucoma

Most cases of primary congenital glaucoma are sporadic without a family history of the disease 201, 203, 198, 222. The significant risk factors for primary congenital glaucoma are consanguineous marriage also known as cousin marriage (a marriage between two individuals related by blood, typically first or second cousins, or closer), genetic predisposition, and first-degree relatives (including siblings) with glaucoma. Approximately 90% of cases belong to this category. About 10-40% are familial with an autosomal recessive inheritance pattern with incomplete penetrance ranging from 40% to 100% 199, 201. Autosomal dominant inheritance has also been reported 223.

Mutations in the CYP1B1 (cytochrome P450 family 1 subfamily B member 1) gene are the predominant genetic anomalies linked to primary congenital glaucoma 208. The CYP1B1 (cytochrome P450 family 1 subfamily B member 1) gene is essential for the formation of the trabecular meshwork and the anterior portion of the eye. The CYP1B1 gene codes for an enzyme that metabolizes compounds vital for the developing eye, such as fatty acids and vitamins 224, and is expressed in fetal and adult neuroepithelium and ciliary body 199, 225. Severe trabecular meshwork atrophy is seen in mouse models deficient of CYP1B1 226. In zebrafish, CYP1B1 has been found to indirectly affect neural crest migration to the anterior segment and angle by playing a role in ocular fissure closure 227. While the exact mechanism by which CYP1B1 mutations causes primary congenital glaucoma is unknown, scientists know that levels of a protein product of this gene are inadequate for appropriate embryogenic ocular development, resulting in goniodysgenesis. A twin study demonstrated that CYP1B1 gene activity may be implicated in a common pathway primary congenital glaucoma, juvenile open-angle glaucoma (JOAG), and primary open angle glaucoma (POAG). Recent studies propose that the CYP1B1 mutation may also interfere with the ability of retinal ganglion cells to respond to the stress generated by high intraocular pressure (IOP) and the resultant increase in reactive oxygen species 228, 229. CYP1B1 mutations are associated with 15-20% of primary congenital glaucoma cases in Japan and the United States, 75-100% of cases in Saudi Arabia, and all cases in Slovakia Roma 230, 231.

Additional implicated genes, including LTBP2 (latent transforming growth factor beta binding protein 2), are located next to the GLC3C locus 232, 233. These genetic mutations cause a dysfunctional trabecular meshwork, obstructing proper drainage of aqueous fluid and increasing intraocular pressure (IOP). Several gene loci have been linked to primary congenital glaucoma, which includes GLC3A, GLC3B, GLC3C, GLC3D, and GLC3E. Locus GLC3A has been linked to the CYP1B1 gene 234.

Mutations in CYP1B1 (cytochrome P450 family 1 subfamily B member 1) gene are most commonly responsible for autosomal recessively inherited cases 235. A recent systematic review reported that CYP1B1 was the most common gene mutation reported in the current literature and that the other gene variants related to childhood glaucoma included MYOC (myocilin), LTBP2, FOXC1 (forkhead box C1), PITX2 (paired-like homeodomain transcription factor 2), ANGPT1 (angiopoietin 1) and TEK (or receptor tyrosine kinase) 236.

Currently, the chance of identifying a genetic cause is 40% when genetic testing is done 237.

Studies from Western countries have reported primary congenital glaucoma incidences ranging from 1 per 10,000 to 1 per 30,000 live births 238. The incidence is reportedly as high as 1/2500 in countries like Saudi Arabia. Slovakian Roma have the greatest incidence at 1/1250 239. The higher incidence in particular countries and ethnic groups is related to the higher prevalence of consanguineous marriages, particularly in those with frequent cousin-cousin marriages.

Approximately 65% to 80% of cases of primary congenital glaucoma are bilateral 237. A male-to-female ratio of 3:2 has been reported in studies from the United States and Europe 240 A Japanese study quoted a male-to-female ratio of 6:5 in patients with CYP1BI mutation and 19:2 without the mutation 241. Several studies have reported that glaucoma accounted for 7% to 18% of children registered in blind schools 242, 243. Asia, India, and Saudi Arabia have a mean presentation age of 3 to 4 months compared to 11 months in Western countries 244. primary congenital glaucoma appears earlier in high-incidence ethnicities.

Risk factors for developing primary congenital glaucoma

The only known risk factors are genetic – consanguinity and affected siblings. Parents of primary congenital glaucoma patients should be aware that the chance of a second child with primary congenital glaucoma is a small but real risk that usually is no more than 3%. If two children have the disease, then the risk of subsequent children increases to as high as 25%, with the assumption of autosomal recessive inheritance 240. In 2018, Yu-Wai-Man et al 237 compiled the clinical utility gene card for primary congenital glaucoma which describes situations for which gene testing may be useful. Carriers of the CYP1B1 gene mutation and double null CYP1B1 alleles are, on average, more likely to have higher intraocular pressure (IOP) and require more surgeries 224.

Primary congenital glaucoma pathophysiology

The primary pathophysiologic process in primary congenital glaucoma is the defect in the development of the trabecular meshwork and the anterior chamber angle. This hampers the aqueous outflow through the anterior chamber and increases intraocular pressure (IOP). In 1955 and 1966, Barkan 245 and Worst 246 proposed that the presence of an imperforate membrane at the angle of the anterior chamber impeded the aqueous outflow; this was later disproved. The obstruction site is trabecular, as opposed to pretrabecular. The isolated maldevelopment of the trabecular meshwork, known as isolated trabeculodysgenesis, is the fundamental disease 247.

The formation of an immature angle is believed to stem from the developmental arrest of tissues originating from neural crest cells during the third trimester of gestation. The degree of angle abnormalities is contingent upon the point at which angle development is halted 248. The pathophysiology is believed to result from compacted thick trabecular sheets that merge and inhibit the posterior movement of the iris during the development of the anterior portion. The trabecular sheets position the iris more anteriorly, leading to the iris’ characteristic “high” insertion in children with primary congenital glaucoma.

Currently, the most accepted theory of the pathogenesis of primary congenital glaucoma proposed by Anderson states that excessive or premature accumulation of collagenous beams within the trabecular meshwork prevents normal insertion of the ciliary body and iris 249. This results in an anteriorly inserted iris root and ciliary muscle, which can obstruct the trabecular meshwork, and narrow or completely compress the Schlemm canal elevating intraocular pressure (IOP) 250, 249. Increased intraocular pressure (IOP) leads to the typical symptoms of buphthalmos (enlargement of the eye) and Haab striae (breaks in the Descemet membrane). Histopathological and electron microscopic studies of primary congenital glaucoma have demonstrated obstruction through the outflow pathway 250. Frequently, the ciliary muscle is inserted high on the trabecular meshwork. Moreover, a detailed framework analysis has shown an excessive amount of collagen in the trabecular meshwork. Other studies have demonstrated fibrillary collagen fibers, elastin fibers, and ground substances in the intervening trabecular meshwork and the canal of Schlemm 251. The microscopic observations explain the clinical manifestation of elevated intraocular pressure (IOP) and optic nerve impairment.

More recently, ultrasound biomicroscopy and anterior segment optical coherence tomography have been used to determine angle abnormalities in primary congenital glaucoma patients 252.

Childhood glaucoma prevention

There is no known way to prevent primary congenital glaucoma. Early detection and treatment are essential to maximize visual potential. A family history of glaucoma and a parental consanguineous marriage are essential elements to consider when considering a diagnosis of primary congenital glaucoma 253.

In the future, prenatal genetic screening may emerge as a preventative measure. It can be offered to parents in at-risk populations, such as those with family history or in consanguineous relationships in areas with higher primary congenital glaucoma prevalence (Slovakia, Saudi Arabia, China, etc.). Parents with unborn children who test positively for mutations in CYP1B1 on genetic screening can be alerted about the potential need for urgent surgical management soon after birth 224.

Childhood glaucoma signs and symptoms

Childhood glaucoma symptoms may not be as obvious in children. The following are the most common symptoms of childhood glaucoma. However, each child may experience symptoms differently. Symptoms may include:

  • Excessive tearing (epiphora)
  • Eye(s) that is sensitive to light (photophobia)
  • Closure of one or both eyes in the light
  • Cloudy, enlarged cornea (cloudy cornea)
  • One eye may be larger than the other (bupthalmos)
  • Vision loss

The classic triad of childhood glaucoma symptoms includes:

  1. Epiphora (watery eyes, tearing).
  2. Photophobia (light sensitivity).
  3. Blepharospasm (uncontrollable eyelid twitching).

Other signs of childhood glaucoma may include:

  • Buphthalmos (enlarged eyeballs or ox-eye).
  • Bluish discoloration of the eyeball.
  • Whitening or clouding of the cornea.

You may or may not be able to tell that your child has vision issues, like:

  • Blurry vision (astigmatism).
  • Nearsightedness (myopia).
  • Favoring one eye (anisometropia).

An eye exam might reveal further signs of glaucoma, like:

  • Corneal edema (swelling).
  • Tears in the cornea.
  • Corneal scarring.

Children usually have signs and symptoms in both eyes. But sometimes, they appear only in one.

If the eye pressure increases rapidly, there may be pain and discomfort. Parents may notice that the child becomes irritable, fussy, and develops a poor appetite. Early detection and diagnosis is very important to prevent loss of vision. The symptoms of glaucoma may resemble other eye problems or medical conditions. Always consult your child’s doctor for a diagnosis.

Childhood glaucoma complications

Untreated intraocular pressure (IOP) or delayed treatment in an infant eye may lead to severe complications and significant visual impairment in addition to permanent optic nerve damage and glaucomatous visual field defects. High intraocular pressure (IOP) causes corneal edema and corneal stretching with development of Haab striae. With prolonged corneal edema, both diffuse and focal overlying Haab striae, the cornea can become permanently opacified. Buphthalmos with axial elongation, and Haab striae cause abnormally high refractive errors including myopia and astigmatism, that can impair vision both by blurring vision and causing refractive amblyopia, which can be exacerbated by anisometropia in unilateral cases. In severe buphthalmos, with continued stretching, the lens could dislocate, and risk of retinal complications increases (i.e. lacquer cracks and retinal detachments). Overcoming these complications can be difficult in severe cases. Corneal transplantation for corneal opacification is avoided if possible due to high risk of failure and complications in young children.

Childhood glaucoma diagnosis

The diagnosis of primary congenital glaucoma can often be made clinically via thorough and precise ophthalmologic assessment, even without an accurate measurement of intraocular pressure (IOP). The hallmark of primary congenital glaucoma, however, is an elevated intraocular pressure (IOP) and ocular stretching in the absence of other ocular and systemic conditions that can cause glaucoma, such as Axenfeld-Reiger syndrome, aniridia, or surgical removal of cataract in infancy (i.e. glaucoma following cataract surgery) 254 .

The clinical diagnosis of primary congenital glaucoma can be difficult, especially when a child does not cooperate with intraocular pressure (IOP) measurement. If a reliable intraocular pressure (IOP) measurement is elevated in the setting of other classic signs of ocular stretching, then the diagnosis of glaucoma is made, and if no other ocular or systemic developmental anomalies are seen, then primary congenital glaucoma is the diagnosis. The presence of Haab striae suggests congenital glaucoma, and if seen without ocular developmental anomalies or systemic syndromes, then primary congenital glaucoma is the diagnosis. If intraocular pressure (IOP) is normal with Haab striae, then one may have a case of spontaneously arrested primary congenital glaucoma, which still needs to be followed over time for elevated intraocular pressure (IOP) 203.

Medical History

Primary congenital glaucoma patients often present to the physician’s office due to abnormal appearance of the eyes such as a cloudiness or a blue tint to the eyes, or patient behavior such as eye rubbing or shying away from light. While there may be tearing, there is no ocular discharge and usually no eye redness. The patients are otherwise healthy. A positive family history is helpful but often is not present since most cases are sporadic.

Physical Examination

The clinical examination must include 255:

  • Fixation of light: The patient’s ability to fixate and follow light should be tested with each eye separately. There may be exotropia (where one or both eyes turn outward, away from the nose) due to poor fixation and nystagmus in long-standing cases.
  • Sclera: The sclera(e) may appear bluish in color because of high myopia, scleral thinning, and exposure to underlying uveal tissue 256.
  • Cornea: Corneal examination might reveal signs of corneal enlargement or buphthalmos. Normal corneal size from birth to 6 months should be between 9.5 to 11.5 mm. A size of greater than 12 mm should raise the suspicion of glaucoma. A corneal diameter of more than 13 mm in any child older than 6 months indicates corneal enlargement. The slit-lamp examination may reveal horizontal or oblique tears and breaks in the Descemet membrane called Haab striae (see Figure 4). Another critical finding is corneal edema. This usually starts as epithelial edema and then gradually involves the deeper layers of the cornea, occasionally causing permanent opacities impairing vision profoundly 257.
  • Anterior chamber: The anterior chamber is usually deep.
  • Iris: Iridodonesis, ectropion uvea, hypoplasia, or any atrophic patches may be present 258.
  • Pupil: The pupil may be oval, dilated, and ischemic.
  • Lens: The clinician should evaluate for lenticular opacities or lens subluxation due to excessive stretching of zonules 259.
  • Optic disc: This typically demonstrates reversible cupping in the early stages. Later stages may present with an enlarged cup-to-disc ratio or even atrophy 260, 261.
  • Intraocular pressure (IOP): Intraocular pressure (IOP) is usually elevated at presentation and can be measured using a pneumotonometer in the outpatient setting 254.

Childhood glaucoma signs

The main clinical signs of primary congenital glaucoma include elevated intraocular pressure (IOP) >21 mmHg, corneal edema and/or enlargement of the eye with buphthalmos, and Haab striae. The intraocular pressure (IOP) at presentation is usually between 30-40 mmHg, though it can be outside this range 262. Intraocular pressure (IOP) in the low-20s mmHg is acceptable if the optic nerve is healthy and the patient’s eye growth is within normal limits, but may not be if there are other more severe signs of primary congenital glaucoma.

With intraocular pressure (IOP) in the 30-40s mmHg, the cornea becomes cloudy due to diffuse and/or focal edema. As in adult eyes, the endothelial cell layer cannot pump fluid out of the cornea in an eye with elevated intraocular pressure (IOP). In young children however, there is the additional insult of corneal stretching from the high intraocular pressure (IOP) causing not only enlargement of the cornea, but Descemet breaks, leading to “striae,” which are areas of bare stroma bordered by two separated edges of Descemet membrane that become ridges due to deposition of hyaline 240. These are called Haab striae and are associated with acute overlying focal corneal edema when the intraocular pressure (IOP) is high. They occur in about 25% of primary congenital glaucoma eyes presenting at birth, and more than 60% of primary congenital glaucoma eyes identified at 6 months of age 263. There may be single or multiple, and are oriented horizontally or obliquely. After normalization of intraocular pressure (IOP), corneal edema may clear; however, Haab striae remain and may be associated with corneal scarring. The poorly controlled cases of primary congenital glaucoma may end up with dense stromal opacification even after intraocular pressure (IOP) is controlled.

A newborn’s cornea is typically 9.5-10.5 mm in diameter and increases to 10.0-11.5 mm by age 1 264. Any diameter above 12.0 mm before 1 year of age suggests an abnormality, especially if there is asymmetry between the two eyes. If the diameter is greater than 13 mm at any age, glaucoma suspicion should be high. Along with corneal stretching in the setting of elevated IOP, there is stretching of the scleral wall and all tissues within the eye leading to buphthalmos. Corneal enlargement stops around age 3 years, while sclera can continue to stretch up to age 10 years of age 240.

Other signs related to the eye distension include abnormally deep anterior chamber, myopia (mainly due to elongation and enlargement of the eye), astigmatism (from Haab striae and corneal stretching), anisometropia (almost always present in unilateral primary congenital glaucoma), and optic nerve cupping.

The optic nerve cupping in very young children may be seen solely due to optic canal stretching and posterior bowing of the lamina cribrosa without a decrease in the neuroretinal rim 265, 266. When the IOP is normalized, there can be notable reversal of cupping. While cupping may resolve, retinal nerve fiber layer damage, if present, is permanent. In older children and those with advanced glaucoma, cupping occurs due to neuroretinal rim tissue loss, especially at the vertical disk poles 240.

Any asymmetry between eyes in the aforementioned signs should raise suspicion of glaucoma. Lastly, amblyopia, either deprivation or both, may also be present with the other signs mentioned above.

Diagnostic procedures

The main diagnostic test for childhood glaucoma is the measurement of the intraocular pressure (IOP), which should be done prior to instilling dilating drops. In a cooperative infant or young child, this measurement can be obtained in the clinic setting with a Perkins applanation tonometer, Tono-pen (a portable Mackay-Marg-type tonometer) and/or Icare rebound tonometer. In older patients, standard Goldmann applanation tonometry can be performed. A pneumotonometer may be useful to confirm intraocular pressure (IOP) during examination under anesthesia or in clinic if available, and may be less influenced by corneal abnormalities. A Schiötz indentation tonometry is not recommended in these patients due to under- or overestimation of intraocular pressure (IOP) in childhood glaucoma 267, 268. For the uncooperative child, an examination under anesthesia should be performed.

Of note, the Icare rebound tonometer has decreased the need for examinations under anesthesia as it does not require a topical anesthetic 269. Two models available in the United States (Icare TAO1i and Icare ic100) require the patient to be upright, while the newest model, recently approved in the US (Icare ic200), allows measurement in a supine patient. The IOP measured by Icare in cooperative, awake children with known or suspected glauacoma, has been shown to be within 3 mmHg of IOP obtained by Goldmann applanation tonometry in 63% and is higher than measured by Goldmann applanation tonometry in 75% of children 270. By contrast, Icare tonometry may under-measure the IOP compared to Tono-pen readings in the setting of corneal edema 271.

Because anesthetic agents variably alter the intraocular pressure (IOP), with most lowering intraocular pressure (IOP), measurements should be obtained as soon as possible after induction of anesthesia and before intubation. If the intraocular pressure (IOP) is actually elevated, it often remains greater than 20 mmHg under anesthesia, which suggests glaucoma. The normal intraocular pressure (IOP) is lower in infants and young children than adults. A newborn has an average intraocular pressure (IOP) of 10-12 mm Hg, increasing to 14 mm Hg by age 7 or 8 years of age. An asymmetric measurement or an elevated intraocular pressure (IOP) measurement in the presence of other clinical signs helps make the diagnosis of glaucoma.

Corneal diameter measurement is another key diagnostic procedure for childhood glaucoma. Some providers check horizontal diameters only, while some check horizontal and vertical diameters. If there is pannus or scarring obscuring the limbus, the measurement may not be accurate. In the office a millimeter ruler can be placed above the eyes and if the child is not cooperative, a close-up digital photograph can be taken with the ruler in place, and a measurement can be made from the photo. This is most amenable to horizontal corneal diameter measurement. While under anesthesia, calipers with the tips placed at the limbus 180 degrees apart are used across the widest diameter, and then measured with a graduated ruler to check the measurement. Ideally, the measurement can be estimated to the nearest 0.25 mm 199.

Examination for Haab striae is done with an oblique slit beam with a portable slit lamp if the patient is younger or under anesthesia, or on a regular slit lamp in the clinic if the patient is older. Retroillumination can also be used to identify Haab striae. . In older patients with treated primary congenital glaucoma, corneal endothelial protuberances and hyperproliferation of the Descemet membrane/pre-Descemet’s layer complex have been demonstrated with anterior segment OCT (ASOCT). These may demarcate areas in which the edges of the Descemet membrane have re-approximated during the healing process 257.

If a view through the cornea allows it, gonioscopy is done in clinic if tolerated, ideally a Sussman (or similar) indirect gonioscopy lens as it fits easily between a young child’s small palpebral fissure. Using a gonioscopy lens without a handle may be easier as it allows the examiner to hold open the eyelids while placing the lens. More commonly, for initial diagnosis of primary congenital glaucoma, gonioscopy is performed under under anesthesia with a Koeppe or similar direct gonioscopy lens and portable slit lamp. There are different sized Koeppe lenses to fit different corneal diameters. The Koeppe lens is best handled with a glove to avoid fingerprint smudges. The Koeppe lens cup is filled with balanced salt solution and placed quickly on the eye or placed on the eye and tilted with one edge abutting the sclera while filling the space between the lens and eye with solution. Then a binocular microscope such as the portable slit lamp is angled towards the angle of interest and the lens can be shifted slightly toward the angle to optimize the view.

Gonioscopy in these cases helps guide surgical planning in cases of primary congenital glaucoma, and may also identify other angle abnormalities which might identify other secondary glaucoma types, for example Axenfeld-Rieger anomaly (many irido-corneal attachments with anteriorly placed Schwalbe line). Infants with primary congenital glaucoma usually do not have a visible scleral spur because the peripheral iris inserts into the trabecular meshwork (in contrast to normal infants whose peripheral iris and ciliary body have recessed to the scleral spur or posterior to it). There may also be scalloped edges of the peripheral iris and pale peripheral iris stroma in front of the angle causing a “morning mist” appearance. If there are peripheral anterior synechiae, posterior embryotoxon, or other abnormalities, then the diagnosis is unlikely primary congenital glaucoma. Gonioscopy photographs can be taken by instilling the eye with coupling gel and angling the camera lens (i.e. RetCam) obliquely toward the angle of interest and adjusting the focus until the angle comes into clear view.

Axial length is measured with A-scan ultrasonography, ideally using the immersion and not contact method, either in clinic or under anesthesia. It is best done under anesthesia, during baseline examination to determine if the axial length is greater than normal for the patient’s age, and repeated approximately every 3-4 months to assess if the growth rate is greater than average. Of note, measuring axial length itself is not an indication for examination under anesthesia if a patient is otherwise doing well, and can be performed at intervals when examination under anesthesia is needed for clinical management. Sampaolesi and Kiskis provided linear regressions from data of normal children. Sampaolesi used immersion A-scans and found the normal axial length for a one-month-old lies between 17.25 mm (5th percentile) and 20.25 mm (95th percentile). Sampaolesi also recommended that axial length be measured after dilation with cycloplegic drops 272, 199.

Optic nerve evaluation is performed with either indirect or direct ophthalmoloscopy with attention to the cup-to-disc ratio. In the setting of a small pupil, a magnified view of the nerve can be obtained by using a direct ophthalmoscope through a Koeppe gonioscopy lens on the eye. Fundus photography is also recommended for comparisons between serial examinations. B-scan ultrasonography is recommended if the cornea does not allow fundus examination to rule out posterior disease. Severe optic nerve cupping may sometimes be noted on the posterior B-scan.

Pachymetry is used to measure central corneal thickness. The central cornea may be thicker due to corneal edema, and has also shown to be thinner in primary congenital glaucoma patients without corneal edema, likely due to stretching of their tissues 273. Other small studies have shown either no significant difference in central corneal thickness between normal eyes and eyes treated for primary congenital glaucoma, or the central corneal thickness was thicker in eyes treated for primary congenital glaucoma than in normal eyes 274, 275. Corneal hysteresis and corneal resistance factor have been found to be lower in eyes with primary congenital glaucoma compared to normal eyes 274, 275.

Perimetry can be attempted starting around age 7-8 years of age if the patient does not have nystagmus, cognitive impairment or severe vision loss. Quicker testing algorithms such as SITA-FAST may allow children to perform more reliably 276. Goldman perimetry can be very helpful in young children.

Standard tabletop optical coherence tomography (OCT) can be considered once a child can be examined at the regular slit lamp to evaluate the retinal nerve fiber layer and ganglion cell layer. It may be helpful especially if the child cannot perform perimetry. While devices currently do not carry normative data for children, studies have collected data on normal children 277, 278, 279, 280. Handheld and mounted spectral-domain OCT devices are emerging technologies that can be used during examination under anesthesia 281, 282.

Childhood glaucoma diagnostic criteria

Childhood glaucoma diagnostic criteria per Childhood Glaucoma Research Network definition 199:

Definition of childhood glaucoma required two or more of the main categories (1‑5)

  • Intraocular pressure (IOP) >21 mm Hg (investigator discretion if examination under anaesthesia data alone)
  • Optic disc cupping
    • Progressive increase in cup‑disc ratio
    • Cup‑disc asymmetry of ≥0.2 when disc sizes are similar
    • Focal rim thinning
  • Corneal findings
    • Haab striae
    • Diameter
      • >11 mm in newborn
      • >12 mm in child <1 year of age
      • >13 mm any age
  • Progressive myopia /myopic shift coupled with an increase in ocular dimensions out of keeping with normal growth
  • Reproducible visual field defect that is consistent with glaucomatous optic neuropathy with no other observable reason for the visual field defect

Childhood glaucoma differential diagnosis

The differential diagnoses of childhood glaucoma can be remembered by the mnemonic STUMPED, which includes the following conditions:

  • S: Sclerocornea, congenital hereditary stromal dystrophy 283. This uncommon disorder is characterized by corneal opacification and may be mistaken for primary congenital glaucoma. A flattened cornea with no concomitant elevation in IOP or optic nerve impairment defines Sclerocornea.
  • T: Trauma, tears in Descemet membrane or endothelial (ie, from forceps).
  • U: Ulcer caused by various factors, including viral, fungal, bacterial, neurotrophic, and pythium (a parasitic aquatic oomycete that causes vision-threatening keratitis) 284, 285, 286
  • M: Metabolic disorders, eg, mucolipidoses, mucopolysaccharidosis, tyrisinosis
  • P: Peters anomaly, an uncommon disorder characterized by central corneal opacity and adherence of the iris to the cornea, may manifest with glaucoma; the principal anomaly is the corneal-lenticular contact 287
  • E: Endothelial dystrophy, congenital hereditary endothelial dystrophy, posterior polymorphous dystrophy, Fuchs dystrophy 288
  • D: Dermoid 289

Other significant differentials which should be kept in mind include:

  • Interstitial keratitis
  • High myopia
  • Megalocornea
  • Corneal abrasion
  • Messman dystrophy
  • Reis-Buckler dystrophy
  • Retinoblastoma
  • Retinopathy of prematurity
  • Persistent primary hyperplastic vitreous
  • Traumatic glaucoma 290
  • Congenital rubella syndrome
  • Sturge-Weber syndrome
  • Aniridia
  • Optic disc pit
  • Optic atrophy 291
  • Coloboma

Childhood glaucoma treatment

It is important for treatment of childhood glaucoma to start as early as possible. The management of childhood glaucoma is directed toward lowering and controlling the intraocular pressure (IOP) and treating the secondary complications such as refractive change and amblyopia that develop during the course of the disease.

Childhood glaucoma treatment may include:

  • Medications. Some medications cause the eye to produce less fluid, while others lower pressure by helping fluid drain from the eye.
  • Surgery. The purpose of surgery is to create a new opening for fluid to leave the eye. Surgical procedures are performed by using microsurgery or lasers.

Both medications and surgery have been successfully used to treat childhood glaucoma. However, surgery is the primary treatment modality for primary congenital glaucoma. In managing secondary childhood glaucoma, medications is the first-line treatment 209.

Surgical procedures used to treat glaucoma in children include the following:

  • Trabeculotomy and goniotomy.
    • Trabeculotomy is a surgical procedure, primarily used in the treatment of childhood glaucoma, that creates a new drainage opening in the eye’s trabecular meshwork, improving the outflow of aqueous humor and reducing the intraocular pressure (IOP).
    • Goniotomy is a microinvasive glaucoma surgery (MIGS) technique that improves fluid flow in the eye to lower intraocular pressure (IOP). A goniotomy involves making a small incision within the trabecular meshwork, the eye’s natural drainage system, to create a more efficient pathway for fluid outflow. This procedure can be used to treat conditions like childhood glaucoma.
  • Trabeculectomy. Trabeculectomy is a surgical procedure that involves the removal of part of the trabecular meshwork drainage system, allowing the fluid to drain from the eye. Trabeculectomy works by creating a new drainage pathway for the fluid (aqueous humor) within the eye, allowing it to drain into a space beneath the outer layer of the eye (conjunctiva). This new pathway, called a bleb, helps reduce eye pressure and can slow or prevent further vision loss.
  • Iridotomy. Iridotomy is a surgical procedure to treat or prevent angle-closure glaucoma, a condition where the iris (colored part of the eye) blocks the drainage angle, leading to increased eye pressure. The eye surgeon may use a laser to create this hole. Laser iridotomy involves using a laser to create a small hole in the iris, allowing fluid to flow freely and preventing or relieving pressure build-up.
  • Cyclophotocoagulation. Cyclophotocoagulation is a laser procedure that uses a laser beam to freeze selected areas of the ciliary body – the part of the eye that produces aqueous humor – to reduce the production of fluid and reduce intraocular pressure (IOP). Cyclophotocoagulation is a type of cyclodestruction procedure, meaning it aims to reduce intraocular pressure (IOP) by damaging the ciliary body, a key part of the eye that produces aqueous humor. This type of surgery may be performed with severe cases of childhood glaucoma.

The primary treatment of childhood glaucoma is angle surgery, either goniotomy or trabeculotomy, to lower intraocular pressure (IOP) by improving aqueous outflow. If angle surgery is not successful, trabeculectomy enhanced with mitomycin C or glaucoma implant surgery with a Molteno, Baerveldt, or Ahmed implant can be performed. In refractory cases, cycloablation can be performed using an Nd:YAG laser, diode laser, or cryotherapy, with diode laser being the most widely used device. Medications, either topically or orally, is typically used as a temporizing measure prior to surgery and to help decrease corneal clouding to facilitate goniotomy, and to supplement intraocular pressure (IOP) control after surgery.

Medications

Medications for childhood glaucoma is typically used as an adjunct (add-on) to surgery. Most medications in the United States have not been approved for children, however many studies have been performed that inform doctors on their safety and efficacy in children. Timolol (a non-selective beta blocker) is the first choice in pediatric glaucoma. In cases with insufficient reduction of the intraocular pressure (intraocular pressure (IOP)), the combination of timolol once a day and dorzolamide twice a day brings about a good control of the intraocular pressure (IOP). Both medications are effective and well tolerated. The alpha2-agonists have more and potentially serious adverse effects in children and are contraindicated for children younger than 2 years of age. Latanoprost tends to be less effective in lowering intraocular pressure (IOP) in children than in adults 292.

  • Beta-blockers (beta-adrenergic antagonists): Topical beta-blockers play a large role in childhood glaucoma treatment and include timolol (non-selective beta-1 and beta-2 blocker, concentrations of 0.1% available in some countries, 0.25% and 0.5% solutions, and 0.25% and 0.5% gel-forming solution), and betaxolol (selective beta-1-blocker, concentrations of 0.25% and 0.5% solutions). Given potentially high plasma levels of the medication from topical instillation in small children, the lowest concentration available should be initiated first. The solution drops are approved for BID dosing though may be just as effective dosed once in the morning. The gel-forming solutions are approved for once daily dosing. Beta-blockers typically reduce intraocular pressure (IOP) by 20-30%. Side effects are mainly systemic and include respiratory distress, caused by apnea or bronchospasm (which may present as coughing instead of wheezing), and bradycardia. Beta-blockers should be avoided in patients with bradycardia, second- or third-degree atrioventricular block, and active asthma or “reactive airways.” Betaxolol may be less likely to cause pulmonary distress (e.g. asthma attacks) and cardiac side effects 293.
  • Carbonic anhydrase inhibitors: Oral carbonic anhydrase inhibitors include acetazolamide (Diamox, dose 10-20 mg/kg/day divided into 3 or 4 doses) and methazolamide (Neptazane, dose < 2 mg/kg/day, divided into 2 doses) 294, 199. Acetazolamide can be prepared in a flavored syrup (have the pharmacist crush the tablets and suspend the powder in syrup) with a concentration of 50 mg/ml for ease of use. Children can also take the tablet crushed in applesauce or something similar. It reduces the intraocular pressure (IOP) about 20-35%. Side effects occur in >40% of patients and include lethargy, decreased appetite, weight loss, gastrointestinal discomfort, diarrhea and metabolic acidosis. Topical carbonic anhydrase inhibitors include dorzolamide 2% (Trusopt) and brinzolamide 1% (Azopt) drops twice a day (BID) or three times a day (TID). These medications may produce less reduction in intraocular pressure (IOP) (about 25%) than oral carbonic anhydrase inhibitors, but also appear to have fewer systemic side effects. Rarely, side effects can occur, particularly in premature infants, such as metabolic acidosis 295. Topical carbonic anhydrase inhibitors ideally should be avoided or used as a later option in the setting of compromised corneas, especially of a corneal transplant 199.
  • Combination beta-blocker/carbonic anhydrase inhibitor: Timolol 0.5%-dorzolamide 2% (Cosopt) drop twice a day (BID) has been shown to be effective in reducing intraocular pressure (IOP) in children requiring more than one topical medication. It is approved for twice a day (BID) dosing, but cautious use in young children is warranted due to the higher concentration of timolol.
  • Adrenergic agonists (avoid or use with caution in children younger than age 6 years or weight less than 20 kg): Apraclonidine 0.5% (Iopidine) and brimonidine (Alphagan, Alphagan P, 0.1%, 0.15%, 0.2%) are alpha-2 selective agonists and are dosed twice a day (BID) to three times a day (TID). Their effectiveness has not been studied specifically for childhood glaucoma. The side effects in children limit their use. Due to being highly lipophilic, brimonidine passes through the blood-brain barrier potentially causing severe sleepiness, respiratory depression, apnea and coma, especially in neonates and infants, thus it is strictly contraindicated in patients 2 years old or younger. It may also cause bradycardia, hypotension, hypotonia, and hypothermia. Apraclonidine is more hydrophilic which reduces its blood-brain barrier penetration and thus has fewer central nervous system side effects than brimonidine. It must still be used with caution and is best used for short- or intermediate-term intraocular pressure (IOP) lowering. Tachyphylaxis and ocular allergy limit its effectiveness long-term.
  • Combination beta-blocker/alpha-2 adrenergic agonists: Timolol 0.5%-brimonidine 0.2% (Combigan) must not be prescribed to children if there is a contraindication to the individual components.
  • Prostaglandin analogs: Latanoprost 0.005% (Xalatan), travoprost 0.004% (Travatan), bimatoprost 0.01% (Lumigan), and tafluprost (Zioptan, preservative-free) are dosed nightly. Latanoprost reduces the intraocular pressure (IOP) in primary congenital glaucoma 15-20% 296. While the FDA has not approved prostaglandin analogs in children, Europe has approved latanoprost for children. Side effects mainly include lash growth, conjunctival injection, and less commonly iris pigmentation alteration, allergy, uveitis and periocular hyperpigmentation. Side effects seem more prominent with use of travoprost and bimatoprost and less with latanoprost. Long-term side effects are still unknown in children. Prostaglandin-related periorbitopathy has been described in children 297. This class of medication is relatively contraindicated when active inflammation or uveitis is present.
  • Combination beta-blocker/prostaglandin analog: Available in countries outside the United States.
  • Miotic agents: These do not play much of a role in childhood glaucoma likely due to their immature angle anatomy and high ciliary muscle insertion. They include echothiophate, phospholine iodide (irreversible cholinesterase and pseudocholinesterase inhibitors) and pilocarpine (direct parasympathomimetic). Miotic are useful perioperatively for angle surgery. Pilocarpine (0.5-6%, most common 1-2%) is dosed once to four times a day, usually 2-3 times a day after angle surgery. Side effects include miosis, decreased heart rate, apnea, sweating, and hypersalivation, and theoretically may induce cataract and retinal detachment.
  • Modified prostaglandin analogs and rho-associated protein kinase inhibitors: Latanoprostene bunod (Vyzulta) and netarsudil (Rhopressa) have not been studied in patients younger than 18 years of age.

Doctors can start with a either a carbonic anhydrase inhibitor, beta-blocker or prostaglandin analog, or a combination, and progressively add another medication class, keeping in mind medications are generally a temporizing measure prior to surgery. If prescribed before initial surgery, medications should not be used without fairly frequent follow-up, and ideally surgery performed within 2 weeks of childhood glaucoma diagnosis. Early discussion preparing family and caregivers for surgery is necessary. Medications should be continued until surgery, and may help maximize corneal clearing by reducing the intraocular pressure (IOP). After surgery, medications may still be needed as an adjunct and family and caregivers should be made aware of this. Compliance may be an issue when the medication regimen becomes complex and should be addressed. Childhood glaucoma requires lifelong serial measurements of intraocular pressure (IOP), corneal diameter, axial length, refractive error, and optic nerve cupping. If an adequate assessment is not possible in the outpatient clinic, an examination under anesthesia should be performed.

Surgery

Surgery is the mainstay of treatment for patients with primary congenital glaucoma. The type of surgical procedure depends on the disease severity, cornea clarity, and surgeon’s choice and experience. There are 4 major surgical options for primary congenital glaucoma; however once the diagnosis of primary congenital glaucoma is established, angle surgery is the first procedure of choice to incise/open the trabecular meshwork with the hope of allowing aqueous flow from the anterior chamber directly into Schlemm canal. It is generally agreed that angle surgery is most successful in infantile-onset primary congenital glaucoma, and less so in newborn or late-recognized primary congenital glaucoma. Goniotomy is preferred by some surgeons when the cornea is clear enough to permit visualization of anterior segment structures (although some prefer trabeculotomy regardless of the corneal clarity, see below). An incision is made across the trabecular meshwork under direct gonioscopic visualization using a goniotomy knife (Swan knife, needle-knife, disposable 25-gauge needle on a syringe) and surgical goniotomy lens (i.e. Barkan or other goniotomy lens). Traditionally, it is first performed nasally, however modifications can be made to complete it temporally as well at one surgical session. If a surgeon is comfortable with devices and modified techniques such as using the Kahook dual blade, Trabectome, gonioscopy-assisted transluminal trabeculotomy with suture or lighted microcatheter, or Omni, these devices can be used safely to perform a goniotomy in children, however it is not recommended to use these devices in a pediatric eye prior to extensive experience in an adult eye. There is no data to suggest these modified techniques do better than traditional goniotomy or trabeculotomy. Complications include hyphema, anterior chamber shallowing, peripheral anterior synechiae, and rarely, iridodialysis, cyclodialysis, cataract, scleral perforation, epithelial ingrowth, and retinal detachment 199, 298.

When the cornea is not clear enough to permit visualization of the angle, or if preferred due to technical factors or surgeon experience or preference, trabeculotomy ab externo (“trabeculotomy”) is the procedure of choice. Access to Schlemm canal is obtained externally via a partial scleral flap to allow cannulation of Schlemm canal. The older technique opened ~90 degrees of Schlemm canal with a curved rigid pronged probe called a trabeculotome, which can then be rotated gently into the anterior chamber to incise through the trabecular meshwork. A trabeculotome curved in the opposite direction can then be used to cannulate another 90 degrees of Schlemm canal and complete 180 degrees of trabeculotomy. Alternatively, and preferred by many angle surgeons at this time, a 6-0 polypropylene (Prolene) suture or an illuminated microcatheter can be threaded into the entire Schlemm canal and pulled across the anterior chamber to complete a 360-degree trabeculotomy. Complications include hyphema, unintentional filtering blebs, choroidal detachment, cyclodialysis, iridodialysis, lens injury, and infection.

Traditional goniotomy and trabeculotomy ab externo (incising 2 quadrants) have success rates ranging for goniotomy 30-65% and for trabeculotomy 40-80%, with success reported as low as 10% to as high as 94% 199, 299.

Combined trabeculotomy and trabeculectomy (CTT) can be performed if Schlemm canal could not be cannulated or prior trabeculotomy failed, in which a trabeculectomy is added to the trabeculotomy by removing a block of tissue in the scleral flap bed followed by a surgical iridectomy as done in regular trabeculectomy. Mitomycin C may be used with care. Combined trabeculotomy and trabeculectomy (CTT) can be an initial surgical procedure, especially in Indian and Middle Eastern patients 300, 301.

Filtering surgery is considered when one or more angle surgeries have failed and includes traditional trabeculectomy with or without mitomycin C and glaucoma drainage device implantation. Trabeculectomy may be best done using techniques of the Moorfields Safer Surgery System, including fornix-based conjunctival flaps, small radial cuts, mitomycin C under the sclera flap and subconjunctival tissue with wider spread to enhance posterior aqueous flow and reduce bleb-related complications. Use of an anterior chamber maintainer in all cases and releasable sutures are also recommended 302, 303, 304. EX-PRESS mini glaucoma shunts are not used commonly in primary congenital glaucoma as safety and efficacy have not been established long-term in young children. Severe complications of trabeculectomy include vitreous loss, ectasia, scleral collapse, retinal detachment, and phthisis. The child is also at lifelong risk of complications and infection including bleb leak, wound rupture, blebitis, and endophthalmitis.

Reported success rates for trabeculectomy performed for primary congenital glaucoma range between 50-87% 199. The risk of failure is 5.6 times higher in patients age 1 year or less 305. The higher risk of failure in advanced primary congenital glaucoma young patients is due to buphthalmos, lack of scleral rigidity, and highly active healing and scarring.

When trabeculectomy fails or is not a desirable option, then the other filtering option with glaucoma drainage device (GDD) surgery or cyclodestructive procedures are the next surgical choices. All models of glaucoma drainage devices (Molteno, Baerveldt, and Ahmed valve) can be used in primary congenital glaucoma patients, and glaucoma drainage devices can be implanted safely in neonates with attention to eye and implant parameters. Generally, it is advisable to use a fornix incision with conjunctival and Tenon capsule incision 8 mm posterior to the limbus, and double-layer closure with running 8-0 polyglactin (Vicryl) suture on a vascular needle. The flexible implants (Baerveldt and Ahmed) can be trimmed posteriorly so as to prevent plate-optic nerve touch. The amount to trim can be calculated with the online Freedman-Margeta GDD calculator (https://people.duke.edu/~freed003/GDDCalculator/306. Some surgeons place the first tube inferior nasal to preserve conjunctiva superiorly for possible trabeculectomy when the patient is older, however others prefer superior temporal placement of the first tube, for better efficacy, and are able to perform successful trabeculectomy superior nasal at a future time. Complications from glaucoma drainage devices are many and include those of trabeculectomy plus cornea-tube touch, tube erosion through the conjunctiva or cornea, implant migration, and cataract. Infection rates are low 307. The Ahmed valve may additionally fail due to fibrovascular ingrowth into the valve chamber 308.

Success rates vary widely for primary congenital glaucoma and childhood glaucoma. For the Molteno, the range is 56-95%, with slightly higher success with the double-plate implant compared to the one-plate implant 309, 310. The Baerveldt success rates range from 80-95% at 12 months, decreasing to < 50% by 60 months 311, 312. The Ahmed glaucoma valve has about a 55% success rate at 5 years 313, 314.

Cyclodestructive procedures are useful tools in managing refractory primary congenital glaucoma after all other options have been tried, to reduce aqueous production. Results are unpredictable and complications exist. Laser cyclophotocoagulation (CPC) has largely replaced cyclocryotherapy, and diode laser is preferred to Nd:YAG laser due to decreased adverse events such as sympathetic ophthalmia. Transscleral and endoscopic application of laser are both options, with endoscopic preferred if the eye anatomy allows. Transscleral Micropulse-cyclophotocoagulation may have less severe complications than traditional transscleral cyclophotocoagulation and be as effective in children 315, though further research is needed. The limbal anatomy may be distorted and blind application of transscleral cyclophotocoagulation may be better guided with ultrasound biomicroscopy 316. A general rule of thumb for all cyclodestructive procedures is to maintain 1-2 clock hours of untouched ciliary processes, even after repeated sessions, thus careful documentation of treated areas is recommended. Rare complications include hypotony, retinal detachment, visual loss, and phthisis.

Success for transscleral cyclophotocoagulation ranges from 30-79% with retreatment in ~70% of patients, and has been comparable to implanting a second glaucoma drainage device (GDD) in children 317, 318, 319, 320. Endoscopic cyclophotocoagulation (ECP) has been reported to be 64% successful at 1 year, and 16% by 5 years, with sequential endoscopic cyclophotocoagulation (ECP) bringing the rate up to 81% at 1 year, and 34% at 5 years 321, 322.

Surgical Complications

Surgical complications include:

  • Hyphema
  • Shallow anterior chamber 323
  • Peripheral anterior synechiae
  • Iridodialysis
  • Cyclodialysis (a condition where the longitudinal ciliary muscle fibers separates from the scleral spur, the area where the muscle attaches to the eye’s wall) 324
  • Cataract 325, 326
  • Epithelial ingrowth
  • Choroidal detachment
  • Retinal detachment
  • Phthisis bulbi

Filtering Procedure-Related Complications:

  • Over or under-filtration
  • Blebitis
  • Vitreous loss
  • Scleral collapse
  • Scleral flap leak
  • Tube lens touch
  • Endothelial decompensation from tube cornea touch
  • Tube erosion
  • Implant migration
  • Diplopia from implant-related restrictions
  • Endophthalmitis

Cyclodestructive Procedure-Related Complications:

  • Hypotony
  • Retinal detachment 327
  • Phthisis

Anesthesia-Related Complications:

  • Oculocardiac reflex
  • Anaphylaxis
  • Malignant hyperthermia
  • Cardiovascular collapse
  • Hepatic porphyria
  • Hypoxic brain injury.

Postoperative and Rehabilitation Care

The children undergoing surgery should be started on topical steroids, either prednisolone 1% or dexamethasone 0.1% for 1 week each using an 8/7/6/5/4/3/2/1 tapering dosage. In addition, a topical antibiotic in the form of tobramycin 0.3% or 0.3% moxifloxacin or gatifloxacin 4 times per day for 20 days should be supplemented to prevent secondary infection. These patients will need close follow-ups postsurgery to look for signs of hypotony, inflammation, or infection. Moreover, the IOP needs to be recorded every 3 to 4 months for at least 2 years postsurgery.

Cycloplegic refraction will be needed every 6 months for these patients. Moreover, lifelong regular follow-up every 6 months is needed for intraocular pressure (IOP) monitoring and early detection of any surgery-related complications. Cases of failing angle or filtration surgery should be counseled for the need for glaucoma drainage device (GDD) and the risk of subsequent failure, amblyopia, blindness, and phthisis bulbi (a shrunken, disfigured, and non-functional eye that has undergone significant damage).

Surgical follow up

In the short term, patients require frequent follow up to follow response to treatment and monitor for hypotony, infection, and excessive inflammation. For young patients, or patients with less than 2 years of intraocular pressure (IOP) control, follow-up is recommended at least every 3-4 months. Regular life-long follow-up is needed (at least every 6 months) because even if long-term intraocular pressure (IOP) control from a surgical intervention is achieved, asymptomatic relapse can occur at any time and will need to be managed with medications or further surgery. Additionally, vision-threatening complications may occur at any time, especially after filtering surgeries.

Glaucoma drainage device (GDD) patency can be assessed with B-scan ultrasonography in the clinic 328.

Childhood glaucoma prognosis

The prognosis for children with primary congenital glaucoma is quite variable, with some achieving good vision, while others go blind 203. While primary congenital glaucoma accounts for less than 0.01% of all patients with eye diseases, it has been blamed for 5% of childhood blindness worldwide 237. Vision loss is secondary to corneal scarring or optic nerve damage, and often amblyopia in asymmetric or unilateral cases. Surgical management is the primary treatment modality. If intraocular pressure (IOP) is controlled, vision in the better eye ultimately can be 20/60 or better 329, 330.

A study from the United States showed a lack of progression following adequate treatment in 90.3% at 1 year, 83.1% at 5 years, 70.8% at 10 years, and 58.3% at 34 years 331. Thus highlighting the importance of appropriate management and follow-ups for these patients. Another study showed that angle procedures were 90% successful among patients presenting between 2 months and 1 year of age, compared to 50% among those presenting either in infantile or late-onset or late-recognized cases 332.

Consideration must also be given to the burden on caregivers, whose actions likely will affect the patient’s prognosis. Recent studies have highlighted that one-third of primary congenital glaucoma caregivers could have moderate to severe depression, and quality of life is poorer for the caregiver if the patient is older and has had the disease longer 333, 334.

Open-angle glaucoma

Open-angle glaucoma is the most common type of glaucoma where the drainage angle between the cornea and the iris is wide open and allows the aqueous fluid of your eye to make its way to the trabecular meshwork – the main site for fluid drainage from your eye 335, 336 , 337. In patients with open-angle glaucoma, non-visible abnormalities in the trabecular meshwork reduce the outflow of aqueous humor 335. Experts aren’t sure what causes open-angle glaucoma, but it may be caused by pressure building up in your eye called raised intraocular pressure (IOP) 28, 335. If the aqueous fluid in your eye can’t drain fast enough, it creates pressure that pushes on the optic nerve in the back of your eye. Over time, the raised intraocular pressure (IOP) damages the optic nerve, which affects your vision. This can eventually lead to blindness. In fact, open-angle glaucoma is the most common type of glaucoma in the United States, where 9 in 10 people with glaucoma have the open-angle type and open-angle glaucoma causes almost 2 in 10 cases of blindness in African Americans 25, 338. In the United States, open-angle glaucoma is what most people mean when they talk about glaucoma. People with high blood pressure (hypertension) or diabetes are at higher risk for open-angle glaucoma. Open-angle glaucoma is painless and causes no vision changes at first. Many people with open-angle glaucoma don’t have any symptoms until they start to lose their vision, and people may not notice vision loss right away.

There are 2 types of open-angle glaucoma that are classified depending on the cause:

  • Primary open-angle glaucoma (POAG): If there is no identifiable factor causing the glaucoma (i.e., the cause of the glaucoma is unknown), this is referred to as primary open-angle glaucoma (POAG) or chronic open-angle glaucoma (COAG). Primary open-angle glaucoma (POAG) is the most common form of glaucoma in the U.S. Primary open-angle glaucoma (POAG) can be of 2 types. Primary open-angle glaucoma (POAG) with either elevated intraocular pressure (IOP) or normal intraocular pressure (IOP) also known as normal tension glaucoma (NTG) or low tension glaucoma.
    1. Primary open-angle glaucoma (POAG) with elevated intraocular pressure (IOP) and no other underlying disease. Primary open-angle glaucoma (POAG) accounts for approximately 75% of glaucoma cases in the United States 339, 340. Primary open-angle glaucoma (POAG) affects an estimated 2 to 3% of U.S. adults age 40 and older, and the prevalence is predicted to increase as the population skews older over time 341. The prevalence of primary open-angle glaucoma (POAG) in people of African and Hispanic descent in the United States is at least 3 times higher than in non-Latino White people 342, 343, 35, 4, 344. The reasons behind this association are unclear and complex, and warrant more study because there are also socioeconomic factors that influence access to care and the detection and treatment of glaucoma 345, 35, 346, 341.
    2. Normal tension glaucoma (NTG), also known as normal or low-pressure glaucoma, is a type of primary open-angle glaucoma (POAG) where the optic nerve damage and vision loss characteristic of glaucoma occur despite eye pressure (intraocular pressure [IOP]) remaining within the normal range and an open, normal appearing anterior chamber angle 131, 2, 3, 32, 33. Normal tension glaucoma (NTG) has the same characteristics as primary open-angle glaucoma (POAG) except the intraocular pressure (IOP) is in the normal range, that is, less than 21 mmHg. One theory regarding the mechanism of injury in normal tension glaucoma (NTG) is insufficient blood flow leading to optic nerve damage. While some try to delineate normal tension glaucoma and primary open angle glaucoma (POAG) as two completely unique disease processes, it has also been suggested that primary open-angle glaucoma (POAG) and normal tension glaucoma (NTG) exist on a continuum with intraocular pressure (IOP) playing a larger role in primary open angle glaucoma (POAG), and vascular or mechanical factors as the root cause in normal tension glaucoma 41.
  • Secondary open-angle glaucoma: Secondary glaucoma is when another condition or problem within the eye increases eye pressure such as eye injury, eye surgery or eye procedures, certain medications especially corticosteroids and cycloplegics or other eye diseases (e.g., pigmentary dispersion syndrome, pseudo-exfoliation syndrome, uveitis) causing the glaucoma. This is when another condition or event increases eye pressure, which leads to glaucoma.

Open-angle glaucoma is a diagnosis of exclusion, and other ocular emergencies such as acute closed-angle glaucoma must be ruled out 347. Diagnosis of open-angle glaucoma requires eye assessment by an eye specialist. This will include examination of the optic nerve where it leaves the eye (the optic disc), a check of the intraocular pressure and a field of vision test. Other tests may include measurement of the central corneal thickness and an examination of the drainage angle with a special lens placed on the eye.

Your eye specialist will conduct a thorough eye assessment to diagnose open-angle glaucoma:

  • Intraocular pressure (IOP)
  • Open- normal appearing anterior chamber angle
  • Characteristics signs of optic disc damage
  • Visual function loss on perimetry

Frequently, the diagnosis is not clear-cut and the patient may present with some risk factors and signs, but not others. In such cases, the patient may be labeled as a glaucoma suspect. These patients require repeat assessments at regular intervals with the frequency of visits dependent upon index of suspicion.

Glaucoma eyesight damage is permanent and it cannot be reversed. But medicine and surgery can help to stop further damage. To treat glaucoma, your ophthalmologist may use one or more of the following treatments.

  • Medications. Glaucoma medications lower pressure inside your eye in different ways. Some of them cause your pupil to relax more, improving aqueous humor drainage. Others slow the production of aqueous humor.
  • Glaucoma surgery. This approach usually aims to improve fluid flow and drainage. Examples of glaucoma surgeries that do this include laser trabeculoplasty, laser iridotomy, trabeculectomy (glaucoma filtration surgery), drainage tubes and minimally invasive glaucoma surgery (MIGS).

Figure 14. Open-angle glaucoma

Open-angle glaucoma

[Source 30 ]

Open-angle glaucoma causes

Scientists aren’t sure what causes glaucoma, but the most common types usually happen when the fluid pressure inside your eye (IOP) slowly rises, damaging the optic nerve. As the optic nerve gradually gets worse, blind spots develop in your vision. Primary open-angle glaucoma (POAG) is the most common type of open-angle glaucoma that is characterized by raised intraocular pressure (IOP) caused by increased resistance to drainage in the trabecular meshwork, despite the drainage angle between the cornea and iris remains open 337. Due to this blockage in the trabecular meshwork, the pressure in the eye gradually increases, resulting in optic nerve damage and progressive visual loss. Many different abnormalities have been noted on histopathological examination of the drainage angle in patients with primary open-angle glaucoma (POAG). These include narrowed intertrabecular spaces, thickened basement membranes, fused trabecular beams, reduction in trabecular endothelial cells, reduction in actin filaments, narrowing of collector channels, foreign material accumulation, scleral spur thickening, and closure of Schlemm’s canal 336 .

Secondary open-angle glaucoma can have multiple causes but is far less common than primary open-angle glaucoma (POAG). Secondary glaucoma is when another condition or problem within the eye increases eye pressure such as eye injury, eye surgery or eye procedures, certain medications especially corticosteroids and cycloplegics or other eye diseases (e.g., pigmentary dispersion syndrome, pseudo-exfoliation syndrome, uveitis) causing the glaucoma.

Genetics

  • Myocilin (MYOC) gene mutation on chromosome 1q24.3: The MYOC (myocilin) gene provides instructions for producing a protein called myocilin 348. Myocilin is found in certain structures of the eye, called the trabecular meshwork and the ciliary body, that regulate the pressure within the eye (intraocular pressure) 348. Myocilin is a cytoskeletal protein expressed in the trabecular meshwork and is also known as trabecular meshwork glucocorticoid-inducible response protein 221. Myocilin’s function is not well understood, but it may help to control the intraocular pressure through its action in the muscle tissue of the ciliary body. Researchers believe that myocilin functions together with other proteins in the eye as part of the extracellular matrix, which is an intricate lattice that forms in the space between cells and provides structural support 348. Myocilin may interact with a number of other proteins that also function in the extracellular matrix. The MYOC gene is implicated in cases of hereditary juvenile open-angle glaucoma and adult open-angle glaucoma 221.
  • WD repeat domain 36 (WDR36) gene mutation on chromosome 5q22.1: The WDR36 (WD repeat domain 36) gene is a gene in humans that encodes a protein containing WD repeat domains that is involved in ribosomal ribonucleic acid (RNA) processing, p53 stress-pathway response, cell cycle progression, signal transduction, apoptosis, and gene regulation 349, 350, 351, 352, 353. WDR36 gene been identified as a causative gene for primary open-angle glaucoma (POAG) 354, 353, 355, 356, 357, 358. WDR36 protein is expressed in the lens, iris, sclera, ciliary muscles, ciliary body, trabecular meshwork, retina, and optic nerve in the eye 335. WDR36 protein is also expressed outside the eye in the human heart, placenta, liver, skeletal muscle, kidney, and pancreas. Four mutations in WDR36 at the GLC1G locus (N355S, A449T, R529Q, and D658G) have been identified, with a study implicating the gene in approximately 6% of patients with primary open-angle glaucoma (POAG) 335. However, results from a recent 2017 Chinese study repudiated this claim by showing that the association between WDR36 and primary open-angle glaucoma (POAG) is inconsistent across populations and calls for more data supporting the WDR36 protein’s role in primary open-angle glaucoma (POAG) 359, 360.
  • CAV1/CAV2 genes mutation on chromosome 7q31:
    • The CAV1 (caveolin-1) gene provides instructions for making a protein called caveolin-1. CAV1 (caveolin-1) protein appears to have diverse functions in cells and tissues throughout the body 361. Caveolin-1 (CAV1) is the major component of caveolae, which are small pouches in the membrane that surrounds cells. Caveolae have multiple functions, some of which are not well understood. They are known to be involved in the transport of molecules from the cell membrane to the interior of the cell (endocytosis), processing of molecules on their way into the cell, maintaining the cell structure, and regulating chemical signaling pathways. Studies suggest that caveolae are particularly numerous in adipocytes, which are cells that store fats for energy. Adipocytes make up most of the body’s fatty (adipose) tissue. In these cells, caveolae appear to be essential for the normal transport, processing, and storage of fats. Caveolin-1 (CAV1) is also found in many other parts of cells, where it regulates various chemical signaling pathways. Through these pathways, caveolin-1 is involved in regulating cell growth and division (proliferation), the process by which cells mature to perform specific functions (differentiation), cell survival and the self-destruction of cells (apoptosis), and cell movement. The functions of caveolin-1 likely differ depending on the type of cell and the part of the cell where the protein is found.
    • The CAV2 (caveolin-2) gene encodes a protein that is a major component of caveolae, which are invaginations of the plasma membrane. It plays a role in various cellular processes like signal transduction, lipid metabolism, and cell growth. Caveolin-2 (CAV2) is co-expressed with CAV1 (caveolin-1), forming hetero-oligomers within caveolae. It also interacts with G-protein alpha subunits and can regulate their activity, as well as acting as a scaffolding protein.
    • Caveolin-1 (CAV1) and Caveolin-2 (CAV2) are expressed in most human cell types, including tissues such as the scleral spur cells, trabecular meshwork, and retinal ganglion cells. In vitro studies of CAV1 showed consistent upregulation in the trabecular meshwork after one hour of increased intraocular pressure (IOP) 362, 363.
    • CAV1/CAV2 genes are associated with primary open-angle glaucoma (POAG) susceptibility in populations of European and East Asian ancestry 335.
  • Cyclin dependent kinase inhibitor 2B antisense noncoding RNA (CDKN2B-AS1) gene mutation on chromosome 9p21: CDKN2B-AS1 is a cyclin-dependent kinase inhibitor 2B antisense long non-coding ribonucleic acid (lncRNA) that regulates cyclin-dependent kinase inhibitor 2A and 2B in the cell cycle 364. A United States-based observational case study found that this region modifies optic nerve vulnerability to glaucomatous change. Single nucleotide polymorphism (SNP) in CDKN2B-AS1 gene is thought to be implicated in primary open-angle glaucoma (POAG) by causing retinal ganglion cells (RGCs) to undergo apoptosis during their quiescent post-mitotic state 365, 366.
  • Optineurin (OPTN) gene on chromosome 10p13: Optineurin (OPTN) is the coiled-coil protein product implicated in adult-onset primary open-angle glaucoma (POAG) and normal-tension glaucoma (NTG) 335, 1, 82. Optineurin (OPTN) is involved in various cellular functions, including apoptosis, cellular morphogenesis, inflammation, vasoconstriction, membrane protein trafficking, vesicular trafficking, and transcription activation 367.
  • Lysyl oxidase-like 1 (LOXL1) gene on chromosome 15q24.1: Lysyl oxidase-like 1 (LOXL1) gene codes for an extracellular copper-dependent amine oxidase enzyme that catalyzes the first step in crosslinking collagen and elastin in the extracellular matrix and is implicated in cases of pseudoexfoliation syndrome. Single nucleotide polymorphism (SNP) in the LOXL1 gene is associated with excessive levels of crosslinked amyloid-like fibrillar glycoproteins that deposit in the anterior segment and are more common in Scandinavian populations 368, 369. Single nucleotide polymorphism (SNP) in the LOXL1 gene can present as exfoliation glaucoma as the first signs of a more systemic severe condition that implicates multiple tissues with the expression of copper-dependent amine oxidase enzyme, including the liver, kidney, and gallbladder 370, 371.

Several study results based on whole-exome sequencing using gene-based and single-variant analyses have revealed more than 40 new previously unreported genes associated with glaucoma phenotypes 372. Understanding open-angle glaucoma genetic and molecular mechanisms is crucial to developing new drug targets 373, 374.

Risk factors for developing open-angle glaucoma

Risk factors for developing open-angle glaucoma include 335:

  • Older age (African American, 40+ years; Caucasians, 65+ years) 375
  • Race (African-American, Afro-Caribbean, and West African patients have a 4-fold increased risk of developing open-angle glaucoma) 375
  • Family history of glaucoma (eg, the Rotterdam Eye study found a 9.2 times higher risk of developing open-angle glaucoma if first-degree relatives had glaucoma) 376
  • Elevated intraocular pressure (IOP) 377
  • Myopia or nearsightedness (eg, results from studies have reported an increased risk of glaucoma of up to 20% for each diopter increase in myopia) 378, 342
  • Increased cup-to-disc ratio 379
  • Optic disc hemorrhage also called Drance hemorrhage 380
  • Thin central corneal thickness 381, 342
  • Low ocular perfusion pressure 382
  • Low blood pressure (systolic and diastolic blood pressure) also known as hypotension 383
  • High blood pressure also known as hypertension (systemic arterial hypertension has been associated with but is not a confirmed risk factor for open-angle glaucoma) 384
  • Type 2 diabetes 385
  • Hypothyroidism (underactive thyroid)
  • Obstructive sleep apnea (OSA)
  • Steroid use
  • High pattern standard deviation on visual fields 386
  • Migraine or vasospasm (a condition of sudden constriction of a blood vessel, reducing its diameter and flow rate) 387
  • Low intracranial pressure or low cerebral spinal fluid (CSF) pressure 388
  • Oral contraceptive pill or hormonal birth control pill 389
  • Lifestyle risk factors include smoking, obesity, alcohol, anxiety, stress, and sleep apnea 390.

Intraocular pressure (IOP)

The most important risk factor for developing open-angle glaucoma is an elevated intraocular pressure (IOP) (i.e., ocular hypertension), usually defined as intraocular pressure (IOP) above 21 mm Hg 377. In patients with ocular hypertension (raised IOP but no signs of glaucomatous optic disc or visual field changes), higher intraocular pressure (IOP) is associated with a higher risk of developing open-angle glaucoma 377. Raised intraocular pressure (IOP) in animal models results in glaucomatous optic neuropathy. Population studies have shown increased prevalence of glaucoma with increasing intraocular pressure (IOP). Approximately 10% of patients with an elevated intraocular pressure (IOP) develop glaucoma after five years, and 30% at 20 years 375. Although the risk of glaucoma progression is directly related to the degree of elevation of intraocular pressure (IOP), up to 40% of patients with glaucoma have normal intraocular pressure (IOP) at initial diagnosis, and about 5% of people in the United States have ocular hypertension without glaucoma 342, 35, 391. Intraocular pressure (IOP) is also thought to be a risk factor for normal tension glaucoma, despite intraocular pressure (IOP) never being higher than the normal range.

Age

The prevalence of open-angle glaucoma increases with age, even after compensating for the association between age and intraocular pressure (IOP). Age was also found to be an important risk factor for the conversion of ocular hypertension to open-angle glaucoma in both Ocular Hypertension Treatment Study (OHTS) and the European Glaucoma Prevention Study (EGPS).

Race

Several studies have shown primary open-angle glaucoma (POAG) to be more prevalent in people of African-Caribbean descent compared with Caucasians 336. Not only is open-angle glaucoma more prevalent in black race, its onset is earlier, and disease progression has been shown to be faster and more refractory to treatment. Black patients with ocular hypertension have been found to be more likely to progress to open-angle glaucoma. The prevalence of primary open-angle glaucoma (POAG) in Hispanics is thought to be between that of African-Caribbean and Caucasian populations.

Refractive error

Myopia has been shown to be a risk factor for primary open-angle glaucoma (POAG) in several studies. However, it can be difficult to diagnose true open-angle glaucoma in myopic patients and controversy exists over whether it is real risk factor. Myopic optic discs are notoriously difficult to assess, and myopic patients may have visual field defects unrelated to any glaucomatous process.

Central corneal thickness

A thinner cornea has been shown to be a risk factor for ocular hypertension patients developing primary open-angle glaucoma (POAG). This may be in part due to IOP measurement error (IOP tends to be read lower in patients with thinner corneas), but there are also theories that a thinner cornea may indicate less rigid support structures around the optic nerve head, and a resultant increased propensity to damage 336.

Family history of glaucoma

A first-degree relative (i.e., parents, brother and sister and children) with primary open-angle glaucoma (POAG) is a risk factor for the development of open-angle glaucoma. This has been reported in several studies with the odds ratio varying from 3 to 13 336. The risk is thought to be higher still if the affected relative is a sibling (brother or sister) 336. Several genes associated with open-angle glaucoma have been identified, though these account for less than 5% of all open-angle glaucoma in the general population 336. It is therefore thought that the hereditary aspect of open-angle glaucoma is likely to be polygenic and that gene-environment interactions are important 336.

Other risk factors

A high prevalence of open-angle glaucoma has been found in diabetic patients, and a high prevalence of diabetes has been found in open-angle glaucoma patients. People with diabetes are 2 times more likely to get glaucoma than people without diabetes 375. However, controversy exists as to whether diabetes truly is a risk factor for open-angle glaucoma, as several large population studies have found no association 336 . The role of blood pressure in the development of open-angle glaucoma is complicated and there is no consensus 336. High blood pressure (hypertension) may predispose to glaucomatous damage via increased peripheral vascular resistance in small vessels, while a low blood pressure may reduce the perfusion pressure of the optic disc 336. There is a relative scarcity of data regarding the lifestyle and nutritional epidemiology of open-angle glaucoma 336. There is a suggestion that exercise and a diet high in green collards and with a high omega 6 to omega 3 fatty acid ratio are protective against open-angle glaucoma 336.

Open-angle glaucoma signs and symptoms

Most patients with primary open angle glaucoma (POAG) have no symptoms of the condition. There is no pain and vision seems normal. In its early stages, glaucoma may not cause any symptoms or have no warning signs. Most people with open-angle glaucoma do not notice any change in their vision until the damage is quite severe. This is why glaucoma is called the “silent thief of sight” and up to half of the people in the United States with glaucoma may not know they have it. And symptoms may not appear until glaucoma causes irreversible eyesight damage. As the disease progresses, blind spots develop in your peripheral (side) vision. Side vision also is called peripheral vision. In later stages, difficulty seeing things in your central vision.

It’s important to have regular eye exams that include measurements of your eye pressure. If glaucoma is found early, vision loss can be slowed or prevented. If you have glaucoma, you’ll need treatment or monitoring for the rest of your life.

Some of the more common glaucoma symptoms include:

  • Eye pain or pressure
  • Headaches
  • Red or bloodshot eyes
  • Double vision (diplopia)
  • Blurred vision
  • Gradually developing low vision
  • Gradually developing blind spots (scotomas) or visual field defects like tunnel vision.

Open-angle glaucoma signs

The International Society for Geographical and Epidemiological Ophthalmology has published a consensus definition for primary open-angle glaucoma (POAG). This definition does not include intraocular pressure (IOP) – i.e. primary open-angle glaucoma (POAG) is diagnosed based on signs of glaucomatous optic neuropathy regardless of the level of intraocular pressure (IOP). Patients can be classified as normal tension glaucoma (NTG) or high tension glaucoma (HTG) based on the intraocular pressure (IOP). Sometimes an intraocular pressure (IOP) spike may be missed in a clinical setting. In these cases, if high tension glaucoma (HTG) is suspected, measurement of intraocular pressure (IOP) at hourly intervals throughout the day, beginning in the early morning, may be indicated. This is termed phasing. If a patient has all the features of primary open-angle glaucoma but consistently normal intraocular pressure (IOP) (less than or equal to 21 mmHg), this is considered normal tension glaucoma (NTG).

Open-angle glaucoma complications

Complications of glaucoma include:

  • Blindness: usually painless
  • Painful blind eye or absolute glaucoma: Open-angle glaucoma predisposes to central retinal venous occlusion (CRVO), leading to neovascular glaucoma and painful blind eye 392.

Open-angle glaucoma diagnosis

The only sure way to diagnose glaucoma is with a complete eye exam. An eye specialist can diagnose glaucoma using an eye exam, including several tests that are part of routine eye exams. A comprehensive eye exam can detect glaucoma long before you have eye damage and the symptoms that follow. Many of these tests involve pupil dilation (mydriasis), so your eye doctor can get a better look inside your eye. Your eye care specialist examines your eyes using a special magnifying lens. This provides a clear view of important tissues at the back of your eye to check for glaucoma or other eye problems. For a few hours after the exam your vision may be blurry and sensitive to light, so you will need someone to take you home.

Some of the most helpful glaucoma tests include:

  • Visual acuity testing. A visual acuity test assesses how clearly someone can see at a distance, typically using a Snellen chart or other standardized chart. The test is performed by an optometrist or ophthalmologist and involves reading progressively smaller letters or identifying shapes, with the results expressed as a fraction like 20/20 or 6/6, indicating the distance at which the person can see the letters or shapes
  • Visual field testing also called perimetry. This check of your peripheral (side) vision allows your eye care provider to find out how well you can see objects off to the side of your vision without moving your eyes. This test measures the entire area the forward-looking eye sees to document straight-ahead (central) and side (peripheral) vision. It measures the dimmest light seen at each spot tested. Each time patients perceive a flash of light, they respond by pressing a button.
  • Depth perception testing. A depth perception test assesses your ability to see the world in three dimensions (3D) and judge distances accurately. It checks if your eyes work together and if your brain processes the visual information correctly. These tests use 3D images or patterns like the Randot Stereo test to gauge how well your eyes coordinate to perceive depth. Some tests involve holding a finger in front of your eyes and focusing on a distant object, checking for double vision of the finger.
  • Tonometry. This measures the pressure inside your eye. Increased eye pressure is the most important risk factor for glaucoma. There are several methods of measuring eye pressure. The most common method is known as applanation, in which a tiny instrument contacts the eye’s surface after it is numbed with an eye drop.
    • Air-puff test. You’ll rest your chin on a machine and your eye specialist will blow a puff of air into your eye. This quick and painless test is used as part of a routine glaucoma screening. If the results show that your eye pressure is high, your eye specialist will do other eye-pressure tests to get a more accurate measurement.
    • Applanation tonometry (Goldmann Applanation Tonometry). Your eye specialist will numb your eyes with drops before measuring your eye pressure using one of these methods:
      • You’ll rest your chin on a special magnifying device called a slit lamp. Your eye care specialist will examine your eye through the slit lamp while gently pressing a special tool on your eye to test the pressure.
      • Your eye care specialist will gently press a handheld device against your eye. The device measures your eye pressure.
  • Pachymetry. Pachymetry is a simple, painless test that measures the thickness of the cornea, the clear front part of the eye. The eye doctor uses an ultrasonic wave instrument to help determine the thickness of the cornea and better evaluate eye pressure.
  • Ophthalmoscopy. Your eye care specialist will do a dilated eye exam to look for damage to your optic nerve. This exam is part of a routine glaucoma check-up. You’ll be given eye drops that widen (dilate) your pupils (the openings that let light into your eyes). You’ll look straight ahead while your eye care specialist looks into your eye using a device with a light and magnifying lens.
  • Slit lamp exam. A slit lamp exam is a common eye test that uses a microscope with a focused beam of light to examine the front of your eye and the back of your eye with the aid of special lenses.
  • Gonioscopy. “Gonio” means angle. Gonioscopy is a specialized eye examination that allows an ophthalmologist to visualize the anterior chamber drainage angle, the space between the iris and the cornea where fluid drains out of the eye. Gonioscopy is a crucial part of diagnosing and monitoring glaucoma and other eye conditions. Eye doctors regularly examine the drainage angle to see if there is any visible obstruction to fluid leaving the eye through the trabecular meshwork. A special lens (gonioscopy lens) is needed to examine the trabecular meshwork. The gonioscopy lens is gently placed against the surface of the cornea and allows eye doctors to see the trabecular meshwork in the drainage angle. Gonioscopy determines whether the diagnosis is considered “open” or “closed” angle glaucoma. Gonioscopy is an acquired skill that allows the ophthalmologist to visualize the angle between the cornea and iris and determine if it is open. The angle between the iris and cornea should be 20° to 45° to be considered “open” so that aqueous humor can circumvent the posterior chamber to the anterior chamber 384.

If your eye specialist has a reason to suspect damage to your retina and/or optic nerve, they may also use additional types of eye imaging. These include:

  • Optical coherence tomography (OCT). Optical Coherence Tomography (OCT) measures the reflection of low-coherence infrared light directed toward the back of the eye, and the path of scattered photons helps recreate an image of the retina. Optical coherence tomography (OCT) provides high-resolution cross-sectional imaging of the retina, optic nerve, and anterior segment. Using optical coherence tomography (OCT) a 3D reconstruction of the optic nerve can be created. Optical coherence tomography (OCT) is valuable for monitoring morphological changes in the optic nerve and retinal nerve fiber layer, especially in patients with ocular hypertension and early-to-moderate glaucoma 117. OCT is highly reproducible and is widely used as an adjunct in routine glaucoma patient management. The most recent advances of OCT include OCT-A or OCT-Angiography, whereby the blood flow to vessels surrounding the optic nerve and in the macula can be measured. This is still an active area of research, but scientists do know that some patients’ optic nerves are very vulnerable to changes in optic nerve blood flow, and this new measurement may be useful in evaluating these patients. Peripapillary retinal nerve fiber layer (RNFL) analysis can show thinning in this layer. As the most commonly used scanning protocol for glaucoma diagnosis, optical coherence tomography (OCT)  samples retinal ganglion cells (RGCs) from the entire retina 393. Some of the drawbacks included variability in optic nerve head morphology from patient to patient. The macular thickness is proposed as a means of glaucoma detection, given that 50% of retinal ganglion cells (RGCs) are found in the macula, and retinal ganglion cell (RGC) bodies are thicker than their axons and potentially easier to detect 394. Retinal nerve fiber layer (RNFL), macular thickness measurements, and visual field results are key in managing cases.
  • Heidelberg Retina Tomograph (HRT): Heidelberg Retina Tomograph (HRT) is also a laser that can produce a 3D representation of the optic nerve.
  • Nerve Fiber Analyzer (GDx): Nerve Fiber Analyzer (GDx) uses laser light to measure the thickness of the nerve fiber layer.
  • Fluorescein angiography. Fluorescein angiography is a diagnostic test used to examine the blood vessels in the retina and choroid of the eye. Fluorescein angiography involves injecting a fluorescent dye into the bloodstream and taking photographs of your retina and its blood vessels as the dye circulates, revealing potential blockages, leaks, or other abnormalities in the blood vessels. Fluorescein angiography is often recommended to find and diagnose eye disease including 118:
    • macular edema (swelling in the retina that distorts vision)
    • diabetic retinopathy (damaged or abnormal blood vessels in the eye caused by diabetes)
    • macular degeneration
    • blockage of veins inside the eye, called branch retinal vein occlusion (BRVO) or central retinal vein occlusion (CRVO)
    • macular pucker (a wrinkle in the retina caused by a buildup of fluid behind it)
    • ocular melanoma (a type of cancer affecting the eye)
    • rack changes in eye disease over time
    • target treatment areas
  • Less commonly, ultrasound, computed tomography (CT) or magnetic resonance imaging (MRI).

The following triad is the cornerstone of Open Angle Glaucoma diagnosis 395:

  1. Optic disc or retinal nerve fiber layer changes
  2. Visual field changes
  3. Elevated intraocular pressure (IOP)

Open-angle glaucoma differential diagnoses

Open angle glaucoma differential diagnoses should include 335:

  • Optic nerve head diseases
    • Physiological cup: Deep cup with healthy neuroretinal rim is seen with normal retinal nerve fiber layer thickness and no visual field defect; disc size may be large
    • Optic disc drusen
    • Optic disc coloboma
    • Anomalous optic disc
    • Tilted disc
    • Ischemic optic neuropathy
  • Retinal diseases causing similar visual field defects
    • Branch retinal vein occlusion (BRVO)
    • Branch retinal artery occlusion (BRAO)
    • Retinitis pigmentosa
    • Panretinal photocoagulation
  • Central nervous system diseases
    • Pituitary tumor: Neuroretinal rim is typically pale, pallor more than cupping, and bitemporal hemianopia exists, which respects the vertical line passing through the fixation (contrary to glaucoma, whose visual field respects the horizontal meridian)
    • Intracranial hypotension due to cerebrospinal fluid shunts or other neurologic pathologies
    • Foster Kennedy Syndrome is a rare neurological condition characterized by optic atrophy in one eye and papilledema (swelling of the optic disc) in the other eye, often accompanied by anosmia (loss of smell). Foster Kennedy Syndrome is usually caused by a mass lesion compressing the optic nerve on one side, leading to optic atrophy, and increased intracranial pressure causing swelling of the optic disc on the other side.
    • Cerebrovascular pathologies or traumas
    • Multiple sclerosis

Open-angle glaucoma treatment

The main goal of treating glaucoma is to keep it from getting worse by lowering the pressure inside your eye (IOP) 336, 396. Below a certain upper limit of intraocular pressure (IOP), it is estimated that the visual field, the optic nerve head, and retinal nerve fiber layer (RNFL) parameters will not deteriorate. This helps to maintain a patient’s quality of life.

Some of the most likely treatments for this include:

  • Medications. This mainly involves medications that lower pressure inside your eye. They can prevent glaucoma from developing if you have higher-than-normal intraocular pressure (ocular hypertension) or keep it from worsening enough to cause damage and symptoms.
  • Glaucoma surgeries. These mainly focus on improving the drainage of aqueous humor fluid to lower pressure inside your eye. Surgery options include trabeculectomy, tube shunts, laser therapy and minimally invasive glaucoma surgeries (MIGS).

There is no strong evidence supporting which of medication, laser or surgical therapy should be given initially. Your eye specialist can tell you more about your treatment options and help you choose one that fits your needs best.

The Collaborative Initial Glaucoma Treatment Study (CIGTS) showed no significant difference in outcome between patients randomized to either medical therapy or trabeculectomy 7784. Commonly, patients are started on medical therapy with possible add-on (adjunctive) laser therapy, and only if these measures fail surgery considered.

Unfortunately, some primary open-angle glaucoma (POAG) patients continue to progress despite lowering of intraocular pressure (IOP). This provides some evidence that there are intraocular pressure (IOP)-independent mechanisms at play 336. Therefore, searching for other reversible risk factors for primary open-angle glaucoma is essential 336. For example, this may come from epidemiological study of primary open-angle glaucoma and identifying lifestyle or nutritional risk factors associated with primary open-angle glaucoma 336.

Open-angle glaucoma prognosis

Advanced open-angle glaucoma may cause optic atrophy and no perception of light, though most open-angle glaucoma patients will not lose vision in their lifetime 335. One study in St Lucia found that 35% of untreated eyes progressed to end-stage disease over 10 years 397. There is strong evidence that lowering intraocular pressure (IOP) can reduce the rate of progression significantly. During 6 years of the Early Manifest Glaucoma Trial, 53% progressed, though treatment reduced the rate of progression by half, and for each mmHg of intraocular pressure (IOP) lowering achieved, the rate was reduced by 10%. However, the rate of progression and risk of blindness may be hard to predict in individual patients 336.

Risk factors for progression of open-angle glaucoma include 398:

  • Old age
  • Elevated intraocular pressure (IOP)
  • Increased cup-to-disc ratio or small optic rim area
  • Beta peripapillary atrophy
  • Disc hemorrhage
  • Thin central corneal thickness
  • Reduced corneal hysteresis
  • Low ocular perfusion pressure
  • Poor compliance with therapy
  • Pseudoexfoliation.

Angle closure glaucoma

Angle closure glaucoma also called closed-angle glaucoma, is one of two main types of glaucoma where the iris, the colored part of your eye, bulges forward and blocks the drainage angle, preventing fluid from flowing out of the eye. This fluid buildup increases pressure inside the eye or intraocular pressure (IOP) elevation, which can damage the optic nerve and lead to vision loss. This can occur suddenly (acute) or gradually (chronic) and can cause vision loss if not treated promptly.

There are 2 main types of angle-closure glaucoma:

  • Acute angle-closure glaucoma: Acute (sudden-onset) angle-closure glaucoma occurs suddenly and is a medical emergency, causing severe eye pain, red eye, blurred vision, and nausea. Acute angle-closure glaucoma is a medical emergency, and you should seek treatment straight away so you don’t lose vision. Go to your nearest emergency department or call your local emergency services number immediately and ask for an ambulance. If you’re diagnosed with acute (sudden-onset) angle-closure glaucoma, you’ll need urgent treatment to reduce the pressure in your eye. This generally will require treatment with medicine and a laser or surgical procedures. You may have a procedure called a laser peripheral iridotomy. The doctor creates a small hole in your iris using a laser. The hole allows fluid to flow through the iris. This helps to open the drainage angle of the eye and relieves eye pressure.
  • Chronic angle-closure glaucoma: This develops gradually, often without noticeable symptoms, but can still cause damage to the optic nerve with associated visual field loss over time.

Angle closure glaucoma is a major cause of blindness worldwide, with a particularly high prevalence in some Inuit and Asian populations. Angle closure glaucoma has a familial tendency and is associated with increasing age and farsightedness (having trouble seeing things up close), or hyperopia.

There are primary and secondary forms of angle closure glaucoma. In primary angle closure glaucoma, the mechanism causing disease is primarily pupillary block (iridotrabecular contact is present in three or more quadrants of the drainage angle), being either functional or absolute, in the presence of documented optic nerve damage and visual field loss. Doctors don’t clearly understand why primary angle closure glaucoma happens. While in secondary angle closure glaucoma there are other underlying identifiable pathologic causes, such as neovascularization or uveitis, that either push the iris forward from behind or pull the iris forward to contact trabecular meshwork. If angle closure results in elevated intraocular pressure (IOP) that causes glaucomatous optic disc damage, it is referred to as secondary angle closure glaucoma.

Both primary and secondary angle closure glaucoma can result in dramatic acute angle closure attacks and chronic asymptomatic angle closure disease leading to glaucoma.

Acute angle closure glaucoma is an urgent but uncommon dramatic symptomatic event with blurring of vision, painful red eye, headache, nausea, and vomiting. Diagnosis is made by noting high intraocular pressure (IOP), corneal edema, shallow anterior chamber, and a closed angle on gonioscopy. Medical or surgical therapy is directed at widening the angle and preventing further angle closure. If glaucoma has developed, it is treated with therapies to lower intraocular pressure.

Chronic angle-closure glaucoma is diagnosed by noting peripheral anterior synechiae on gonioscopy, as well as progressive damage to the optic nerve and characteristic visual field loss. Chronic angle-closure glaucoma is treated with therapies to lower intraocular pressure.

A thorough history and eye examination are essential in differentiating between primary and secondary angle closure glaucoma. This distinction is crucial as the treatment of each form can vary greatly. The mainstays of therapy are medications that lower intraocular pressure and laser peripheral iridotomy for any component of pupillary block. Although angle closure glaucoma in all its forms is vision threatening, early diagnosis and appropriate management can stabilize the angle closure glaucoma and minimize vision loss.

Figure 15. Angle closure glaucoma (blocked aqueous outflow increases intraocular pressure causing glaucoma)

angle closure glaucoma

When to see a doctor

Angle-closure glaucoma usually develops quickly, so it needs immediate medical attention to prevent permanent damage and vision loss. The symptoms to watch for are the sudden onset or worsening of:

  • Severe eye pain or pressure
  • Headaches
  • Double vision (diplopia) or blurred vision
  • Nausea and vomiting that happen with eye pain/pressure
  • Rainbow-colored halos around lights
  • Vision loss of any kind
  • Flashing lights in your vision
  • Appearance or increase in visible floaters

Angle closure glaucoma causes

Primary angle closure glaucoma

Primary angle closure glaucoma is caused by relative pupillary block in the majority of cases. In pupillary block, aqueous humor encounters increased resistance as it flows from the posterior to anterior chamber through the iris-lens channel. Some degree of relative pupillary block is present in most phakic eyes. The risk of pupillary block is highest with a mid-dilated pupil where there appears to be maximum contact between the iris and the lens. In eyes with angle closure, other factors exacerbate the block, such as the front lens surface being anterior to the plane of iris insertion into the ciliary body base. The increased pressure gradient across the pupil causes the peripheral iris to bow forward and cover some or all of the filtering portion of the trabecular meshwork, resulting in appositional angle closure. Peripheral anterior synechiae form after prolonged or repeated contacts of the peripheral iris with trabecular meshwork. Another mechanism thought to be important in primary angle closure is iris angle crowding, which is caused by a thickened peripheral iris filling the space between the trabecular meshwork and angle recess under dark conditions 399.

Eyes that experience angle closure glaucoma are anatomically different than normal eyes — they have shorter axial lengths, shallower anterior chambers, thicker and relatively anteriorly positioned lenses, and flatter corneas 400 and they are also physiologically different. Thicker irides may increase the posterior to anterior pressure differential 401. Dynamic factors in angle closure eyes that can contribute to increased pupillary block are the tendency to retain more iris volume after dilation and choroidal expansion causing forward lens movement 402. As imaging modalities, such as ultrasound biomicroscopy and anterior segment optical coherence tomography improve, these dynamic factors will be better studied and understood.

A less common cause of primary angle closure glaucoma is anterior non-pupillary block. This is observed in eyes in which angle closure progresses despite a patent iridotomy, for example, as seen in plateau iris. Plateau iris configuration is characterized by a normal central anterior chamber depth, flat iris profile, and crowding of the angle by the iris base. There is a forward displacement of the iris base by anteriorly located ciliary processes that can lead to subsequent angle closure. Plateau iris syndrome occurs when an eye with plateau iris configuration develops a closed angle 403. Prominent last iris roll is another mechanism of anterior nonpupillary block in which a very thick iris with prominent peripheral circumferential folds becomes more pronounced and contacts trabecular meshwork with dilation 404.

Secondary angle closure glaucoma

Secondary angle closure glaucoma is caused by a myriad of other eye diseases. There are several secondary causes of angle closure that involve relative and absolute pupillary block. In phacomorphic glaucoma, the mass effect of a thickened or intumescent cataract pushes the iris forward and causes pathological angle narrowing. Forward displacement of the lens in ectopia lentis or microspherophakia can also push the iris forward and shallow the angle. Absolute pupillary block occurs when there is no movement of aqueous through the pupil because of 360 degrees posterior synechiae between the iris and a crystalline lens, an intraocular lens, capsular remnants, or the vitreous face. In secondary angle closure glaucoma without pupillary block, angle closure is due to either a.) contraction of an inflammatory, hemorrhagic, or vascular membrane in the angle leading to peripheral anterior synechiae
, or b.) forward displacement of the lens-iris diaphragm, often associated with ciliary body swelling and anterior rotation.

Mechanisms that push the iris forward from behind:

  • Relative pupillary block (primary angle closure)
  • Plateau iris configuration (primary angle closure)
  • Absolute pupillary block – 360o posterior synechiae secluding pupil
  • Aqueous misdirection or malignant glaucoma
  • Ciliary body swelling, inflammation or cysts
  • Choroidal swelling, effusions, or detachments
  • Posterior segment tumors or space-occupying substances (silicone oil or gas bubble)
  • Contracting retrolental tissue as seen in retinopathy of prematurity
  • Anteriorly displaced lens
  • Encircling retinal bands/buckles

Mechanisms that pull the iris forward into contact with the trabecular meshwork:

  • Contraction of inflammatory membrane or fibrovascular tissue
  • Iridocorneal endothelial (ICE) syndrome with migration of corneal endothelium
  • Fibrous ingrowth
  • Epithelial downgrowth
  • Iris incarceration in traumatic or surgical wound

Angle closure glaucoma risk factors

A large number of angle closure glaucoma risk factors have been identified:

  • Hyperopia (farsightedness or hypermetropia) 405
  • Family history of angle closure 406
  • Advancing age 407
  • Female gender 408
  • Asian or Inuit descent 409
  • Shallow anterior chamber depth 410
  • Shorter axial length 411
  • Thicker lens 412

Risk factors for secondary angle closure glaucoma are related to the underlying diseases that cause it.

Angle closure glaucoma signs and symptoms

Patients with anatomic narrow angles without acute angle closure are typically asymptomatic in both the primary and secondary angle closure glaucoma. Similarly, primary and secondary chronic angle closure  glaucoma patients often experience no symptoms unless they develop end-stage glaucoma, in which case they may complain of decreased vision or reduced peripheral vision.

Acute angle closure glaucoma, on the other hand, usually presents with dramatic symptoms from the quick rise in intraocular pressure. Patients complain of blurred vision, rainbows, halos around lights, or even transient loss of vision. They often have intense pain that may be localized to the eye, may follow the trigeminal distribution, or may be described as diffuse discomfort. Nausea and vomiting are common.

Subacute or intermittent angle closure attacks are brief episodes of angle closure that resolve spontaneously. Patients experience the above symptoms of acute angle closure, but on a milder scale. They will typically experience some blurring of the vision or halos with mild to moderate eye pain, brow ache, or headache. These attacks are often resolved by entering a well lit room which may cause miosis or sleep as sleep-induced miosis ameliorates the lesser degree of pupillary block in these patients.

Angle closure glaucoma signs

Signs of a primary anatomic narrow angle on slit lamp exam can be subtle and include a shallow anterior chamber and an anteriorly bowed iris.

Both primary and secondary forms of angle closure can cause acute angle closure attacks. The intraocular pressure usually exceeds 40 mmHg and may rise to as high as 80 mmHg. The conjunctiva is significantly injected. The cornea develops stromal and microcystic edema in response to the acute rise in eye pressure and decompensation of the endothelial pump mechanism. Iris sphincter ischemia leads to a fixed, mid-dilated pupil. Sectoral iris atrophy may occur, releasing pigment into the anterior chamber that dusts the corneal endothelium and anterior lens capsule. The overall anterior chamber is shallow. The center is usually formed, but the mid-peripheral iris bows forward and may touch the peripheral cornea. Often there is anterior chamber inflammation. The fundus is typically difficult to examine because of corneal edema. If visualized, the optic nerve head may be hyperemic and edematous. The angle is often difficult to examine with gonioscopy because of corneal edema, but if visualized, reveals contact of the peripheral iris with cornea.

In the aftermath of an acute attack, pupillary distortion may result if there is permanent sphincter damage and/or iris atrophy. Anterior lens capsule opacities, known as glaukomflecken, may result from damage to the anterior lens epithelium from high intraocular pressure. The angle may have permanent synechiae formation. Intraocular pressure may be low if the ciliary body is so ischemic that aqueous humor production is compromised. As the ciliary body recovers, normal aqueous humor production resumes and the intraocular pressure rises.

Angle closure glaucoma exhibits signs of optic neuropathy in the typical glaucomatous pattern with increased cupping of the optic nerve and retinal nerve fiber layer dropout.

Angle closure glaucoma complications

Complications of acute angle attacks are the result of a rapid, extreme rise in IOP. Possible sequelae include corneal decompensation, cataractous lens changes, iris ischemia resulting in atrophy and distortion, ciliary body shutdown with resultant hypotony, central retinal vein occlusion, optic nerve ischemia, and acute permanent vision loss.

Complications of chronic disease include all the same ones that can be seen in acute disease. The difference in chronic disease is that these conditions develop in a more insidious fashion over a longer period of time. These patients typically have asymptomatic progression of glaucomatous optic neuropathy with corresponding visual field defects developing over time.

Angle closure glaucoma diagnosis

Diagnosis of both primary and secondary angle closure glaucoma is based on history and eye exam. Gonioscopy is the gold standard for evaluating the anterior chamber angle, but imaging modalities assist in quantifying and objectifying angle characteristics. To distinguish between primary and secondary causes, the clinician must actively look for signs and symptoms of possible secondary causes and rule each out before the patient can be diagnosed with primary angle closure glaucoma.

History

  • History of present illness – typical signs and symptoms of acute or subacute angle closure attacks and if patient was upset or in the dark when symptoms started
  • Past ocular history
    • Trauma – can cause zonular weakness or dehiscence allowing lens to displace anteriorly
    • Incisional or laser surgery – can cause anterior chamber inflammation or predispose to epithelial/fibrous ingrowth; may also lead to aqueous misdirection (e.g. after laser peripheral iridotomy) or ciliary body engorgement (e.g. after extensive panretinal photocoagulation).
    • History of prior retinal vein occlusion – can cause angle neovascularization or ciliary body engorgement rotating lens forward
  • Past medical history – history of diabetes or carotid stenosis disease that can cause angle neovascularization
  • Medications – use of systemic medications, such as sulfonamide, topiramate, and phenothizaines, that may cause ciliary body engorgement or suprachoroidal effusion; use of medications to treat allergy, bladder dysfunction, or depression; use of anticholinergics or sympathomimetics that can dilate pupil
  • Family history of acute angle glaucoma

Physical examination

  • Refractive status – hyperopic eyes tend to have shallower anterior chamber angles which places them at risk for angle closure
  • Pupil size and reactivity
  • Slit lamp exam
    • Conjunctiva – injection
    • Cornea – clarity, presence of edema, evidence of surgical or traumatic wounds
    • Anterior chamber – central and peripheral depth, inflammation
    • Iris – areas of atrophy, mass, neovascularization, or posterior synechiae
    • Lens – thickness, phacodonesis, subluxation, glaucomflecken (necrosis of anterior lens capsule; may indicate previous attacks)
  • Intraocular pressure measurement, preferably with applanation prior to gonioscopy
  • Gonioscopy of both eyes with indentation to evaluate for appositional versus synechial angle closure
  • Evaluation of fundus and optic nerve – dilation is often not advisable in primary angle closure attack until an iridotomy has been performed and/or the acute attack has resolved as dilation can exacerbate the condition. In contrast, dilation may be permissible as the appropriate treatment in certain forms of secondary angle closure. The fundus should be examined for underlying causes leading to the angle closure.

Gonioscopy

The key to diagnosis of anatomic narrow angle or angle closure is gonioscopy, which is still the gold standard method of angle evaluation. The ideal way to perform gonioscopy is in a dark room using a small rectangle of light only as bright as necessary to view the angle structures, as light can open an appositionally closed angle in about one-third of cases 413. Dynamic or compression or indentation gonioscopy is essential to differentiate appositional closure from synechial closure. Gentle pressure on the cornea with the goniolens pushes back the iris and reveals whether the angle can be opened any further; if not, synechial closure is present. This maneuver can also help break acute attacks by forcing fluid into the periphery and opening areas of appositional closure.

Occludable angles are typically described as eyes in which the posterior, usually pigmented, trabecular meshwork is seen for less than 90° of the angle circumference or if the angle width is less than 20° 414.

Angle Grading and Classification Systems

A. Scheie system (R)

  • 0 – entire angle visible with wide ciliary body band
  • I – last roll of iris obscuring part of the ciliary body
  • II – nothing posterior to trabecular meshwork visible
  • III – posterior portion of trabecular meshwork not visible
  • IV – no structures posterior to Schwalbe’s line visible

Pigmentation graded 0 (no pigmentation) to 4 (heavily pigmented)

B. Shaffer system (R)

  • 0 – closed or slit
  • 1 – extremely narrow, ≤10 degrees
  • 2 – narrow, 20 degrees
  • 3 – open, 20-35 degrees
  • 4 – wide open, 35-45 degrees

C. Spaeth system (R)

Level of iris insertion:

  • A – anterior to trabecular meshwork
  • B – anterior to posterior limit of trabecular meshwork
  • C – posterior to scleral spur
  • D – into the mid-ciliary body face (anterior ciliary body band visible)
  • E – posterior ciliary body (wide band of ciliary body band visible)

Angle width – estimated in degrees from line tangential to the trabecular meshwork to line tangential to the iris surface one third of the way from the periphery (ranges from 0 – 40 degrees)

Curvature of iris:

  • r – regular configuration, no significant forward or backward arching of iris
  • s – steep or forward bowing (convex) curve
  • q – queer or posterior bowing (concave) curve

Pigmentation: 0 (no pigment) to 4 (heavy pigmentation)

Change in angle configuration after indentation performed described by putting the original insertion in parenthesis, followed by the insertion after indentation. For example, if indentation shows that the insertion is actually a D when it originally appeared to be a C, it is indicated as a (C)D.

Provocative tests

Various provocative tests have been developed in an attempt to separate out patients who may be at higher risk of angle closure. In these tests, different maneuvers are used in an attempt to induce pupillary block, and then the pressure is rechecked and the angle is examined for narrowing. A test is considered positive if the IOP increases by 8 or more mmHg. In the dark room test, patients are placed in a dark room for 1-2 hours to dilate the pupil and increase resistance at the lens-iris channel.

The prone test involves placing the patient in the prone position for 1-2 hours without sleeping to anteriorly displace the lens and increase pupillary block. These tests have not been found to be very predictive of angle closure 415. Combining anterior chamber imaging (e.g. ultrasound biomicroscopy) with provocative testing assists in detecting apposition and allows measurement of various parameters of the angle 416, but their ability to predict future angle closure is not well established.

Pharmacologic provocative tests using mydriatic eye drops to increase pupillary block via pupil dilation have fallen out of favor as they carry a significant risk of angle closure in and of themselves.

Imaging modalities

To supplement information obtained through gonioscopy, there are several anterior segment imaging devices available that provide detailed images of structures and quantitative measurements. They are useful in primary angle closure but can also help detect secondary cases of angle closure, such as ciliary body masses or anterior rotation. At this time, there are no widely agreed upon quantitative measurement cutoffs obtained from these devices that distinguish a narrow angle from an open one.

Ultrasound biomicroscopy

This high-frequency B scan ultrasound provides high-resolution cross-sectional images of the anterior segment of the eye to the anterior vitreous. Because it uses sound, it can pass through opaque structures to visualize structures hidden from direct clinical examination, such as the ciliary body 417. It is particularly helpful for evaluating plateau iris and other ciliary body pathology. The disadvantages of ultrasound biomicroscopy include: requirement of a water bath immersion, specialized equipment, and a skilled technician to operate; it is also relatively costly and time consuming.

Anterior segment optical coherence tomography

This modality uses a diode light source instead of sound to produce highly detailed images of the cornea, angle region, and anterior ciliary body similar to those seen with ultrasound biomicroscopy. Compared to ultrasound biomicroscopy, anterior segment optical coherence tomography (OCT) is unable to image structures posterior to the iris plane well because of posterior pigmented iris shadowing and scleral light scattering 417. The advantages of anterior segment optical coherence tomography are that it is a noncontact exam: the patient can be imaged in an upright position avoiding positional lens changes, and all four quadrants can be scanned at once.

Scheimpflug photography

Digital images of the anterior chamber angle can be obtained using a Scheimpflug camera. Rotating versions of the camera provide three-dimensional photos that can be analyzed by computer software to measure specific parameters of the angle. The camera has an easy-to-use slit lamp type configuration but is expensive and requires special equipment. It cannot image the ciliary processes or body behind the iris.

Optic nerve assessment and imaging, retinal nerve fiber layer analysis, and visual field testing should be preformed to assess for signs of glaucomatous optic neuropathy in any patient with angle narrowing or angle closure glaucoma.

Summary of clinical findings in various types of secondary angle closure glaucoma

  • Neovascularization of the angle – blood vessels from iris that cross scleral spur to arborize along trabecular meshwork, peripheral anterior synechiae
  • Anterior chamber inflammation – keratic precipitates, posterior synechiae, iris bombé, inferior PAS (as opposed to primary angle closure where PAS tend to develop superiorly)
  • Iridocorneal endothelial (ICE) syndrome – beaten-bronze corneal endothelium, corneal edema, high PAS that can extend anterior to the Schwalbe line, iris atrophy, corectopia
  • Drug induced (e.g. systemic topiramate) – acute bilateral disease, acute myopic shift, uniformly shallow chamber with anterior iris and lens displacement, ciliochoroidal effusion or detachment
  • Aqueous misdirection (malignant glaucoma) – flattening of central and periperhal anterior chamber, anterior displacement of lens (cystralline or intraocular lens) or vitreous face, clear “aqueous” zones in vitreous
  • Lens-induced angle closure – thick cataract, unstable or subluxed lens
  • Nanophthalmos – small but normal eye with short axial length, microcornea, large lens, and thickened sclera; choroidal effusion
  • Retinopathy of prematurity or persistent hyperplastic primary vitreous – contracting retrolental tissue
  • Iris or ciliary body mass lesions or cysts – irregular contour of iris or neovascularization, hyphema or vitreous hemorrhage, episcleral sentinel vessel, ciliary body mass through dilated pupil or on ultrasound biomicroscopy
  • Posterior segment mass or large serous retinal detachment – signs of each on funduscopic exam
  • Epithelial and/or fibrous downgrowth – wound dehiscence or gape, epithelial cysts in anterior chamber adjacent to wound, gray sheetlike membrane covering anterior segment structures that whitens with argon laser
  • Pseudophakic or aphakic pupillary block – pupilary obstruction or synechiae to anterior hyaloid surface, the intraocular lens, or posterior capsule
  • Ciliary body engorgement associated with retinal vascular occlusion or panretinal photocoagulation – anterior rotation of iris and lens; retinal signs of each underlying etiology, e.g. retinal hemorrhages for vein occlusion and extensive retinal laser scars from photocoagulation.
  • Retinal surgery
    • Encircling scleral buckle – relatively deep central anterior chamber with peripheral iris flattening, choroidal effusion
    • Pars plana vitrectomy – pupillary block from expansile gases pushing iris and lens forward, silicone oil in the anterior chamber, non-patent iridotomy

Angle closure glaucoma treatment

The overall goals for management are to reverse or prevent the angle closure process, control intraocular pressure elevation, and prevent damage to the optic nerve. Some primary and secondary forms of angle closure may be treated similarly, while others require very different treatment approaches based on their underlying pathophysiology. Intraocular pressure (IOP) is lowered with glaucoma medications. Iridotomy is an essential part of treatment in primary angle closure glaucoma, but may not be indicated in some forms of secondary angle closure glaucoma. Trabeculectomy and tube shunts may also not be indicated for certain secondary forms of angle closure glaucoma.

Medical therapy

The role of medical therapy in acute angle closure attacks is to lower IOP, reduce pain, and clear corneal edema in preparation for iridotomy. The medications below can be used, provided the patient has no condition contraindicating them:

Topical

  • Beta blockers
  • Selective alpha agonists
  • Carbonic anhydrase inhibitors
  • Miotics (e.g., pilocarpine 2%) may help break an early angle-closure attack, but may be ineffective if the iris is already ischemic. High-concentration miotics (e.g., pilocarpine 4%) should be avoided because of the potential for forward displacement iris-lens diaphragm.
  • Prostaglandin analogues – unreliable effect in acute attack because of slow onset of action
  • Hyperosmolar agent (e.g. 5% sodium chloride) – assists in clearing corneal edema
  • Prednisolone 1% – decreases inflammation

Systemic

  • Carbonic anhydrase inhibitors – oral acetazolamide’s maximum intraocular pressure reduction is reached in 2-4 hours and lasts for 6-8 hours. Intravenous acetazolamide drops the intraocular pressure (IOP) within 2 minutes with a peak effect noted by 10-15 minutes. In acute situations, a single dose of 500 mg acetazolamide should be given orally if the patient is not vomiting. Regular acetazolamide is preferred over the sustained-release sequel form because of quicker onset of action. If the patient is vomiting, acetazolamide can be given intravenously.
  • Osmotic agents
    1. Mannitol can decrease the intraocular pressure (IOP) 30 mm Hg or more within 30 minutes of administration. The recommended intravenous dose is 0.5-1.5 g/kg body weight as a 15% or 20% solution, delivered at 3 to 5 mL/minute. Frail patients with cardiac or conditions may develop circulatory overload, pulmonary edema, congestive heart failure, and electrolyte imbalance. A rapid reduction in cerebral volume may result in subdural hematomas from vein rupture between the sagittal sinus and cortical surface. Therefore, patients receiving IV mannitol should be monitored in a hospital setting.
    2. Oral osmotic agents:
      • Glycerin: 1 to 1.5 g/kg body weight of a 50% solution. Onset of pressure reduction is typically 10 to 30 minutes. Avoid in diabetics because the increased caloric load can cause ketoacidosis.
      • Isosorbide is commercially available as a 45% (45 g/100 mL) solution (Ismotic; Alcon Surgical). The recommended dose is 1 to 1.5 g/kg body weight. Its effect is similar to glycerin’s but is safe for use in diabetics because it is not metabolized.
      • Although less common, oral agents can also cause subdural hematomas. Headache and gastrointestinal upset are common adverse reactions.

Paracentesis

Paracentesis can be perfomed in an acute setting. Technically, it can be difficult to perform on a phakic eye in pain with a shallow chamber, and there is a risk of permanent damage to the cornea, lens, and iris. Devastating complications such as endophthalmitis and choroidal hemorrhage from a rapid pressure drop may occur. Also the effects are typically short-term, because, as the ciliary body begins to form aqueous again, the intraocular pressure (IOP) will inevitably rise. This procedure can be used in cases of extreme intraocular pressure (IOP) elevation to “buy time” until medications take effect or iridotomy can be performed.

Laser Iridotomy

Laser Iridotomy should be performed as soon as possible in the affected eye and in the contralteral eye to avoid an attack of acute angle closure glaucoma in the future.

Chronic angle closure glaucoma treatment

Very few studies exist to address medical therapy in chronic angle closure glaucoma after laser iridotomy. In cases where elevated intraocular pressure becomes an issue, aqueous suppressants are helpful in reducing intraocular pressure (IOP) 418. Prostaglandin analogues have been shown to be effective in lowering intraocular pressure, even in angles that are partially closed 419. Evidence is not conclusive, however, regarding their effectiveness in cases of 360° degrees of synechial closure 420. The role of peripheral iridotomy and other surgical interventions are described below.

Peripheral iridotomy

See prevention section for information regarding prophylactic laser peripheral iridotomy for narrow angles.

Acute Angle Closure and Fellow Eyes

In angle closure secondary to pupillary block, an iridotomy is the definitive treatment. Laser peripheral iridotomy is considered an effective and safe treatment. It often breaks an attack of acute angle closure and can prevent future attacks. An incisional iridectomy may be necessary in cases of cloudy corneas, flat anterior chamber, poor patient cooperation at the laser, or inability to substantially lower the IOP with medications after a failed laser peripheral iridotomy attempt.

The fellow eyes of patients that have undergone primary acute angle-closure are generally at significant risk for an acute attack and should receive an iridotomy 421. An untreated fellow eye has a 40% to 50% chance of developing an acute primary angle-closure attack over the next 5 to 10 years 421. Chronic miotic therapy is not an acceptable alternative, as 50% of contralateral eyes of individuals suffering acute primary angle-closure developed acute attaks when treated with pilocarpine alone. This is in contrast to the 1.8% of patients treated with prophylactic incisional iridectomy who developed an attack during this same time period 422.

Chronic Angle Closure and Angle Closure Glaucoma

Laser peripheral iridotomy relieves the pupillary block component in chronic disease and may halt the progression of synechial closure and progressive IOP elevation 423. Its ability to control IOP, however, may not be long-lasting, especially in eyes where glaucomatous optic neuropathy has already developed. Additional medications or surgical treatment is often necessary 424. In cases where laser peripheral iridotomy does successfully lower IOP, eyes still need to be monitored routinely as IOP can increase months or years after the procedure 425.

Persistent or progressive rise in IOP after laser peripheral iridotomy

  • Damage to trabecular meshwork and/or formation of PAS has occurred when iridocorneal apposition present
  • Pupillary block may recur if iridotomy becomes occluded
  • Factors other than pupillary block causing angle closure may have gone unrecognized until after the LPI (e.g. plateau iris syndrome)
  • Angle closure may have been superimposed on pre-exisitng open angle glaucoma or another eye condition causing IOP elevation, such as pseudoexfolation
  • There may be co-existing chronic open angle glaucoma

Complications of laser peripheral iridotomy

The most common complications of laser peripheral iridotomy are transient bleeding at site of treatment, hyphema, postoperative pressure spike, and anterior chamber inflammation. Occasionally, patients may complain of a seeing a double image if the lid does not cover the iridotomy site. More severe but rare complications include aqueous misdirection and injury to the cornea, lens, or retina.

Iridoplasty

In laser iridoplasty, contraction burns of long duration, low power, and large spot size are placed on the peripheral iris to contract the iris stroma and physically pull the iris from the drainage angle in an attempt to open the angle. In acute angle closure, iridoplasty has been found effective and safe in short-term lowering of IOP 426. It can be used in cases that are medically unresponsive, in which systemic carbonic anhydrase inhibitors must be avoided, when immediate iridotomy is not possible, or rarely when the attack continues despite a successful laser peripheral iridotomy. It is the procedure of choice for plateau iris syndrome when the angle fails to open and IOP remains elevated despite a patent peripheral iridotomy. In chronic cases of angle closure, iridoplasty may slightly decrease the formation of peripheral anterior synechiae 427.

It is important to note that iridoplasty does not eliminate pupillary block, so iridotomy remains necessary if pupillary block is the mechanism of angle closure 428. Potential complications include IOP spike, iris atrophy from destruction of iris vessels, corneal burns, marked anterior chamber inflammation, and corneal endothelial damage 420.

Cataract extraction

In primary angle closure glaucoma, since the lens is a key player in development of relative pupillary block, it makes sense that cataract extraction can lower IOP in both acute and chronic angle closure. Removal of the lens from an eye with a crowded anterior chamber opens the angle and may prevent or reduce peripheral anterior synechiae formation. In one study, early phacoemulsification was found to be better than laser peripheral iridotomy at preventing IOP rise after an acute angle closure event was controlled medically 429. Cataract extraction in the setting of an acute angle-closure attack, however, is technically difficult. The eye is inflamed with significant corneal edema, a shallow anterior chamber, an atrophic and atonic iris that is difficult to dilate, and possible zonular weakness. It may be more prudent to control the acute attack with medications and laser peripheral iridotomy first and then wait to perform surgery when the eye has recovered and is less inflamed.

In the chronic phase when patent laser iridotomy and medical treatment have failed to adequately control IOP, lens extraction many months after the initial attack has been found to reduce IOP and reduce the need for IOP medications 430. Cataract extraction can be combined with goniosynechialysis to further improve IOP control in the short term 431. Methods of goniosynechialysis include breaking synechiae with a heavy viscoelastic (viscogonioplasty), forceps, or a cyclodialysis spatula.

In lens-induced angle closure glaucoma, which includes phacomorphic glaucoma and angle closure due to forward subluxation of the lens, the definitive treatment is lensectomy. Acute attacks can be first controlled with medical therapy and/or laser peripheral iridotomy with lens extraction performed when the eye is quiet.

Filtration surgery

Filtration surgery has been performed for both acute and chronic angle closure glaucoma 432. The indications to perform filtration surgery in primary angle closure glaucoma are similar to those for surgery in POAG. In chronic cases, surgery is considered if the optic neuropathy is progressing and IOP is at a level believed to be contributing to the progression. Reasons for performing filtration surgery in the setting of an acute closure attack include medical unresponsiveness, lack of laser availability, or signs of glaucomatous optic neuropathy already present 433. The same techniques of filtration surgery are used as in primary open angle glaucoma, although some surgeons advocate tighter suturing of the trabeculectomy flap to avoid low IOP in the immediate postoperative period. A low IOP may contribute to further anterior chamber shallowing, which may lead to a higher rate of malignant glaucoma postoperatively 434.

Whether to perform filtration surgery versus a tube shunt procedure for secondary angle closure glaucoma depends on the underlying etiology. In most cases, dealing with the underlying pathology (e.g. stopping topiramate in drug-induced glaucoma, removing the lens in lens-induced glaucoma) will slow or stop the progression of disease. If medical therapy is not sufficient to control IOP, even when the primary pathology has been addressed, glaucoma surgery may be necessary and filtration surgery may be appropriate. Conditions such as neovascular glaucoma and ICE, however, tend to do better with glaucoma drainage implants. (see below). It has been proposed that combined phacotrabeculectomy may be more effective at controlling IOP than cataract extraction alone. Recent study results have been mixed, with some showing phacoemulsification as being superior for deepening the chamber 435, the two procedures being equal in terms of IOP control 436 and combined procedures being superior for IOP control 437. Of note, one randomized control trial found that patients who undergo combined procedures have more postoperative complications and progression of their optic neuropathy compared to the phacoemulsification alone group 438, so the risks and benefits of each procedure need to be carefully considered in each case until more definitive evidence is available.

Glaucoma drainage implants

A limited number of studies look at the use of tube shunt devices in primary angle closure glaucoma. Primary angle closure glaucoma eyes are often placed into the category of refractory glaucoma along with other types of glaucoma and thus are not separately evaluated. Based on the limited data available in these studies, tube implants appear effective in controlling IOP in primary angle closure glaucoma 439. Drainage implants are considered more effective than filtering procedures in neovascular glaucoma and ICE syndrome because, in these diseases, a fibrovascular membrane often grows over the sclerostomy site causing bleb failure.

Other surgical procedures

  • May be required depending on the etiology of the underlying disease.
  • Aqueous misdirection: Nd:YAG laser can be used to disrupt the anterior vitreous in aphakic and pseudophakic eyes. If that is not successful, definitive surgical treatment in the form of vitrectromy with anterior hyaloid disruption combined with an anterior chamber deepening procedure may be indicated.
  • Epithelial and/or fibrous downgrowth: radical surgery to remove the intraocular membrane and affected tissues may be necessary.
  • Angle closure that develops after retina surgery may require removing or loosening encircling bands, opening up iridectomy sites, or removing silicone oil.

Surgical follow up

Patients typically need to be watched closely in the immediate postoperative period, sometimes weekly for several months. Once IOP control is achieved and the eye is stabilized, follow up can follow the schedule of routine primary angle closure glaucoma patients.

Angle closure glaucoma prognosis

Primary angle closure suspects

Primary angle-closure suspect or anatomic narrow angle – an eye in which the anterior chamber angle recess has an abnormally narrow angular width. The peripheral iris is located close to, but not touching, the posterior pigmented trabecular meshwork. No peripheral anterior synechiae are present. IOP, optic nerve, and visual field are normal.

The overall likelihood of an individual with a anatomic narrow angle developing acute angle closure glaucoma in the United States is less than 10% 440. A prospective multicenter study, however, found that patients judged to be at risk for developing angle closure glaucoma by experienced ophthalmologists through careful slit lamp exam and gonioscopy had a 30% risk of developing angle closure within 5 years when no prophylactic intervention was performed 415.

In contrast, fellow eyes in which the other eye has already suffered an acute angle closure attack have a much worse prognosis without prophylactic treatment. An untreated fellow eye has a 40% to 50% chance of developing an acute primary angle closure attack over the next 5 to 10 years 425. An iridectomy or iridotomy virtually eliminates the risk 421. The long-term fate (4–6 years) in terms of IOP and glaucomatous optic neuropathy is good for most fellow eyes after laser peripheral iridotomy with the majority not requiring any additional glaucoma treatment and retaining good vision 441.

Acute angle closure glaucoma

In an eye suffering an acute angle-closure attack, the long-term outcomes vary depending on ethnicity (which may be a reflection of mechanism of angle closure), duration of attack, and severity of attack in terms of whether or not it can be aborted by medical treatment alone. If promptly treated, most (60%–75%) symptomatic episodes of angle closure recover without visual field or optic disc damage in the short term 442. Six months post-attack, one study reported 38% of eyes to have visual field loss secondary to nerve fiber bundle loss 442. In another study, 2 – 16 weeks after an attack, changes were noted in optic disk morphology, with preferential loss in the superotemporal and inferotemporal areas 443.

The longer the duration of an attack and the more difficult to manage, the worse the outcome for the eye, regardless of the initial IOP measurement 444. Patients with a 24 to 72 hour delay in presentation had a relative risk of 2.78 for developing chronic glaucoma, whereas those requiring laser iridotomy to control IOP or a trabeculectomy had relative risks of 3.63 and 4.83, respectively 444. In Singapore, risk of visual field loss was significant if the duration of symptoms was longer than 7 days 442.

Even despite successful termination of an acute attack by surgical iridectomy, late IOP increase is reported in 19% to 24% of cases 445. There may be ethnic differences in the rate of IOP control by laser peripheral iridotomy alone 407. A greater extent of peripheral anterior synechiae, a high presenting IOP, and a larger cup to disc ratio have been reported as predictors of poor pressure control following iridotomy in Asian and African eyes 446. A significant portion of patients go on to develop primary angle closure glaucoma and require further medical or surgical intervention in an attempt to control their disease. Unfortunately, up to 20% these patients deteriorate to lose significant vision and are classified as blind in that eye 447.

Chronic angle closure glaucoma

Asymptomatic angle-closure patients present with more severe visual field defects than symptomatic patients 448. In one study which examined visual field loss on presentation, 52.8% of asymptomatic primary angle-closure glaucoma had end stage visual field loss by Advanced Glaucoma Intervention Study criteria compared to only 17.5% of symptomatic primary angle-closure glaucoma cases. Although presenting IOP was considerably higher in the symptomatic group, the level of IOP was not found to be a significant predictor for visual field outcome 449. It is likely that the duration of elevated IOP has a major influence in causing optic nerve damage in primary angle-closure glaucoma, possibly more so than the level of IOP. Once glaucomatous optic neuropathy has developed, almost all cases will require further treatment to control IOP (94%–100%) 450. Closer monitoring of IOP is recommended in chronic angle closure glaucoma compared to primary open angle glaucoma, despite the efficacy of medical therapy. Chronic angle-closure glaucoma tends to progress more quickly and fail medical therapy sooner than primary open angle glaucoma 451. With aggressive management, however, chronic angle-closure glaucoma patients can maintain stable fields and long-term IOP control 411.

Secondary angle closure glaucoma

The prognosis for secondary angle closure patients depends on the underlying cause. Early recognition of the underlying pathology and timely directed treatment helps to improve outcomes.

Glaucoma causes

Scientists aren’t sure what causes glaucoma, but the most common types usually happen when the fluid pressure inside your eye (IOP) slowly rises, damaging the optic nerve. As the optic nerve gradually gets worse, blind spots develop in your vision. For reasons that eye doctors don’t fully understand, optic nerve damage is usually related to increased pressure in the eye.

Other types of glaucoma may be caused by medical conditions, or a baby may be born with it.

Raised eye pressure happens as the result of a buildup of fluid called aqueous humor that flows throughout the inside of your eye. This fluid, called the aqueous humor, usually drains through a tissue located at the drainage angle where the iris and cornea meet. This tissue is called the trabecular meshwork (see Figure 2 above). As new aqueous flows into your eye, the same amount should drain out. This process keeps pressure in the eye (intraocular pressure or IOP) stable. But if the drainage angle is not working properly or when your eye makes too much fluid (aqueous humor), fluid builds up inside your eyeball. Pressure inside your eye rises, damaging the optic nerve. The optic nerve is made of more than a million tiny nerve fibers. It is like an electric cable made up of many small wires. As these nerve fibers die, you will develop blind spots in your vision. You may not notice these blind spots until most of your optic nerve fibers have died. If all of the fibers die, you will become blind.

However, not everyone with high eye pressure develops glaucoma. Some people may even get glaucoma with normal eye pressure. The amount of pressure your optic nerve can handle is different for each person. For reasons unknown, some people can have optic nerves that are sensitive to normal eye pressure. This means their risk of getting glaucoma is higher than normal. Getting regular dilated eye exams helps your eye doctor figure out what level of eye pressure is normal for you and find early signs of damage to their optic nerve.

Risk factors for glaucoma

Several risk factors can contribute to glaucoma:

  • Age. Most types of glaucoma affect people age 40 and older (congenital glaucoma is the biggest exception to this). Experts estimate that 10% of people age 75 and older have glaucoma.
  • Race. Black people have a much higher risk of developing primary open-angle glaucoma, especially people of Afro-Caribbean descent. People of African descent are 15 times more likely to have blindness from open-angle glaucoma. People of Asian and Inuit descent have a higher risk of angle-closure glaucoma.
  • Sex. Women have a higher risk of angle-closure glaucoma. Experts suspect this is mainly because of sex-linked differences in eye anatomy.
  • Refractive errors. People with extreme nearsightedness (myopia) have a higher risk of open-angle glaucoma. People with extreme farsightedness (hyperopia) have a higher risk of angle-closure glaucoma.
  • Family history. There’s evidence that a family history of glaucoma, especially a first-degree biological relative (a parent, child or sibling), means you also have a higher risk of developing it. And several conditions that cause secondary glaucoma are genetic, too 222.
  • Certain medical conditions. Research links some conditions, like high blood pressure (hypertension), migraine, sickle cell anemia and diabetes, to much higher odds of developing glaucoma.
  • Corneas that are thin in the center.
  • Eye injury
  • Certain types of eye surgery.
  • Taking corticosteroid medicines, especially eye drops, for a long time.

If you’re at higher risk for glaucoma, you should get comprehensive eye exams every 1 to 2 years. You’re at higher risk if you:

  • Are Black or African American and over age 40
  • Are over age 60, especially if you’re Hispanic or Latino
  • Have a family history of glaucoma

You’re also at higher risk if you have high blood pressure or diabetes and should get a dilated eye exam at least once a year. Talk with your eye specialist about your risk and how often you should get your eyes checked.

Glaucoma pathophysiology

The exact cause of glaucoma is not fully known, but the underlying pathology lies in the apoptosis of retinal ganglion cells 221. Retinal ganglion cells are the third class of photoreceptors recently noted in scientific literature for their intrinsic photosensitivity. Retinal Ganglion Cellss form functional microcircuits with rods, cones, amacrine cells, and bipolar cells that help transmit image-forming and non-image-forming information to the brain. Retinal Ganglion Cells axons target the suprachiasmatic nucleus, intergeniculate leaflet, olivary pretectal nucleus, ventral division of the lateral geniculate nucleus, and the preoptic area. Retinal Ganglion Cellss play a role in modulating circadian rhythm, releasing melatonin, regulating pupil size, and forming relays in on-off retina centers 452. Retinal Ganglion Cells axons remain unmyelinated until they have passed through the lamina cribrosa and converged to form the optic nerve. After passing through the lamina cribrosa, oligodendrocytes myelinate the axons of the Retinal Ganglion Cells. Iatrogenically induced ocular hypertension in mice, cats, and monkeys has shown blockade of orthograde and retrograde axonal transport, integral in transporting growth factors such as brain-derived neurotrophic factors (BDNF). Regardless of whether intraocular pressure (IOP) is elevated, normal, or decreased, the death of Retinal Ganglion Cellss underlies glaucoma pathology 453, 454.

The optic nerve carries over 1 million nerve fibers that transmit visual signals from the photoreceptors in the outer retina to the visual processing areas of the occipital lobe. Damage to the retinal nerve fiber layer occurs in various types of glaucoma. Aqueous humor, the fluid in the anterior chamber of the eye, is crucial in maintaining intraocular pressure and nourishing ocular tissues. Aqueous humor is produced by the non-pigmented epithelial cells of the ciliary body processes and follows a circadian production pattern 455. Aqueous humor drains continuously through the pupil, then via the trabecular meshwork anterior to the scleral spur and iris insertion, into Schlemm canal, and finally into the episcleral venous system, larger orbital venous system, and systemic venous circulation. The trabecular meshwork consists of multiple layers of connective tissue and the endothelium of Schlemm canal, forming the primary drainage pathway for aqueous humor 456.

Aqueous humor dynamics and circulation are vital in the pathogenesis of glaucomatous damage to the optic nerve 10, 16, 17. One of the primary risk factors and causative factors in glaucoma is an elevated intraocular pressure (IOP) above the normal physiologic range of 12 to 21 mmHg 10, 16, 17. Extended periods of elevated intraocular pressure (IOP) above 22 mmHg (especially if IOP rises above 26 mm Hg to 30 mm Hg) have been associated with development of glaucomatous damage 10, 16, 17. About 40% to 50% of all open-angle glaucoma cases have intraocular pressure (IOP) below 22 mm Hg in a single screening 384, 457. Elevated intraocular pressure from increased resistance and decreased outflow of aqueous humor and/or increased production leads to compression of the optic nerve which limits oxygen and nutrition supply to the nerve. There are two main categories of glaucoma, each involving aqueous humor circulation and production in relation to resulting optic neuropathy. Reduction of intraocular pressure (IOP) leads to less progression or stabilization of the glaucomatous optic nerve and visual field changes 335.

Primary open angle glaucoma (POAG) causes optic neuropathy in the setting of an ‘open angle’, while primary angle closure glaucoma (PACG) in the setting of peripheral angle obstruction or narrowing 10, 16, 17. The ‘angle’ refers to the intersection of the peripheral iris with the cornea, a key anatomical player in conventional aqueous humor drainage 10, 16, 17 as this is where the trabecular, Sclemm’s canal are located. In primary open angle glaucoma (POAG) where the iris-cornea angle remains open for drainage, the mechanism for glaucomatous damage includes overall increased resistance to outflow despite maintaining an open angle, and increased aqueous humor production 10, 16, 17. In primary angle closure glaucoma (PACG), the iris-cornea angle is acutely or chronically obstructed which leads to significant decrease in aqueous humor outflow and an elevation in intraocular pressure 10, 16, 17.

Ocular hypertension is defined as elevated intraocular pressure without any signs of vision loss or optic neuropathy otherwise classified as glaucomatous changes 458. Patients with ocular hypertension have demonstrated reduced trabecular outflow and uveoscleral outflow with normal aqueous production which explains the mechanism for elevated intraocular pressure 458. This is a similar mechanism to patients with primary open angle glaucoma (POAG). However, the understanding of elevated intraocular pressure in the setting of ocular hypertension or primary open angle glaucoma (POAG) is limited, such as in situations with ocular hypertension without primary open angle glaucoma (POAG) or normal tension glaucoma 458.

Current glaucoma pharmacologic and surgical therapies are targeted at decreasing aqueous humor production, such as carbonic anhydrase inhibitors or ciliary body ablative laser treatments, or increasing outflow, such as prostaglandin agonists, adrenergic agonists, and physical modifications to optimize or bypass the outflow pathways 10, 17.

Glaucoma prevention

Glaucoma isn’t preventable, but early detection and treatment can manage pressure inside your eye and help prevent or delay vision loss. There are a few main ways that can happen:

  • Regular eye exams. Pressure increases in your eyes are often detectable before they can cause glaucoma and its symptoms. Early detection lets you and your eye specialist try to prevent glaucoma from worsening or, at least, slow down its progress.
  • Managing ocular hypertension. If you have higher-than-normal pressure in your eyes, your eye care specialist can offer treatment options. Following their guidance can be crucial and help you avoid — or at least delay — vision loss.
  • Knowing and managing your risk factors. Some of the conditions that you can try to manage or prevent include high blood pressure and diabetes. Using protective items like safety glasses and goggles can help prevent eye injuries.
  • Eat a healthy diet. Eating a healthy diet can help you maintain your health, but it won’t prevent glaucoma from worsening. Several vitamins and nutrients are important to eye health, including zinc, copper, selenium, and antioxidant vitamins C, E and A.
  • Exercise regularly. Regular exercise may reduce eye pressure.
  • Limit your caffeine. Drinking beverages with large amounts of caffeine may increase your eye pressure.
  • Take prescribed medicine. Using your eye drops or other medicines as prescribed can help you get the best possible result from your treatment. Be sure to use the eye drops exactly as prescribed. Otherwise, your optic nerve damage could get worse.

Glaucoma signs and symptoms

In its early stages, glaucoma may not cause any symptoms or have no warning signs. This is why glaucoma is called the “silent thief of sight”. Up to half of the people in the United States with glaucoma may not know they have it. And symptoms may not appear until glaucoma causes irreversible eyesight damage. Having regular eye exams can help your ophthalmologist find this disease before you lose vision. Your ophthalmologist can tell you how often you should be examined.

Some of the more common glaucoma symptoms include:

  • Eye pain or pressure
  • Headaches
  • Red or bloodshot eyes
  • Double vision (diplopia)
  • Blurred vision
  • Gradually developing low vision
  • Gradually developing blind spots (scotomas) or visual field defects like tunnel vision

Some types of glaucoma, particularly angle closure glaucoma, can cause sudden, severe symptoms that need immediate medical attention to prevent permanent vision loss. Emergency glaucoma symptoms include:

  • Blood gathering in front of your iris (hyphema)
  • Bulging or enlarged eyeballs (buphthalmos = “ox eye”)
  • Nausea and vomiting that happen with eye pain/pressure
  • Rainbow-colored halos around lights
  • Sudden appearance or increase in floaters (myodesopsias)
  • Sudden vision loss of any kind
  • Suddenly seeing flashing lights (photopsias) in your vision.

Open-angle glaucoma symptoms

With open-angle glaucoma, there are no warning signs or obvious symptoms in the early stages. As the disease progresses, blind spots develop in your peripheral (side) vision. Side vision also is called peripheral vision. In later stages, difficulty seeing things in your central vision.

Most people with open-angle glaucoma do not notice any change in their vision until the damage is quite severe. This is why glaucoma is called the “silent thief of sight”. Having regular eye exams can help your ophthalmologist find this disease before you lose vision. Your ophthalmologist can tell you how often you should be examined.

Acute angle-closure glaucoma symptoms

People at risk for angle-closure glaucoma usually show no symptoms before an attack. Some early symptoms of an attack may include blurred vision, halos, mild headaches or eye pain. People with these symptoms should be checked by their ophthalmologist as soon as possible. An attack of angle-closure glaucoma includes the following:

  • Severe eye pain
  • Bad headache or forehead pain
  • Eye redness
  • Decreased vision or blurred vision
  • Seeing rainbows (colored rings around lights) or halos
  • Headache
  • Nausea
  • Vomiting.

Normal-tension glaucoma symptoms

  • No symptoms in early stages.
  • Gradually, blurred vision.
  • In later stages, loss of side vision.

Glaucoma in children symptoms

  • A dull or cloudy eye (infants).
  • Increased blinking (infants).
  • Tears without crying (infants).
  • Blurred vision.
  • Nearsightedness that gets worse.
  • Headache.

Pigmentary glaucoma symptoms

  • Halos around lights.
  • Blurred vision with exercise.
  • Gradual loss of side vision.

Glaucoma complications

Glaucoma damages your retina and optic nerve to the point where they stop working, resulting in vision loss and, eventually, total blindness. The end result is what experts call “absolute glaucoma” – meaning you’re totally blind in the affected eye. The affected eye feels hard and maybe even painful when you touch it.

Glaucoma diagnosis

The only sure way to diagnose glaucoma is with a complete eye exam. An eye specialist can diagnose glaucoma using an eye exam, including several tests that are part of routine eye exams. A comprehensive eye exam can detect glaucoma long before you have eye damage and the symptoms that follow. Many of these tests involve pupil dilation (mydriasis), so your eye doctor can get a better look inside your eye. Your eye care specialist examines your eyes using a special magnifying lens. This provides a clear view of important tissues at the back of your eye to check for glaucoma or other eye problems. For a few hours after the exam your vision may be blurry and sensitive to light, so you will need someone to take you home.

Some of the most helpful glaucoma tests include:

  • Visual acuity testing. A visual acuity test assesses how clearly someone can see at a distance, typically using a Snellen chart or other standardized chart. The test is performed by an optometrist or ophthalmologist and involves reading progressively smaller letters or identifying shapes, with the results expressed as a fraction like 20/20 or 6/6, indicating the distance at which the person can see the letters or shapes
  • Visual field testing also called perimetry. This check of your peripheral (side) vision allows your eye care provider to find out how well you can see objects off to the side of your vision without moving your eyes. This test measures the entire area the forward-looking eye sees to document straight-ahead (central) and side (peripheral) vision. It measures the dimmest light seen at each spot tested. Each time patients perceive a flash of light, they respond by pressing a button.
  • Depth perception testing. A depth perception test assesses your ability to see the world in three dimensions (3D) and judge distances accurately. It checks if your eyes work together and if your brain processes the visual information correctly. These tests use 3D images or patterns like the Randot Stereo test to gauge how well your eyes coordinate to perceive depth. Some tests involve holding a finger in front of your eyes and focusing on a distant object, checking for double vision of the finger.
  • Tonometry. This measures the pressure inside your eye. Increased eye pressure is the most important risk factor for glaucoma. There are several methods of measuring eye pressure. The most common method is known as applanation, in which a tiny instrument contacts the eye’s surface after it is numbed with an eye drop.
    • Air-puff test. You’ll rest your chin on a machine and your eye specialist will blow a puff of air into your eye. This quick and painless test is used as part of a routine glaucoma screening. If the results show that your eye pressure is high, your eye specialist will do other eye-pressure tests to get a more accurate measurement.
    • Applanation tonometry. Your eye specialist will numb your eyes with drops before measuring your eye pressure using one of these methods:
      • You’ll rest your chin on a special magnifying device called a slit lamp. Your eye care specialist will examine your eye through the slit lamp while gently pressing a special tool on your eye to test the pressure.
      • Your eye care specialist will gently press a handheld device against your eye. The device measures your eye pressure.
  • Pachymetry. Pachymetry is a simple, painless test that measures the thickness of the cornea, the clear front part of the eye. The eye doctor uses an ultrasonic wave instrument to help determine the thickness of the cornea and better evaluate eye pressure.
  • Ophthalmoscopy. Your eye care specialist will do a dilated eye exam to look for damage to your optic nerve. This exam is part of a routine glaucoma check-up. You’ll be given eye drops that widen (dilate) your pupils (the openings that let light into your eyes). You’ll look straight ahead while your eye care specialist looks into your eye using a device with a light and magnifying lens.
  • Slit lamp exam. A slit lamp exam is a common eye test that uses a microscope with a focused beam of light to examine the front of your eye and the back of your eye with the aid of special lenses.
  • Gonioscopy. Gonioscopy is a specialized eye examination that allows an ophthalmologist to visualize the anterior chamber drainage angle, the space between the iris and the cornea where fluid drains out of the eye. Gonioscopy is a crucial part of diagnosing and monitoring glaucoma and other eye conditions. Eye doctors regularly examine the drainage angle to see if there is any visible obstruction to fluid leaving the eye through the trabecular meshwork. A special lens (gonioscopy lens) is needed to examine the trabecular meshwork. The gonioscopy lens is gently placed against the surface of the cornea and allows eye doctors to see the trabecular meshwork in the drainage angle.

If your eye specialist has a reason to suspect damage to your retina and/or optic nerve, they may also use additional types of eye imaging. These include:

  • Optical coherence tomography (OCT). Optical Coherence Tomography (OCT) measures the reflection of laser light similar to the way that ultrasound measures the reflection of sound. Using this device, a 3D reconstruction of the optic nerve can be created. Optical coherence tomography (OCT) is valuable for monitoring morphological changes in the optic nerve and retinal nerve fiber layer, especially in patients with ocular hypertension and early-to-moderate glaucoma 117. The most recent advances of OCT include OCT-A, or OCT-Angiography, whereby the blood flow to vessels surrounding the optic nerve and in the macula can be measured. This is still an active area of research, but scientists do know that some patients’ optic nerves are very vulnerable to changes in optic nerve blood flow, and this new measurement may be useful in evaluating these patients.
  • Heidelberg Retina Tomograph (HRT): Heidelberg Retina Tomograph (HRT) is also a laser that can produce a 3D representation of the optic nerve.
  • Nerve Fiber Analyzer (GDx): Nerve Fiber Analyzer (GDx) uses laser light to measure the thickness of the nerve fiber layer.
  • Fluorescein angiography. Fluorescein angiography is a diagnostic test used to examine the blood vessels in the retina and choroid of the eye. Fluorescein angiography involves injecting a fluorescent dye into the bloodstream and taking photographs of your retina and its blood vessels as the dye circulates, revealing potential blockages, leaks, or other abnormalities in the blood vessels. Fluorescein angiography is often recommended to find and diagnose eye disease including 118:
    • macular edema (swelling in the retina that distorts vision)
    • diabetic retinopathy (damaged or abnormal blood vessels in the eye caused by diabetes)
    • macular degeneration
    • blockage of veins inside the eye, called branch retinal vein occlusion (BRVO) or central retinal vein occlusion (CRVO)
    • macular pucker (a wrinkle in the retina caused by a buildup of fluid behind it)
    • ocular melanoma (a type of cancer affecting the eye)
    • rack changes in eye disease over time
    • target treatment areas
  • Less commonly, ultrasound, computed tomography (CT) or magnetic resonance imaging (MRI).

Figure 16. Goldmann Applanation Tonometry

Goldmann Applanation Tonometry

Figure 17. Gonioscopy (the drainage angle is examined using a special lens)

Gonioscopy

Footnotes: Gonioscopy. (A) and (B) Gonioscopy lens. (C) The gonioscopy lens is gently held against the cornea. Eye doctors look through the gonioscopy lens to see the drainage angle.

[Source 459 ]

Figure 18. Humphrey Field Analyzer

Humphrey Field Analyzer

Optic Nerve

The optic nerve should be evaluated using a slit lamp and 90D or 78D lens so that the three-dimensional features of the optic nerve are appreciated 335. The inferior neuroretinal rim (neuroretinal rim) is the thickest, followed by superior, nasal, and temporal neuroretinal rim; this is called the ISNT rule (inferior [I] ≥ superior [S] ≥ nasal [N] ≥ temporal [T] rule) 460, 461. The ISNT rule (inferior [I] ≥ superior [S] ≥ nasal [N] ≥ temporal [T]) refers to the characteristic pattern of neuroretinal rim width in a normal optic disc 460. In a healthy eye, the neuroretinal rim is typically broadest inferiorly, followed by superiorly, nasally, and thinnest temporally 460. This pattern, represented as inferior [I] ≥ superior [S] ≥ nasal [N] ≥ temporal [T], can be used as a reference point for evaluating optic disc rim changes, especially in the context of glaucoma diagnosis. In open-angle glaucoma, the superior and inferior neuroretinal rim are thinned, breaking the ISNT rule 335. The optic cup should be determined by its contour and not color. A recent Journal of the Americal Medical Association Rational Clinical Examination systematic review of primary open-angle glaucoma diagnosis found that the risk of glaucoma was highest when an examination revealed an increased cup-disk ratio (CDR), cup-disk ratio asymmetry, disc hemorrhage, or elevated intraocular pressure (IOP) 462.

Typical optic nerve head changes in open-angle glaucoma include 335:

  • Diffuse or focal narrowing (notching/shelving) of the neuroretinal rim precisely at the superior or inferior part of the optic disc
  • Symmetrically enlarged cup-to-disc ratios greater than 0.5
  • Increased vertical cup-to-disc ratio and thinning of neuroretinal rim
  • Asymmetry of cup-disk ratio (CDR) of 0.2 or more
  • Hemorrhage at or around the optic disc
  • Peripapillary atrophy
  • Baring of circumlinear vessels (the gap between the superficial vessels and disc margin)
  • Bayonetting of vessels (the vessel first goes back and then climbs along the wall of the deep cup and then angles again on the disc margin)
  • Very deep (excavated) cup with bean-pot cupping and laminar dot sign
  • Nasalization of optic disc vessels
  • Diffuse or focal (arcuate) thinning/defect of the retinal nerve fiber layer contiguous with an area of neuroretinal rim-notch
  • The neuroretinal rim is typically pink and not pale in open-angle glaucoma. The pallor of the neuroretinal rim usually denotes an atrophic optic nerve, as seen in primary angle closure glaucoma (POAG).

Figure 19. ISNT rule

ISNT rule

Footnotes: ISNT rule for a normal optic nerve. The ISNT rule is that optic disc rim thickness shows a characteristic configuration of inferior (I) greater than or equal to superior (S) greater than or equal to nasal (N) greater than or equal to temporal (T) (or inferior [I] ≥ superior [S] ≥ nasal [N] ≥ temporal [T]).

[Source 460 ]

Figure 20. Glaucomatous Optic Nerve

Glaucomatous Optic Nerve

Footnotes: (A) Normal optic nerve. The pink area of neural tissue forms the neuroretinal rim, whereas the central empty space corresponds to the cup. (B) Glaucomatous optic nerve showing loss of superior neural retinal rim (thinning) and excavation with enlargement of the cup. The arrowheads point to an associated retinal nerve fiber layer defect, which appears as a wedge-shaped dark area emanating from the optic nerve head. The superior neural losses correspond to the inferior defect (black scotoma) seen on the visual field. There is also a small retinal nerve fiber layer defect inferiorly, but the corresponding hemifield of the visual field remains within normal limits. (C) More extensive neural tissue loss from glaucoma with severe neuroretinal rim loss, excavation, and enlargement of the cup. There is severe loss of visual field both in the superior as well as in the inferior hemifield.

[Source 221 ]

Figure 21. Optical Coherence Tomography (OCT) of Glaucomatous Optic Nerve

Optical Coherence Tomography of Glaucomatous Optic Nerve

Footnotes: (A) The arrowheads point to a retinal nerve fiber layer (RNFL) defect. (B) Areas of thicker retinal nerve fiber layer (RNFL) appear in yellow and red. Arrowheads point to the retinal nerve fiber layer (RNFL) defect. A deviation map compares the retinal nerve fiber layer (RNFL) thickness values with a normative database and highlights the defect. (E) Arrowheads point to a visual field defect.

[Source 221 ]

Visual Field

Perimetry also known as visual field testing is an important diagnostic tool that maps out your visual field on a printout, making it helpful and necessary in diagnosing and managing open-angle glaucoma. Perimetry testing provides a baseline visual field for glaucoma suspects and confirmed open-angle glaucoma cases so clinicians can track disease progression 335.

To make a diagnosis of acquired glaucomatous visual field defect, Hoddap–Parrish–Anderson criteria are used 463:

  • Glaucoma hemifield test outside normal limits on at least 2 fields.
  • A cluster of 3 or more non-edge points in a location typical for glaucoma, all of which are depressed on the pattern deviation plot at a perimetry (P) <5% and one of which is depressed at a perimetry (P) <1% on 2 consecutive fields.
  • A corrected pattern standard deviation that occurs in less than 5% of normal fields on 2 consecutive fields.

Static automated threshold perimetry is used with white stimulus on a white background. Most studies used the Humphrey Field Analyzer, but other perimeters like Octopus have also been used successfully. Non-conventional perimetry like SWAP (short-wavelength automated perimetry using blue stimulus on yellow background), pulsar perimetry, rarebit, Matrix and frequency-doubling technology have been proposed in earlier studies for the detection of early glaucoma visual field loss, however, tend not to be used in routine clinical practice 464, 465, 466, 467, 468, 469, 470.

The visual field must be reliable, and defects should be repeatable on at least 2 fields. When treating patients long-term, it is preferable to use the same machine, the same degree of field, and protocol (eg, 24-2, 30-3, or 10-2) to compare the fields to note for progression or stability. At least 40% to 50% ganglion cell loss is needed to reliably show visual field defects in threshold perimetry 471, 472.

Structural changes of the optic nerve and retinal nerve fiber layer (RNFL) tend to occur earlier than functional change (visual field loss) in most patients with open-angle glaucoma. This is relevant to the concept of preperimetric glaucoma, which is defined as ‘the presence of characteristic glaucomatous changes in the optic disc and increased vulnerability to damage in the retinal nerve fiber layer (RNFL), without the presence of visual field defects detectable with standard automated perimetry 473. For patients with risk factors, suspect optic discs, and/or ocular hypertension, periodic visual field and optical coherence tomography (OCT) testing are recommended to detect early visual field defects and changes to determine whether therapy is needed.

Typical visual field changes in open-angle glaucoma include 335:

  • Increased variability of responses in an area that later developed field defects
  • Asymmetry of the visual field between the eyes
  • Paracentral scotoma- commonly superonasal
  • Roenne’s nasal step- an area of depression above or below the horizontal meridian on the nasal side
  • Temporal wedge
  • Sickle-shaped (Seidel) scotoma
  • Bjerrum scotoma or arcuate scotoma
  • Annular or ring scotoma when arcuate scotoma is present on both above and below the horizontal meridian
  • General constriction of peripheral field
  • A temporal island of the visual field.

Figure 22. Hoddap–Parrish–Anderson criteria

Hoddap–Parrish–Anderson criteria

Abbreviations: MD = mean deviation; dB = decibels (refers to the logarithmic scale used to measure visual sensitivity and it represents the intensity of light stimuli that a person can see at a specific location in their visual field)

[Source 463 ]

Intraocular Pressure

Intraocular pressure (IOP) is measured with tonometry and several different tonometers are used 474, 475. The gold standard for intraocular pressure (IOP) measurement is Goldmann Applanation Tonometry (GAT) 476. However, Goldmann Applanation Tonometry is not available in all instances, and non-contact tonometry is also frequently used. In the UK, this is certainly true in the community where optometrists’ preferred method of IOP measurement is non-contact 336.

When determining the intraocular pressure (IOP) of a patient using tonometry, certain variables must be considered. Tonometry measurements can, for example, vary between examiners by approximately 10% per individual, translating to a difference in intraocular pressure (IOP) measurement of 1 mm Hg to 2 mm Hg 477.

An individual’s corneal thickness measured with pachymetry or diurnal variations of intraocular pressure (IOP) (eg, higher intraocular pressure (IOP) in early morning hours or other times of the day or variability in the time of day of maximal intraocular pressure (IOP) between patients) can also have a tremendous effect on the accuracy of intraocular pressure (IOP) measurements. Study results have shown that intraocular pressure (IOP) is overestimated in individuals with thick corneas or central corneal thickness (CCT) while underestimated in those with thinner corneas 478, 479. Newer methods of IOP measurement aim to overcome variations in corneal biomechanics and give a more accurate estimate of true IOP. These include the Reichert Ocular Response Analyzer (ORA) and the Pascal Dynamic Contour Tonometer (DCT). The ORA is a non-contact tonometer that measures a biomechanical attribute of the cornea termed hysteresis. The DCT uses uses principle of contour matching instead of applanation to reduce the effect of corneal biomechanics. Furthermore, multiple measurements should be taken and correlated with optic nerve and visual field examinations when a patient is suspected of elevated intraocular pressure (IOP).

If previous tonometry measurements are available, they should be reviewed and compared to the most recent ones. Also, the intraocular pressure (IOP) may be different on different days, and different instruments may capture different values of intraocular pressure (IOP). If a difference of 3 mm Hg or more is noted between the 2 eyes, there is an increased suspicion of glaucoma 335. Clinicians should expect approximately 10% variation between individual and repeat measurements over 2 to 3 occasions before deciding on the treatment 335.

Elevated intraocular pressure (IOP) is an important and modifiable risk factor; however, it is not a diagnostic factor for open-angle glaucoma. An ophthalmologist should check the patient’s intraocular pressure (IOP) using applanation tonometry, remaining aware that the applanation tonometry test causes patients to squeeze their eyes and elevate their pressure readings. Normal intraocular pressure (IOP) should range between 12 mm Hg and 21 mm Hg 335. Approximately 75% of patients with elevated intraocular pressure (IOP) never develop glaucomatous optic nerve atrophy or visual field deficits. When a patient has recorded a reliably high intraocular pressure (IOP) reading above 21 mm Hg, they are deemed patients with glaucoma or patients with ocular hypertension 384, 480.

Corneal Photokeratoscopy

Corneal photokeratoscopy or corneal topography, is a potential biological marker to monitor patients with primary open-angle glaucoma (POAG). Preliminary results have shown a forward shift of the posterior and anterior corneal surfaces; this is correlated with more advanced stages of functional damage, indicating a link between corneal structural changes and the duration and intensity of elevated IOP. Further studies are needed to validate this marker in patients with primary open-angle glaucoma (POAG) 481.

Glaucoma Stages

The Americal Academy of Ophthalmology’s classifies the severity of glaucomatous damage into different categories 398:

  • Mild: Definite optic disc or retinal nerve fiber layer (RNFL) abnormalities consistent with glaucoma as detailed above and a normal visual field tested with standard automated perimetry (SAP) are seen.
  • Moderate: Definite optic disc or retinal nerve fiber layer (RNFL) abnormalities consistent with glaucoma, as detailed above, are seen, and visual field abnormalities in one hemifield are not within 5° of fixation as tested with standard automated perimetry (SAP).
  • Severe: Definite optic disc or retinal nerve fiber layer (RNFL) abnormalities consistent with glaucoma as detailed above are seen, and visual field abnormalities in both hemifields or loss within 5° of fixation in at least one hemifield as tested with standard automated perimetry (SAP).
  • Indeterminate: Definite optic disc or retinal nerve fiber layer (RNFL) abnormalities consistent with glaucoma, as detailed above, are seen, and the patient cannot perform visual field testing, has unreliable or uninterpretable visual field test results, or has not performed visual fields yet.

Several other different staging systems are recognized based on visual field damage, optic disc cupping, and retinal nerve fiber layer (RNFL) defects with optical coherence tomograpy (OCT), some of which are applicable in a routine clinical setting or clinical trials 482, 483, 484, 485, 486, 487.

Glaucoma treatment

The main goal of treating glaucoma is to keep it from getting worse by lowering the pressure inside your eye (IOP).

Some of the most likely treatments for this include:

  • Medications. This mainly involves medications that lower pressure inside your eye. They can prevent glaucoma from developing if you have higher-than-normal intraocular pressure (ocular hypertension) or keep it from worsening enough to cause damage and symptoms.
  • Glaucoma surgeries. These mainly focus on improving the drainage of aqueous humor fluid to lower pressure inside your eye. Surgery options include trabeculectomy, tube shunts, laser therapy and minimally invasive glaucoma surgeries (MIGS).

Other treatments are possible, depending on what type of glaucoma you have, how it’s affecting your eye and other factors. Your eye specialist can tell you more about your treatment options and help you choose one that fits your needs best.

Table 1. Glaucoma medications

Intraocular pressure lowering agents
Drug ClassExampleMechanismIOP LoweringDosingSide EffectsNotes
Prostaglandin F2a analoguesLatanoprost (Xalatan)

Travoprost (Travatan)

Bimatoprost (Lumigan)

Tafluprost (Zioptan; preservative free)

Latanoprostene bunod (Vyzulta)

Increase outflow via uveoscleral pathway; decrease outflow resistance; mechanism unclearLatanaprost and Travoprost: 25-32%

Bimatoprost: 27-33%

Tafluprost: 27-31%

Latanoprostene:
29-32%1

As a class, most potent

Once daily, usually in eveningOcular: Irritation, increased pigmentation of iris, lashes, and skin, hypertrichosis, prostaglandin associated periorbitopathy, loss of orbital fat over time
Often preferred as first line; highly effective at IOP lowering, with minimal systemic side effects.

Among preservative containing drops, side effects tend to be greatest with bimatoprost, and least with latanoprost.

Preservative free tafluprost may reduce irritation and ocular side effects and improve compliance in some patients.2

Systemic:  flu-like symptoms, myalgias and arthralgias, nasal congestion; generally very well tolerated
Beta blockers (beta-adrenergic antagonists)Non-Selective: Timolol maleate (Timoptic;
Timoptic occudose = preservative free)
Timolol hemihydrate (Betimol)Levobunolol (Betagan)MetipranololCarteolol HCl (partial alpha agonist)
Decrease aqueous production20-30%1-2 times daily

OR

Once daily in morning; more effective in AM

Ocular: Blurring, irritation, punctate keratitis; metipranolol associated with uveitisGenerally well tolerated from the perspective of ocular side effects.

History of asthma or other airway disease is a strong contraindication.

Suspect side effects of therapy in a patient with new onset depression, lethargy, or sexual dysfunction.

Carteolol may have less detrimental effect on lipid profile in some patients.3

Betaxolol may be slightly less likely to cause asthma or lung disease exacerbation, but still carries significant risk.4

Systemic: symptoms of b-blockade: heart block and bradycardia, decreased exercise tolerance, asthma and lung disease exacerbation, decreased symptoms of hypoglycemia in diabetes, depression, sexual dysfunction, lipid profile changes
Selective b 1:
Betaxalol
Same15-20%2 times daily
Alpha agonists (Alpha2-adrenergic agonists)Apraclonidine HCl (Iopidine)

Brimonidine tartrate (Alphagan)

Decreases aqueous production, increases uveoscleral outflow to lesser extent20-30%2-3 times dailyOcular: Irritation, allergy, pruritis, dry eyeAs a class, most likely to cause ocular irritation.

High risk of apnea and CNS depression in infants with brimonidine. If CNS depression is a particular concern in any patient, apraclonidine does not cross blood-brain barrier and may be better choice.5,6

Systemic: Lethargy, hypotension, vasovagal attack, headache, dry mouth and nose, insomnia, anxiety; risk of apnea and CNS depression in infants with brimonidine, apraclonidine will not cross blood-brain barrier
Carbonic anhydrase inhibitorsOral:
Acetazolamide (Diamox)
Methazolamide (Neptazane)
Decreases aqueous production15-20%Acetazolamide: 500-1000mg per day in 2-4 doses

Methazolamide: 25-50mg 2-3 times per day

IV Acetazolamide: 5-10mg/kg q6-8 hours

Ocular: None
Sulfa allergies are often cited as a contraindication to use of these drugs, but there is little structural overlap with antibiotic sulfa drugs, and thus most patients allergic to sulfa drugs will not be affected by carbonic anhydrase inhibitors. 7
Systemic: hypokalemia, poor tolerance of carbonated beverages, acidosis, paresthesias, blood dyscrasias, lethargy, nephrolithiasis, others

 

Topical:
Dorzolamide (Trusopt)
Brinzolamide (Azopt)
SameGenerally not as effective as oral acetazolamide2 times daily (only FDA approved for 3 times daily)Ocular:  Induced myopia, blurred vision, stinging (less with Brinzolamide), allergic conjunctivitis, keratopathyGenerally very safe, often used in infants and pregnant women.
Systemic: Less than with oral agents; bad taste in mouth
Parasympathomimetic agentsPilocarpine HCl

Echothiophate iodide (Phospholine iodide)

Miotics; increase trabecular outflow; ciliary muscles contract, put traction on scleral spur, open trabecular meshwork15-25%Pilocarpine: 2-4 times daily, usually 4 times daily

Echothiopentate: 1-2 times daily, usually 2 times daily

Ocular: Posterior synechiae, intense miosis, keratitis, cataract, retinal detachment, angle closure, epiphora, induced myopiaConsider echothiophate in patients with a complicated anterior segment, e.g. aphakia, PK, ACIOL, etc., with inadequate pressure control. In certain patients it can be the sole effective drug.8
Systemic: increased salivation, increased gastric secretion, abdominal cramps
Rho kinase inhibitorsNetarsudil (Rhopressa)Increases trabecular outflow, decreases episcleral venous pressure9Reduces IOP by ~5.5-6 mmHg regardless of baseline IOP10FDA approved for once daily, more efficacious when used twice daily in studiesOcular: conjunctival hyperemia and hemorrhage, corneal deposits (verticillata), blurry vision, epiphora, pruritis, punctate keratitis, eyelid erythema, conjunctival edema, foreign body sensationThough not as effective at IOP reduction as other classes, may have benefit in patients with lower baseline IOP (IOP reduction less dependent on baseline IOP than other classes), and as an adjunct to other drugs, as mechanism of action (increases trabecular outflow rather than uveoscleral) is different from the most commonly used classes.10
Systemic: None achieving statistical significance in recent studies10
Hyperosmotic agentsMannitol 20% (parenteral)
Glycerol 50% (oral)
Creates osmotic gradient, dehydrates vitreousMannitol: 0.5-2.0 g/kg
Glycerol: 1-1.5 g/kg
Ocular: IOP rebound, aqueous flare
Systemic: urinary retention, headache, nausea, vomiting, diarrhea, electrolyte disturbances, cardiac complications, contraindicated in renal failure
[Source 488 ]

Eye drops

Glaucoma treatment often starts with prescription eye drops. Some may decrease eye pressure by improving how fluid drains from your eye. Others decrease the amount of fluid your eye makes. Depending on how low your eye pressure needs to be, more than one eye drop may be prescribed.

Prescription eye drop medicines include:

  • Prostaglandins. These increase the outflow of the fluid in your eye, helping to reduce eye pressure (IOP). Medicines in this category include latanoprost (Xalatan), travoprost (Travatan Z), tafluprost (Zioptan), bimatoprost (Lumigan) and latanoprostene bunod (Vyzulta). Possible side effects include mild reddening and stinging of the eyes, darkening of the iris, darkening of the pigment of the eyelashes or eyelid skin, and blurred vision. This class of medicine is prescribed for once-a-day use.
  • Beta blockers. These reduce the production of fluid in your eye, helping to lower eye pressure. Examples include timolol (Betimol, Istalol, Timoptic) and betaxolol (Betoptic S). Possible side effects include difficulty breathing, slowed heart rate, lower blood pressure, impotence and fatigue. This class of medicine can be prescribed for once- or twice-daily use depending on your condition.
  • Alpha-adrenergic agonists. These reduce the production of the fluid that flows throughout the inside of your eye. They also increase the outflow of fluid in the eye. Examples include apraclonidine (Iopidine) and brimonidine (Alphagan P, Qoliana). Possible side effects include irregular heart rate; high blood pressure; fatigue; red, itchy or swollen eyes; and dry mouth. This class of medicine is usually prescribed for twice-daily use but sometimes can be prescribed for use three times a day.
  • Carbonic anhydrase inhibitors. These medicines reduce the production of fluid in your eye. Examples include dorzolamide and brinzolamide (Azopt). Possible side effects include a metallic taste, frequent urination, and tingling in the fingers and toes. This class of medicine is usually prescribed for twice-daily use but sometimes can be prescribed for use three times a day.
  • Rho kinase inhibitor. This medicine lowers eye pressure by suppressing the rho kinase enzymes responsible for fluid increase. It is available as netarsudil (Rhopressa) and is prescribed for once-a-day use. Possible side effects include eye redness and eye discomfort.
  • Miotic or cholinergic agents. These increase the outflow of fluid from your eye. An example is pilocarpine (Isopto Carpine). Side effects include headache, eye pain, smaller pupils, possible blurred or dim vision, and nearsightedness. This class of medicine is usually prescribed to be used up to four times a day. Because of potential side effects and the need for frequent daily use, these medicines are not prescribed very often anymore.

Combination drugs:

  • Timolol/Brinzolamide (Azarga-not available in the US)
  • Timolol/Dorzolamide (Cosopt)
  • Timolol/Latanoprost (Xalacom-not available in the US)
  • Timolol/Bimatoprost (Ganfort-not available in the US)
  • Timolol/Brimonidine (Combigan)
  • Brinzolamide/Brimonidine (Simbrinza)

Because some of the eye drop medicine is absorbed into the bloodstream, you may experience some systemic side effects unrelated to your eyes. To minimize this absorption, close your eyes for 1 to 2 minutes after putting the drops in. You also may press lightly at the corner of your eyes near your nose to close the tear duct for 1 to 2 minutes. Wipe off any unused drops from your eyelid.

You may be prescribed multiple eye drops or need to use artificial tears. Make sure you wait at least five minutes in between using different drops.

Never change or stop taking your glaucoma medications without talking to your ophthalmologist. If you are about to run out of your medication, ask your ophthalmologist if you should have your prescription refilled.

Alpha agonists for glaucoma

Alpha agonists also called alpha2-adrenergic agonists work by reducing the amount of fluid your eye produces. Alpha agonists also increase the amount of fluid that drains out of the eyes. This helps lower eye pressure and, hopefully, saves your vision.

Possible side effects of alpha agonists include:

  • red, stinging or painful eyes after using drops
  • blurry vision
  • allergy (redness, itching, tearing and swelling of the eye)
  • a large (dilated) pupil
  • headaches
  • dry mouth
  • feeling tired, weak or dizzy
  • decreased blood pressure
  • a fast or irregular heartbeat
  • feeling nervous

Blurry vision, stinging, and redness may improve with time. But if the side effects still bother you, call your ophthalmologist. They may be able to lower your dose or change your medicine. Most side effects go away when the medicine is stopped. If you feel you need to stop taking this medication due to side effects or other concerns, please discuss with your doctor.

Beta-blockers for glaucoma

Beta-blockers also called beta-adrenergic antagonists work by reducing the amount of fluid your eye produces. This helps lower pressure in your eye and, hopefully, saves your vision.

Possible side effects of beta-blockers include:

  • red, stinging or painful eyes after using drops.
  • blurry vision
  • breathing problems in people with asthma, emphysema, or chronic obstructive pulmonary disease (COPD)
  • a slow or irregular heartbeat
  • feeling tired
  • depression
  • dizziness
  • a change in sex drive or sexual function
  • getting overly tired during exercise
  • in people with diabetes, low blood sugar symptoms becoming difficult to notice

Blurry vision, stinging, and redness may improve with time. But if the side effects still bother you, call your ophthalmologist. They may be able to lower your dose or change your medicine. Most side effects go away when the medication is stopped. Never suddenly quit taking your medicine unless your doctor tells you to.

Carbonic anhydrase inhibitors for glaucoma

Carbonic anhydrase inhibitors work by reducing the amount of fluid your eye produces. This helps lower eye pressure and, hopefully, saves your vision. Your ophthalmologist may have you take this medicine as an eye drop or by mouth as a pill.

Possible side effects of carbonic anhydrase inhibitors include:

  • stinging eyes after putting drops in.
  • red eyes
  • blurry vision
  • a skin rash (especially in people who are allergic to sulfa drugs)
  • changes in how things taste to you (especially with carbonated drinks)
  • bad taste or upset stomach (nausea)
  • feeling tired
  • decreased energy
  • increase in urination (with the pills)
  • tingling around the mouth and fingertips (with the pills)
  • lower potassium (with the pills)

Blurry vision, stinging, and redness may improve with time. But if the side effects still bother you, call your ophthalmologist. They may be able to lower your dose or change your medicine. Most side effects go away when the medication is stopped. Never suddenly quit taking your medicine unless your doctor tells you to.

Miotics for glaucoma

Miotics also called cholinergic agents make your pupil constrict (get smaller or miosis), increasing the amount of fluid that drains out of the eye. This helps lower eye pressure and, hopefully, helps protect your vision.

Possible side effects of miotics include:

  • blurred vision
  • nearsightedness (trouble focusing on distant objects)
  • dim vision with difficulty seeing in the dark or at night
  • headache or brow ache (aching around eye)

While very rare, there is the possibility that your retina could detach. This is when the light-sensitive tissue lining the back of the eye pulls away. You would suddenly notice dark specks or spots (floaters) or flashing lights in your vision. If you have these symptoms, see your ophthalmologist immediately.

Side effects may go away after you take the medicine for a while. But if the side effects still bother you, call your ophthalmologist. They may be able to lower your dose or change your medicine. If you feel you need to stop taking this medication due to side effects or other concerns, please discuss with your doctor.

Prostaglandin analogs for glaucoma

Prostaglandin analogs also called prostaglandin F2a analogues work by increasing the drainage of fluid out of your eye. This helps lower eye pressure and, hopefully, saves your vision.

Possible side effects of prostaglandin analogs include:

  • red, stinging or painful eyes after using drops.
  • feeling like something is in your eye
  • blurry vision
  • a permanent change in your eye color (occurs mostly in hazel eyes)
  • an increase in thickness, number and length of eyelashes
  • darkening of the eyelid
  • worsening of existing angina and asthma
  • joint aches
  • light sensitivity
  • eyes gradually sinking deeper into their sockets, keeping eyelids from working properly

Blurry vision, stinging, and redness may improve with time. But if the side effects still bother you, call your ophthalmologist. They may be able to lower your dose or change your medicine. Most side effects go away when the medication is stopped. Never suddenly quit taking your medicine unless your doctor tells you to.

Oral medicines

Eye drops alone may not bring eye pressure down to the desired level. So an eye doctor also may prescribe oral medicine. This medicine is usually a carbonic anhydrase inhibitor. Possible side effects include frequent urination, tingling in the fingers and toes, depression, stomach upset, and kidney stones.

Surgery, laser and other therapies

Other treatment options include laser therapy and surgery. The following techniques may help to drain fluid within the eye and lower eye pressure:

  • Laser trabeculoplasty is an option if eye drops can’t be tolerated. Laser trabeculoplasty also may be used if medicine hasn’t slowed the progression of the glaucoma. An eye doctor also may recommend laser surgery before using eye drops. It’s done in the eye doctor’s office. An eye doctor uses a small laser to improve the drainage of the tissue located at the angle where the iris and cornea meet. It may take a few weeks before the full effect of this procedure becomes apparent.
  • Laser iridotomy. Laser iridotomy is for people who have angle-closure glaucoma. The ophthalmologist uses a laser to create a tiny hole in the iris. This hole helps fluid flow to the drainage angle.
  • Glaucoma filtration surgery also called a trabeculectomy. The eye doctor (ophthalmologist) creates an opening in the white of your eye, which also is known as the sclera. This is where your eye surgeon creates a tiny flap in the sclera. They will also create a bubble like a pocket in the conjunctiva called a filtration bleb. It is usually hidden under the upper eyelid and cannot be seen. Aqueous humor will be able to drain out of the eye through the flap and into the bleb, lowering eye pressure. In the bleb, the fluid is absorbed by tissue around your eye.
  • Drainage tubes. In this procedure, the eye surgeon inserts a small drainage tube in your eye to drain excess fluid to lower eye pressure. The glaucoma drainage implant sends the fluid to a collection area called a reservoir. Your eye surgeon creates this reservoir beneath the conjunctiva. The fluid is then absorbed into nearby blood vessels.
  • Minimally invasive glaucoma surgery (MIGS). An eye doctor may suggest a minimally invasive glaucoma surgery (MIGS) procedure to lower eye pressure. This procedure generally require less immediate postoperative care and have less risk than trabeculectomy or using a drainage device. A minimally invasive glaucoma surgery (MIGS) procedure is often combined with cataract surgery. There are a number of minimally invasive glaucoma surgery (MIGS) techniques available.
  • Cataract surgery. For some people with narrow angles, removing the eye’s natural lens can lower eye pressure. With narrow angles, the iris and the cornea are too close together. This can cover (block) the eye’s drainage channel. Removing the eye’s lens with cataract surgery creates more space for fluid to leave the eye. This can lower eye pressure.

After your procedure, you’ll need to see your eye doctor for follow-up exams. You can expect to visit your ophthalmologist about every 3 to 6 months. However, this can vary depending on your treatment needs. And you may eventually need to undergo additional procedures if your eye pressure begins to rise or other changes happen in your eye.

Living with glaucoma

If you have glaucoma, the best thing you can do is follow your eye specialist’s guidance on treating and managing your glaucoma. Your eye specialist may also recommend making certain changes to your life, habits or routine. These can include:

  • Not ignoring new symptoms or vision changes
  • Reaching and maintaining a weight that’s healthy for you
  • Staying physically active (but be sure to ask about which activities to avoid, as some can increase eye pressure)
  • Seeing your eye specialist as recommended

You should also see your eye specialist if you notice new symptoms, if treatments aren’t as effective or if you have treatment side effects that are disrupting your life.

Glaucoma prognosis

Without treatment, glaucoma inevitably causes permanent vision loss and blindness. With treatment, it’s possible to slow the progress of your glaucoma or stop it entirely. The higher the intraocular pressure (IOP) and the duration of elevated IOP, the greater the risk of damaging the optic nerve. But because the range of possibilities can vary so widely between indivuals, your eye specialist is the best person to talk to about this. Your eye doctor can tell you the likely prognosis (outlook) for your specific case and what you can do to help tilt the scales in your favor. Timely diagnosis plays a critical role in mitigating glaucomatous eye damage, and early intervention is crucial in preventing and slowing down vision loss progression. Effective treatment, especially in maintaining low intraocular pressure (IOP) levels, can often lead to positive outcomes, preserving visual field integrity and halting disease advancement 28.

  1. Gosling D, Meyer JJ. Normal Tension Glaucoma. [Updated 2022 Dec 12]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK576377[][][][][][][][][][][][][][][][][]
  2. Shields MB. Normal-tension glaucoma: is it different from primary open-angle glaucoma? Curr Opin Ophthalmol. 2008 Mar;19(2):85-8. doi: 10.1097/ICU.0b013e3282f3919b[][][][]
  3. Lee BL, Bathija R, Weinreb RN. The definition of normal-tension glaucoma. J Glaucoma. 1998 Dec;7(6):366-71.[][][]
  4. Allison K, Patel D, Alabi O. Epidemiology of Glaucoma: The Past, Present, and Predictions for the Future. Cureus. 2020 Nov 24;12(11):e11686. doi: 10.7759/cureus.11686[][]
  5. Quigley HA, Broman AT. The number of people with glaucoma worldwide in 2010 and 2020. Br J Ophthalmol. 2006;90(3):262–267. doi: 10.1136/bjo.2005.081224[]
  6. Quigley HA, Broman AT. The number of people with glaucoma worldwide in 2010 and 2020. Br J Ophthalmol. 2006;90:262–7. doi: 10.1136/bjo.2005.081224[]
  7. Thylefors B, Négrel AD. The global impact of glaucoma. Bull World Health Organ. 1994;72(3):323-6. https://pmc.ncbi.nlm.nih.gov/articles/instance/2486713/pdf/bullwho00414-0005.pdf[]
  8. What is glaucoma? https://wga.one/what-is-glaucoma[][]
  9. Shahror, R.A., Morris, C.A., Mohammed, A.A. et al. Role of myeloid cells in ischemic retinopathies: recent advances and unanswered questions. J Neuroinflammation 21, 65 (2024). https://doi.org/10.1186/s12974-024-03058-y[]
  10. Goel M, Picciani RG, Lee RK, Bhattacharya SK. Aqueous humor dynamics: a review. Open Ophthalmol J. Sep 3 2010;4:52-9. doi:10.2174/1874364101004010052[][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][]
  11. Toris CB, Koepsell SA, Yablonski ME, Camras CB. Aqueous humor dynamics in ocular hypertensive patients. J Glaucoma. 2002 Jun;11(3):253-8. doi: 10.1097/00061198-200206000-00015[][][][][]
  12. Macknight AD, McLaughlin CW, Peart D, Purves RD, Carré DA, Civan MM. Formation of the aqueous humor. Clin Exp Pharmacol Physiol. 2000 Jan-Feb;27(1-2):100-6. doi: 10.1046/j.1440-1681.2000.03208.x[]
  13. Ferreira SM, Lerner SF, Brunzini R, Evelson PA, Llesuy SF. Oxidative stress markers in aqueous humor of glaucoma patients. Am J Ophthalmol. 2004 Jan;137(1):62-9. doi: 10.1016/s0002-9394(03)00788-8[][]
  14. Kaeslin MA, Killer HE, Fuhrer CA, Zeleny N, Huber AR, Neutzner A. Changes to the Aqueous Humor Proteome during Glaucoma. PLoS One. 2016 Oct 27;11(10):e0165314. doi: 10.1371/journal.pone.0165314[][]
  15. Brubaker RF. The flow of aqueous humor in the human eye. Trans Am Ophthalmol Soc. 1982;80:391-474. https://pmc.ncbi.nlm.nih.gov/articles/instance/1312276/pdf/taos00019-0414.pdf[][][][][]
  16. R TCYMT. Aqueous humor dynamics. In: DC CNL, ed. Atlas of Glaucoma. Second edition ed. Informa HK; 2007:13 – 28:chap 3.[][][][][][][][][][][][][][][][][][][][][][]
  17. Kiel JW, Hollingsworth M, Rao R, Chen M, Reitsamer HA. Ciliary blood flow and aqueous humor production. Prog Retin Eye Res. Jan 2011;30(1):1-17. doi:10.1016/j.preteyeres.2010.08.001[][][][][][][][][][][][][]
  18. Macknight AD, McLaughlin CW, Peart D, Purves RD, Carre DA, Civan MM. Formation of the aqueous humor. Clin Exp Pharmacol Physiol. Jan-Feb 2000;27(1-2):100-6. doi:10.1046/j.1440-1681.2000.03208.x[]
  19. Johnson M, McLaren JW, Overby DR. Unconventional aqueous humor outflow: A review. Exp Eye Res. May 2017;158:94-111. doi:10.1016/j.exer.2016.01.017[][][][][][][][][][][][][][]
  20. Roy Chowdhury U, Hann CR, Stamer WD, Fautsch MP. Aqueous humor outflow: dynamics and disease. Invest Ophthalmol Vis Sci. May 2015;56(5):2993-3003. doi:10.1167/iovs.15-16744[][][][]
  21. Fautsch MP, Johnson DH. Aqueous humor outflow: what do we know? Where will it lead us? Invest Ophthalmol Vis Sci. Oct 2006;47(10):4181-7. doi:10.1167/iovs.06-0830[][][]
  22. Trabecular Meshwork Research. https://ethier.gatech.edu/anterior-segmenttrabecular-meshwork-biomechanics[][]
  23. J. Buffault, A. Labbé, P. Hamard, F. Brignole-Baudouin, C. Baudouin. The trabecular meshwork: Structure, function and clinical implications. A review of the literature. Journal Français
    d’Ophtalmologie, 2020, 43 (7), pp.e217-e230. https://hal.sorbonne-universite.fr/hal-02962228v1/document[]
  24. Medical Management of Glaucoma in the 21st Century from a Canadian Perspective. Harasymowycz P, Birt C, Gooi P, et al. J Ophthalmol. 2016;2016:6509809. doi: 10.1155/2016/6509809[]
  25. Glaucoma. https://www.nei.nih.gov/learn-about-eye-health/eye-conditions-and-diseases/glaucoma[][]
  26. Congenital Glaucoma. https://glaucoma.org/types/congenital-glaucoma[][]
  27. Glaucoma in Infants. https://www.rch.org.au/ophthal/patient_information/Glaucoma/[][]
  28. Dietze J, Blair K, Zeppieri M, et al. Glaucoma. [Updated 2024 Mar 16]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK538217[][][]
  29. Kelly SR, Khawaja AP, Bryan SR, Azuara-Blanco A, Sparrow JM, Crabb DP. Progression from ocular hypertension to visual field loss in the English hospital eye service. Br J Ophthalmol. 2020 Oct;104(10):1406-1411. doi: 10.1136/bjophthalmol-2019-315052[]
  30. Open-angle glaucoma: Causes, symptoms and treatment. https://www.allaboutvision.com/conditions/glaucoma/primary-open-angle[][]
  31. Salvetat ML, Pellegrini F, Spadea L, Salati C, Zeppieri M. Pharmaceutical Approaches to Normal Tension Glaucoma. Pharmaceuticals (Basel). 2023 Aug 17;16(8):1172. doi: 10.3390/ph16081172[][][][][][][]
  32. Van Buskirk EM. The tale of normal-tension glaucoma. J Glaucoma. 1998 Dec;7(6):363-5.[][]
  33. Sowka J. New thoughts on normal tension glaucoma. Optometry. 2005 Oct;76(10):600-8. doi: 10.1016/j.optm.2005.08.020[][]
  34. European Glaucoma Society Terminology and Guidelines for Glaucoma, 5th Edition. British Journal of Ophthalmology 2021;105:1-169. doi: 10.1136/bjophthalmol-2021-egsguidelines[]
  35. Gedde SJ, Vinod K, Wright MM, Muir KW, Lind JT, Chen PP, Li T, Mansberger SL; American Academy of Ophthalmology Preferred Practice Pattern Glaucoma Panel. Primary Open-Angle Glaucoma Preferred Practice Pattern®. Ophthalmology. 2021 Jan;128(1):P71-P150. doi: 10.1016/j.ophtha.2020.10.022[][][][]
  36. Wu AM, Wu CM, Young BK, Wu DJ, Chen A, Margo CE, Greenberg PB. Evaluation of primary open-angle glaucoma clinical practice guidelines. Can J Ophthalmol. 2015 Jun;50(3):192-6. doi: 10.1016/j.jcjo.2015.03.005[]
  37. Asia-Pacific Glaucoma Society (APGS) Amsterdam, The Netherlands: Kugler Publications, 3th Edition. 2016. http://medi-guide.meditool.cn/ymtpdf/80C3C8C0-BED9-6460-F2FA-8C56BBF88EDF.pdf[]
  38. Kiuchi Y, Inoue T, Shoji N, Nakamura M, Tanito M; Glaucoma Guideline Preparation Committee, Japan Glaucoma Society. The Japan Glaucoma Society guidelines for glaucoma 5th edition. Jpn J Ophthalmol. 2023 Mar;67(2):189-254. doi: 10.1007/s10384-022-00970-9[]
  39. Colton T, Ederer F. The distribution of intraocular pressures in the general population. Surv Ophthalmol. 1980 Nov-Dec;25(3):123-9. doi: 10.1016/0039-6257(80)90086-7[]
  40. Drance SM, Morgan RW, Sweeney VP. Shock-induced optic neuropathy: a cause of nonprogressive glaucoma. N Engl J Med. 1973 Feb 22;288(8):392-5. doi: 10.1056/NEJM197302222880804[][][]
  41. Normal Tension Glaucoma. https://eyewiki.org/Normal_Tension_Glaucoma[][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][][]
  42. Tomita G. The optic nerve head in normal-tension glaucoma. Curr Opin Ophthalmol. 2000 Apr;11(2):116-20. doi: 10.1097/00055735-200004000-00009[]
  43. Drance SM. Disc hemorrhages in the glaucomas. Surv Ophthalmol. 1989 Mar-Apr;33(5):331-7. doi: 10.1016/0039-6257(89)90010-6[][][]
  44. Uhler TA, Piltz-Seymour J. Optic disc hemorrhages in glaucoma and ocular hypertension: implications and recommendations. Curr Opin Ophthalmol. 2008 Mar;19(2):89-94. doi: 10.1097/ICU.0b013e3282f3e6bc[][]
  45. Budenz DL, Anderson DR, Feuer WJ, Beiser JA, Schiffman J, Parrish RK 2nd, Piltz-Seymour JR, Gordon MO, Kass MA; Ocular Hypertension Treatment Study Group. Detection and prognostic significance of optic disc hemorrhages during the Ocular Hypertension Treatment Study. Ophthalmology. 2006 Dec;113(12):2137-43. doi: 10.1016/j.ophtha.2006.06.022[][]
  46. Schacknow PN, Samples JR eds. The glaucoma book: a practical, evidence-based approach to patient care. New York: Springer; 2010.[][]
  47. Optic Disc Hemorrhage. https://eyewiki.org/Optic_Disc_Hemorrhage[]
  48. Kosior-Jarecka E, Wróbel-Dudzińska D, Łukasik U, Żarnowski T. Ocular and Systemic Risk Factors of Different Morphologies of Scotoma in Patients with Normal-Tension Glaucoma. J Ophthalmol. 2017;2017:1480746. doi: 10.1155/2017/1480746[]
  49. Kottler MS, Drance SM. Studies of hemorrhage on the optic disc. Can J Ophthalmol. 1976 Apr;11(2):102-5.[]
  50. Anderson DR; Normal Tension Glaucoma Study. Collaborative normal tension glaucoma study. Curr Opin Ophthalmol. 2003 Apr;14(2):86-90. doi: 10.1097/00055735-200304000-00006[]
  51. Mullany S, Xiao L, Qassim A, Marshall H, Gharahkhani P, MacGregor S, Hassall MM, Siggs OM, Souzeau E, Craig JE. Normal-tension glaucoma is associated with cognitive impairment. Br J Ophthalmol. 2022 Jul;106(7):952-956. doi: 10.1136/bjophthalmol-2020-317461[][]
  52. Crump C, Sundquist J, Sieh W, Sundquist K. Risk of Alzheimer’s Disease and Related Dementias in Persons with Glaucoma: A National Cohort Study. Ophthalmology. 2024 Mar;131(3):302-309. doi: 10.1016/j.ophtha.2023.10.014[][]
  53. Klein B.E., Klein R., Sponsel W.E., Franke T., Cantor L.B., Martone J., Menage M.J. Prevalence of glaucoma. The Beaver Dam Eye Study. Ophthalmology. 1992;99:1499–1504. doi: 10.1016/S0161-6420(92)31774-9[]
  54. Leung DYL, Tham CC. Normal-tension glaucoma: Current concepts and approaches-A review. Clin Exp Ophthalmol. 2022 Mar;50(2):247-259. doi: 10.1111/ceo.14043[][]
  55. Zhao J., Solano M.M., Oldenburg C.E., Liu T., Wang Y., Wang N., Lin S.C. Prevalence of Normal-Tension Glaucoma in the Chinese Population: A Systematic Review and Meta-Analysis. Am. J. Ophthalmol. 2019;199:101–110. doi: 10.1016/j.ajo.2018.10.017[]
  56. Collaborative Normal-Tension Glaucoma Study Group The effectiveness of intraocular pressure reduction in the treatment of normal-tension glaucoma. Am. J. Ophthalmol. 1998;126:498–505. doi: 10.1016/S0002-9394(98)00272-4[][][]
  57. Heijl A., Leske M.C., Bengtsson B., Hyman L., Bengtsson B., Hussein M., Early Manifest Glaucoma Trial Group Reduction of intraocular pressure and glaucoma progression: Results from the Early Manifest Glaucoma Trial. Arch. Ophthalmol. 2002;120:1268–1279. doi: 10.1001/archopht.120.10.1268[][]
  58. Killer H.E., Pircher A. Normal tension glaucoma: Review of current understanding and mechanisms of the pathogenesis. Eye Lond. 2018;32:924–930. doi: 10.1038/s41433-018-0042-2[]
  59. Razeghinejad M.R., Lee D. Managing normal tension glaucoma by lowering the intraocular pressure. Surv. Ophthalmol. 2019;64:111–116. doi: 10.1016/j.survophthal.2018.07.003[][][][][][]
  60. Lusthaus J., Goldberg I. Current management of glaucoma. Med. J. Aust. 2019;210:180–187. doi: 10.5694/mja2.50020[]
  61. Hoyng P.F., Kitazawa Y. Medical treatment of normal tension glaucoma. Surv. Ophthalmol. 2002;47, Suppl. S1:S116–S124. doi: 10.1016/S0039-6257(02)00322-3[][]
  62. Kim J.M., Sung K.R., Kim H.K., Park S.W., Lee E.J., Jeoung J.W., Park H.L., Ahn J., Yoo C., Kim C.Y. Long-Term Effectiveness and Safety of Tafluprost, Travoprost, and Latanoprost in Korean Patients with Primary Open-Angle Glaucoma or Normal-Tension Glaucoma: A Multicenter Retrospective Cohort Study (LOTUS Study) J. Clin. Med. 2021;10:2717. doi: 10.3390/jcm10122717[]
  63. Islam S, Spry C. Prostaglandin Analogues for Ophthalmic Use: A Review of Comparative Clinical Effectiveness, Cost-Effectiveness, and Guidelines [Internet]. Ottawa (ON): Canadian Agency for Drugs and Technologies in Health; 2020 Feb 18. Available from: https://www.ncbi.nlm.nih.gov/books/NBK562924[][]
  64. Quaranta L., Katsanos A., Russo A., Riva I. 24-hour intraocular pressure and ocular perfusion pressure in glaucoma. Surv. Ophthalmol. 2013;58:26–41. doi: 10.1016/j.survophthal.2012.05.003[]
  65. McKibbin M., Menage M.J. The effect of once-daily latanoprost on intraocular pressure and pulsatile ocular blood flow in normal tension glaucoma. Pt 1Eye Lond. 1999;13:31–34. doi: 10.1038/eye.1999.6[]
  66. Araie M., Mayama C. Use of calcium channel blockers for glaucoma. Prog. Retin. Eye Res. 2011;30:54–71. doi: 10.1016/j.preteyeres.2010.09.002[]
  67. Dinakaran S., Mehta P., Mehta R., Tilva B., Arora D., Tejwani S. Significance of non-intraocular pressure (IOP)-related factors particularly in normal tension glaucoma: Looking beyond IOP. Indian J. Ophthalmol. 2022;70:569–573. doi: 10.4103/ijo.IJO_861_21[][]
  68. Shin J.W., Jo Y.H., Song M.K., Won H.J., Kook M.S. Nocturnal blood pressure dip and parapapillary choroidal microvasculature dropout in normal-tension glaucoma. Sci. Rep. 2021;11:206. doi: 10.1038/s41598-020-80705-3[]
  69. Shen J., Wang Y., Yao K. Protection of retinal ganglion cells in glaucoma: Current status and future. Exp. Eye Res. 2021;205:108506. doi: 10.1016/j.exer.2021.108506[]
  70. Krupin T., Liebmann J.M., Greenfield D.S., Ritch R., Gardiner S., Low-Pressure Glaucoma Study Group A randomized trial of brimonidine versus timolol in preserving visual function: Results from the Low-Pressure Glaucoma Treatment Study. Am. J. Ophthalmol. 2011;151:671–681. doi: 10.1016/j.ajo.2010.09.026. Erratum in Am. J. Ophthalmol. 2011, 151, 1108[]
  71. Nocentini A., Supuran C.T. Adrenergic agonists and antagonists as antiglaucoma agents: A literature and patent review (2013–2019) Expert Opin. Ther. Pat. 2019;29:805–815. doi: 10.1080/13543776.2019.1665023[]
  72. Liu B., Liu Y.J. Carvedilol Promotes Retinal Ganglion Cell Survival Following Optic Nerve Injury via ASK1-p38 MAPK Pathway. CNS Neurol. Disord Drug Targets. 2019;18:695–704. doi: 10.2174/1871527318666191002095456[]
  73. Szumny D., Szeląg A. The influence of new beta-adrenolytics nebivolol and carvedilol on intraocular pressure and iris blood flow in rabbits. Graefes Arch. Clin. Exp. Ophthalmol. 2014;252:917–923. doi: 10.1007/s00417-014-2623-5[]
  74. Oh D.J., Chen J.L., Vajaranant T.S., Dikopf M.S. Brimonidine tartrate for the treatment of glaucoma. Expert Opin. Pharmacother. 2019;20:115–122. doi: 10.1080/14656566.2018.1544241[]
  75. Schulzer M., The Normal Tension Glaucoma Study Group Intraocular pressure reduction in normal-tension glaucoma patients. Ophthalmology. 1992;99:1468–1470. doi: 10.1016/S0161-6420(92)31782-8[]
  76. Optic disc hemorrhage in normal tension glaucoma. https://eyerounds.org/atlas/pages/Optic-Disc-Hemorrhage-in-NTG.htm#gsc.tab=0[]
  77. The effectiveness of intraocular pressure reduction in the treatment of normal-tension glaucoma. Collaborative Normal-Tension Glaucoma Study Group. Am J Ophthalmol. 1998 Oct;126(4):498-505. doi: 10.1016/s0002-9394(98)00272-4[][][][][][][][]
  78. Heijl A, Leske MC, Bengtsson B, et al. Reduction of Intraocular Pressure and Glaucoma Progression: Results From the Early Manifest Glaucoma Trial. Arch Ophthalmol. 2002;120(10):1268–1279. doi:10.1001/archopht.120.10.1268[][]
  79. Mozaffarieh M, Flammer J. New insights in the pathogenesis and treatment of normal tension glaucoma. Curr Opin Pharmacol. 2013 Feb;13(1):43-9. doi: 10.1016/j.coph.2012.10.001[]
  80. Crawford Downs J, Roberts MD, Sigal IA. Glaucomatous cupping of the lamina cribrosa: a review of the evidence for active progressive remodeling as a mechanism. Exp Eye Res. 2011 Aug;93(2):133-40. doi: 10.1016/j.exer.2010.08.004[]
  81. Wostyn P, Killer HE. Normal-Tension Glaucoma: A Glymphopathy? Eye Brain. 2023 Apr 6;15:37-44. doi: 10.2147/EB.S401306[]
  82. Pan Y, Iwata T. Molecular genetics of inherited normal tension glaucoma. Indian J Ophthalmol. 2024 May 1;72(Suppl 3):S335-S344. doi: 10.4103/IJO.IJO_3204_23[][][][][]
  83. Adeghate J, Rahmatnejad K, Waisbourd M, Katz LJ. Intraocular pressure-independent management of normal tension glaucoma. Surv Ophthalmol. 2019 Jan-Feb;64(1):101-110. doi: 10.1016/j.survophthal.2018.08.005[]
  84. Comparison of glaucomatous progression between untreated patients with normal-tension glaucoma and patients with therapeutically reduced intraocular pressures. Collaborative Normal-Tension Glaucoma Study Group. Am J Ophthalmol. 1998 Oct;126(4):487-97. doi: 10.1016/s0002-9394(98)00223-2. Erratum in: Am J Ophthalmol 1999 Jan;127(1):120.[][][][][][][][]
  85. Holländer R. Der Einfluss der Membranpermeabilität auf die TMPD-Oxydase-Aktivität bei Bakterien [The Influence of the Permeability of the Cell Membrane on TMPD Oxydase Activity (author’s transl)]. Zentralbl Bakteriol Orig A. 1977;237(2-3):351-7. German.[]
  86. Hirooka K, Tenkumo K, Fujiwara A, Baba T, Sato S, Shiraga F. Evaluation of peripapillary choroidal thickness in patients with normal-tension glaucoma. BMC Ophthalmol. 2012 Jul 28;12:29. doi: 10.1186/1471-2415-12-29[]
  87. Firat PG, Ozsoy E, Demirel S, Cumurcu T, Gunduz A. Evaluation of peripapillary retinal nerve fiber layer, macula and ganglion cell thickness in amblyopia using spectral optical coherence tomography. Int J Ophthalmol. 2013;6(1):90-4. doi: 10.3980/j.issn.2222-3959.2013.01.19[]
  88. Zheng Y, Cheung N, Aung T, Mitchell P, He M, Wong TY. Relationship of retinal vascular caliber with retinal nerve fiber layer thickness: the singapore malay eye study. Invest Ophthalmol Vis Sci. 2009 Sep;50(9):4091-6. doi: 10.1167/iovs.09-3444[]
  89. Kim JM, Sae Kim M, Ju Jang H, Ho Park K, Caprioli J. The association between retinal vessel diameter and retinal nerve fiber layer thickness in asymmetric normal tension glaucoma patients. Invest Ophthalmol Vis Sci. 2012 Aug 17;53(9):5609-14. doi: 10.1167/iovs.12-9783[]
  90. Lee JY, Yoo C, Park JH, Kim YY. Retinal vessel diameter in young patients with open-angle glaucoma: comparison between high-tension and normal-tension glaucoma. Acta Ophthalmol. 2012 Nov;90(7):e570-1. doi: 10.1111/j.1755-3768.2011.02371.x[]
  91. Fox AR, Fingert JH. Familial normal tension glaucoma genetics. Prog Retin Eye Res. 2023 Sep;96:101191. doi: 10.1016/j.preteyeres.2023.101191[]
  92. Burk RO, Rohrschneider K, Noack H, Völcker HE. Are large optic nerve heads susceptible to glaucomatous damage at normal intraocular pressure? A three-dimensional study by laser scanning tomography. Graefes Arch Clin Exp Ophthalmol. 1992;230(6):552-60. doi: 10.1007/BF00181778[]
  93. Tuulonen A, Takamoto T, Wu DC, Schwartz B. Optic disk cupping and pallor measurements of patients with a disk hemorrhage. Am J Ophthalmol. 1987 Apr 15;103(4):505-11. doi: 10.1016/s0002-9394(14)74272-2[]
  94. Vogel W, Dietze O, Judmaier G, Then P, Schmid T, Margreiter R. Delayed clearance of HBsAG after transplantation for fulminant delta-hepatitis. Lancet. 1988 Jan 2-9;1(8575-6):52. doi: 10.1016/s0140-6736(88)91025-2[]
  95. Buus DR, Anderson DR. Peripapillary crescents and halos in normal-tension glaucoma and ocular hypertension. Ophthalmology. 1989 Jan;96(1):16-9. doi: 10.1016/s0161-6420(89)32930-7[]
  96. Yamazaki Y, Hayamizu F, Miyamoto S, Nakagami T, Tanaka C, Inui S. Optic disc findings in normal tension glaucoma. Jpn J Ophthalmol. 1997 Jul-Aug;41(4):260-7. doi: 10.1016/s0021-5155(97)00052-x[]
  97. Gramer E, Leydhecker W. Glaukom ohne Hochdruck. Eine klinische Studie [Glaucoma without ocular hypertension. A clinical study]. Klin Monbl Augenheilkd. 1985 Apr;186(4):262-7. German. doi: 10.1055/s-2008-1050918[][]
  98. Krupin T, Liebmann JM, Greenfield DS, Ritch R, Gardiner S; Low-Pressure Glaucoma Study Group. A randomized trial of brimonidine versus timolol in preserving visual function: results from the Low-Pressure Glaucoma Treatment Study. Am J Ophthalmol. 2011 Apr;151(4):671-81. doi: 10.1016/j.ajo.2010.09.026 Erratum in: Am J Ophthalmol. 2011 Jun;151(6):1108.[]
  99. Chuang LH, Koh YY, Chen HSL, Lo YL, Yu CC, Yeung L, Lai CC. Normal tension glaucoma in obstructive sleep apnea syndrome: A structural and functional study. Medicine (Baltimore). 2020 Mar;99(13):e19468. doi: 10.1097/MD.0000000000019468[]
  100. Lin PW, Friedman M, Lin HC, Chang HW, Wilson M, Lin MC. Normal tension glaucoma in patients with obstructive sleep apnea/hypopnea syndrome. J Glaucoma. 2011 Dec;20(9):553-8. doi: 10.1097/IJG.0b013e3181f3eb81[]
  101. Bilgin G. Normal-tension glaucoma and obstructive sleep apnea syndrome: a prospective study. BMC Ophthalmol. 2014 Mar 10;14:27. doi: 10.1186/1471-2415-14-27[]
  102. Leggewie B, Gouveris H, Bahr K. A Narrative Review of the Association between Obstructive Sleep Apnea and Glaucoma in Adults. Int J Mol Sci. 2022 Sep 3;23(17):10080. doi: 10.3390/ijms231710080[]
  103. Phelps CD, Corbett JJ. Migraine and low-tension glaucoma. A case-control study. Invest Ophthalmol Vis Sci. 1985 Aug;26(8):1105-8. https://iovs.arvojournals.org/article.aspx?articleid=2159734[]
  104. Flammer J, Konieczka K, Flammer AJ. The primary vascular dysregulation syndrome: implications for eye diseases. EPMA J. 2013 Jun 7;4(1):14. doi: 10.1186/1878-5085-4-14[]
  105. Hirooka K, Higashide T, Sakaguchi K, Udagawa S, Sugiyama K, Oki K, Kometani M, Yoneda T, Fukunaga K, Akita T, Baba T, Kiuchi Y. Prevalence of Normal-Tension Glaucoma in Patients With Primary Aldosteronism. Am J Ophthalmol. 2025 Jan;269:339-345. doi: 10.1016/j.ajo.2024.09.014[][][]
  106. Ho K, Bodi NE, Sharma TP. Normal-Tension Glaucoma and Potential Clinical Links to Alzheimer’s Disease. J Clin Med. 2024 Mar 27;13(7):1948. doi: 10.3390/jcm13071948[][][]
  107. Fan N, Tan J, Liu X. Is “normal tension glaucoma” glaucoma? Med Hypotheses. 2019 Dec;133:109405. doi: 10.1016/j.mehy.2019.109405[]
  108. Killer HE, Pircher A. Normal tension glaucoma: review of current understanding and mechanisms of the pathogenesis. Eye (Lond). 2018 May;32(5):924-930. doi: 10.1038/s41433-018-0042-2[][]
  109. Chen YY, Lai YJ, Yen YF, Shen YC, Wang CY, Liang CY, Lin KH, Fan LW. Association between normal tension glaucoma and the risk of Alzheimer’s disease: a nationwide population-based cohort study in Taiwan. BMJ Open. 2018 Nov 5;8(11):e022987. doi: 10.1136/bmjopen-2018-022987[]
  110. Drance S, Anderson DR, Schulzer M; Collaborative Normal-Tension Glaucoma Study Group. Risk factors for progression of visual field abnormalities in normal-tension glaucoma. Am J Ophthalmol. 2001 Jun;131(6):699-708. doi: 10.1016/s0002-9394(01)00964-3[][]
  111. Konieczka K, Ritch R, Traverso CE, Kim DM, Kook MS, Gallino A, Golubnitschaja O, Erb C, Reitsamer HA, Kida T, Kurysheva N, Yao K. Flammer syndrome. EPMA J. 2014 Jul 8;5(1):11. doi: 10.1186/1878-5085-5-11[]
  112. Xie Y, Jiang J, Liu C, Lin H, Wang L, Zhang C, Chen J, Liang Y, Congdon N, Zhang S. Performance of a Glaucoma Screening Program Compared With Opportunistic Detection in China. J Glaucoma. 2023 Feb 1;32(2):80-84. doi: 10.1097/IJG.0000000000002125[][][][]
  113. Normal Tension Glaucoma. https://glaucoma.org.au/what-is-glaucoma/types-of-glaucoma/normal-tension-glaucoma[][]
  114. Quigley HA. Glaucoma. Lancet. 2011 Apr 16;377(9774):1367-77. doi: 10.1016/S0140-6736(10)61423-7[]
  115. Lawlor M, Quartilho A, Bunce C, Nathwani N, Dowse E, Kamal D, Gazzard G. Patients With Normal Tension Glaucoma Have Relative Sparing of the Relative Afferent Pupillary Defect Compared to Those With Open Angle Glaucoma and Elevated Intraocular Pressure. Invest Ophthalmol Vis Sci. 2017 Oct 1;58(12):5237-5241. doi: 10.1167/iovs.17-21688[]
  116. Spaeth GL, Henderer J, Liu C, Kesen M, Altangerel U, Bayer A, Katz LJ, Myers J, Rhee D, Steinmann W. The disc damage likelihood scale: reproducibility of a new method of estimating the amount of optic nerve damage caused by glaucoma. Trans Am Ophthalmol Soc. 2002;100:181-5; discussion 185-6[]
  117. Mahmoudinezhad G, Moghimi S, Proudfoot JA, Brye N, Nishida T, Yarmohammadi A, Kamalipour A, Zangwill LM, Weinreb RN. Effect of Testing Frequency on the Time to Detect Glaucoma Progression With Optical Coherence Tomography (OCT) and OCT Angiography. Am J Ophthalmol. 2023 Jan;245:184-192. doi: 10.1016/j.ajo.2022.08.030[][][][]
  118. What Is Fluorescein Angiography? https://www.aao.org/eye-health/treatments/what-is-fluorescein-angiography[][][][]
  119. Seo SB, Cho HK. Deep learning classification of early normal-tension glaucoma and glaucoma suspects using Bruch’s membrane opening-minimum rim width and RNFL. Sci Rep. 2020 Nov 4;10(1):19042. doi: 10.1038/s41598-020-76154-7[][]
  120. Betzler BK, Siat DJY, Agrawal R, Dorairaj S, Ang BCH. Comparison of Peripapillary Choroidal Thickness Between Primary Open-angle Glaucoma, Normal Tension Glaucoma, and Normal Eyes: A Systematic Review and Meta-analysis. Ophthalmol Glaucoma. 2024 Jul-Aug;7(4):359-371. doi: 10.1016/j.ogla.2024.02.008[][][][][]
  121. Shih CY, Graff Zivin JS, Trokel SL, Tsai JC. Clinical significance of central corneal thickness in the management of glaucoma. Arch Ophthalmol. 2004 Sep;122(9):1270-5. doi: 10.1001/archopht.122.9.1270[][]
  122. Shetgar AC, Mulimani MB. The central corneal thickness in normal tension glaucoma, primary open angle glaucoma and ocular hypertension. J Clin Diagn Res. 2013 Jun;7(6):1063-7. doi: 10.7860/JCDR/2013/4292.3022[][]
  123. Krupin T, Liebmann JM, Greenfield DS, Ritch R, Gardiner S; Low-Pressure Glaucoma Study Group. A randomized trial of brimonidine versus timolol in preserving visual function: results from the Low-Pressure Glaucoma Treatment Study. Am J Ophthalmol. 2011 Apr;151(4):671-81. doi: 10.1016/j.ajo.2010.09.026. Erratum in: Am J Ophthalmol. 2011 Jun;151(6):1108.[][][][]
  124. Tanna AP, Johnson M. Rho Kinase Inhibitors as a Novel Treatment for Glaucoma and Ocular Hypertension. Ophthalmology. 2018 Nov;125(11):1741-1756. doi: 10.1016/j.ophtha.2018.04.040[]
  125. Thomas NM, Nagrale P. Rho Kinase Inhibitors as a Neuroprotective Pharmacological Intervention for the Treatment of Glaucoma. Cureus. 2022 Aug 26;14(8):e28445. doi: 10.7759/cureus.28445[]
  126. Chihara E, Dimitrova G, Chihara T. Increase in the OCT angiographic peripapillary vessel density by ROCK inhibitor ripasudil instillation: a comparison with brimonidine. Graefes Arch Clin Exp Ophthalmol. 2018 Jul;256(7):1257-1264. doi: 10.1007/s00417-018-3945-5[]
  127. Kaneko Y, Ohta M, Inoue T, Mizuno K, Isobe T, Tanabe S, Tanihara H. Effects of K-115 (Ripasudil), a novel ROCK inhibitor, on trabecular meshwork and Schlemm’s canal endothelial cells. Sci Rep. 2016 Jan 19;6:19640. doi: 10.1038/srep19640[]
  128. El Mallah MK, Walsh MM, Stinnett SS, Asrani SG. Selective laser trabeculoplasty reduces mean IOP and IOP variation in normal tension glaucoma patients. Clin Ophthalmol. 2010 Aug 9;4:889-93. doi: 10.2147/opth.s11787[]
  129. Yang SA, Mitchell WG, Hall N, Elze T, Miller JW, Lorch AC, Zebardast N. Usage Patterns of Minimally Invasive Glaucoma Surgery (MIGS) Differ by Glaucoma Type: IRIS Registry Analysis 2013-2018. Ophthalmic Epidemiol. 2022 Aug;29(4):443-451. doi: 10.1080/09286586.2021.1955391[][]
  130. Schargus M, Theilig T, Rehak M, Busch C, Bormann C, Unterlauft JD. Outcome of a single XEN microstent implant for glaucoma patients with different types of glaucoma. BMC Ophthalmol. 2020 Dec 17;20(1):490. doi: 10.1186/s12886-020-01764-8[]
  131. Oo HH, Hong ASY, Lim SY, Ang BCH. Angle-based minimally invasive glaucoma surgery in normal tension glaucoma: A systematic review and meta-analysis. Clin Exp Ophthalmol. 2024 Sep-Oct;52(7):740-760. doi: 10.1111/ceo.14408[][][][][]
  132. Baek S.U., Ha A., Kim D.W., Jeoung J.W., Park K.H., Kim Y.K. Risk factors for disease progression in low-teens normal-tension glaucoma. Br. J. Ophthalmol. 2020;104:81–86. doi: 10.1136/bjophthalmol-2018-313375[][]
  133. Sawada A., Rivera J.A., Takagi D., Nishida T., Yamamoto T. Progression to Legal Blindness in Patients with Normal Tension Glaucoma: Hospital-Based Study. Investig. Ophthalmol. Vis. Sci. 2015;56:3635–3641. doi: 10.1167/iovs.14-16093[][][][]
  134. Anderson D.R., Drance S.M., Schulzer M., Collaborative Normal-Tension Glaucoma Study Group Factors that predict the benefit of lowering intraocular pressure in normal tension glaucoma. Am. J. Ophthalmol. 2003;136:820–829. doi: 10.1016/S0002-9394(03)00478-1[][][]
  135. Bach-Holm D., Jensen P.K., Kessing S.V. Long-term rate of progression and target intraocular pressure in patients with normal-tension glaucoma in clinical care. Acta Ophthalmol. 2018;96:e1034–e1035. doi: 10.1111/aos.13767[][][]
  136. Sakata R., Yoshitomi T., Iwase A., Matsumoto C., Higashide T., Shirakashi M., Aihara M., Sugiyama K., Araie M., Lower Normal Pressure Glaucoma Study Members in Japan Glaucoma Society Factors Associated with Progression of Japanese Open-Angle Glaucoma with Lower Normal Intraocular Pressure. Ophthalmology. 2019;126:1107–1116. doi: 10.1016/j.ophtha.2018.12.029[][]
  137. Daugeliene L., Yamamoto T., Kitazawa Y. Effect of trabeculectomy on visual field in progressive normal-tension glaucoma. Jpn. J. Ophthalmol. 1998;42:286–292. doi: 10.1016/S0021-5155(98)00013-6[]
  138. Gurnani B, Kaur K. Pigment Dispersion Syndrome. [Updated 2023 Jun 11]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK580526[][][]
  139. Pigmentary Glaucoma and Pigment Dispersion Syndrome. https://eyewiki.org/Pigmentary_Glaucoma_and_Pigment_Dispersion_Syndrome[][]
  140. What Is Pigment Dispersion Syndrome? https://www.aao.org/eye-health/diseases/what-is-pigment-dispersion-syndrome[]
  141. Pigmentary Glaucoma. https://glaucoma.org/types/pigmentary-glaucoma[]
  142. Zeppieri M. Pigment dispersion syndrome: A brief overview. J Clin Transl Res. 2022 Sep 7;8(5):344-350. https://pmc.ncbi.nlm.nih.gov/articles/PMC9741933[][][]
  143. Scuderi G, Contestabile MT, Scuderi L, Librando A, Fenicia V, Rahimi S. Pigment Dispersion Syndrome and Pigmentary Glaucoma:A Review and Update. Int Ophthalmol. 2019;39:1651–62. doi: 10.1007/s10792-018-0938-7[][]
  144. Niyadurupola N, Broadway DC. Pigment dispersion syndrome and pigmentary glaucoma–a major review. Clin Exp Ophthalmol. 2008;36:868–82. doi: 10.1111/j.1442-9071.2009.01920.x[]
  145. Campbell DG. Pigmentary Dispersion and Glaucoma: A New Theory. Arch Ophthalmol. 1979;97(9):1667–1672. doi:10.1001/archopht.1979.01020020235011[][][][][][]
  146. Kampik A, Green WR, Quigley HA, Pierce LH. Scanning and transmission electron microscopic studies of two cases of pigment dispersion syndrome. Am J Ophthalmol. 1981 May;91(5):573-87. doi: 10.1016/0002-9394(81)90055-6[][][][][][]
  147. Mardin CY, Küchle M, Nguyen NX, Martus P, Naumann GO. Quantification of aqueous melanin granules, intraocular pressure and glaucomatous damage in primary pigment dispersion syndrome. Ophthalmology. 2000 Mar;107(3):435-40. doi: 10.1016/s0161-6420(99)00089-5[][][][]
  148. Campbell DG, Schertzer RM. Pathophysiology of Pigment Dispersion Syndrome and Pigmentary Glaucoma. Curr Opin Ophthalmol. 1995;6:96–101. doi: 10.1097/00055735-199504000-00015[][]
  149. Ritch R. A unification hypothesis of pigment dispersion syndrome. Trans Am Ophthalmol Soc. 1996;94:381-405; discussion 405-9. https://pmc.ncbi.nlm.nih.gov/articles/instance/1312105/pdf/taos00005-0402.pdf[][]
  150. Potash SD, Tello C, Liebmann J, Ritch R. Ultrasound Biomicroscopy in Pigment Dispersion Syndrome. Ophthalmology. 1994;101:332–9. doi: 10.1016/s0161-6420(94)31331-5[][]
  151. Karickhoff JR. Pigmentary dispersion syndrome and pigmentary glaucoma: a new mechanism concept, a new treatment, and a new technique. Ophthalmic Surg. 1992 Apr;23(4):269-77.[][][]
  152. Jewelewicz DA, Radcliffe NM, Liebmann J, Ritch R. Temporal evolution of intraocular pressure elevation after pupillary dilation in pigment dispersion syndrome. J Glaucoma. 2009 Mar;18(3):184-5. doi: 10.1097/IJG.0b013e318182edbf[][]
  153. Richter CU, Richardson TM, Grant WM. Pigmentary Dispersion Syndrome and Pigmentary Glaucoma: A Prospective Study of the Natural History. Arch Ophthalmol. 1986;104(2):211–215. doi:10.1001/archopht.1986.01050140065021[][]
  154. Richardson TM, Hutchinson BT, Grant WM. The Outflow Tract in Pigmentary Glaucoma: A Light and Electron Microscopic Study. Arch Ophthalmol. 1977;95(6):1015–1025. doi:10.1001/archopht.1977.04450060101010[][]
  155. Lichter PR. Pigmentary glaucoma–current concepts. Trans Am Acad Ophthalmol Otolaryngol. 1974 Mar-Apr;78(2):OP309-13.[][]
  156. Kupfer C, Kuwabara T, Kaiser-Kupfer M. The Histopathology of Pigmentary Dispersion Syndrome with Glaucoma. Am J Ophthalmol. 1975;80:857–62. doi: 10.1016/0002-9394(75)90283-4[][]
  157. Fine BS, Yanoff M, Scheie HG. Pigmentary “glaucoma”. A histologic study. Trans Am Acad Ophthalmol Otolaryngol. 1974 Mar-Apr;78(2):OP314-25.[][]
  158. Mauksch H. Über Idiopathischen Zerfall des Retinalen Pigmentblattes der Iris bei Zwei Brudern. Z Augenheilkd. 1925;57:262.[][]
  159. McDermott JA, Ritch R, Berger A, Wang RF. Inheritance of Pigmentary Dispersion Syndrome. Invest Ophthalmol Vis Sci. 1987;28:153.[][]
  160. Scheie HG, Cameron JD. Pigment Dispersion Syndrome:A Clinical Study. Br J Ophthalmol. 1981;65:264–9. doi: 10.1136/bjo.65.4.264[][]
  161. Andersen JS, Pralea AM, DelBono EA, Haines JL, Gorin MB, Schuman JS, et al. A Gene Responsible for the Pigment Dispersion Syndrome Maps to Chromosome 7q35-q36. Arch Ophthalmol. 1997;115:384–8. doi: 10.1001/archopht.1997.01100150386012[][]
  162. Lascaratos G, Shah A, Garway-Heath DF. The Genetics of Pigment Dispersion Syndrome and Pigmentary Glaucoma. Surv Ophthalmol. 2013;58:164–75. doi: 10.1016/j.survophthal.2012.08.002[][]
  163. Roberts DK, Meetz RE, Chaglasian MA. The inheritance of the pigment dispersion syndrome in blacks. J Glaucoma. 1999 Aug;8(4):250-6.[][]
  164. McDermott JA et al. Familial occurrence of pigmentary dispersion syndrome. Invest Ophthalmolol Vis Sci. 1987;28(suppl):136.[][]
  165. Siddiqui Y, Ten Hulzen RD, Cameron JD, Hodge DO, Johnson DH. What is the risk of developing pigmentary glaucoma from pigment dispersion syndrome? Am J Ophthalmol. 2003 Jun;135(6):794-9. doi: 10.1016/s0002-9394(02)02289-4. Erratum in: Am J Ophthalmol. 2003 Sep;136(3):592.[][][][][][][][][]
  166. Gaton D, Barak A, Segev S, Yassur Y, Treister G. [Prevalence of pigmentary dispersion syndrome in Israel]. Harefuah. 1998 Mar 1;134(5):337-9, 424. Hebrew.[][]
  167. STANKOVIC I. [A contribution to the knowledge of the heredity of pigment glaucoma]. Klin Monbl Augenheilkd Augenarztl Fortbild. 1961 Sep;139:165-74. German.[][]
  168. Andersen JS, Pralea AM, DelBono EA, Haines JL, Gorin MB, Schuman JS, Mattox CG, Wiggs JL. A gene responsible for the pigment dispersion syndrome maps to chromosome 7q35-q36. Arch Ophthalmol. 1997 Mar;115(3):384-8. doi: 10.1001/archopht.1997.01100150386012[][]
  169. Scheie HG, Cameron JD. Pigment dispersion syndrome: a clinical study. Br J Ophthalmol. 1981 Apr;65(4):264-9. doi: 10.1136/bjo.65.4.264[][][][]
  170. Sugar HS. Pigmentary glaucoma. A 25-year review. Am J Ophthalmol. 1966 Sep;62(3):499-507. doi: 10.1016/0002-9394(66)91330-4[][][][][][]
  171. Gillies WE, Brooks AM. Clinical features at presentation of anterior segment pigment dispersion syndrome. Clin Exp Ophthalmol. 2001 Jun;29(3):125-7. doi: 10.1046/j.1442-9071.2001.00391.x[][][][]
  172. Lichter PR, Shaffer RN. Diagnostic and prognostic signs in pigmentary glaucoma. Trans Am Acad Ophthalmol Otolaryngol. 1970 Sep-Oct;74(5):984-98.[][][]
  173. BICK MW. Sex differences in pigmentary glaucoma. Am J Ophthalmol. 1962 Nov;54:831-7. doi: 10.1016/0002-9394(62)94170-3[][]
  174. Migliazzo CV, Shaffer RN, Nykin R, Magee S. Long-term analysis of pigmentary dispersion syndrome and pigmentary glaucoma. Ophthalmology. 1986 Dec;93(12):1528-36. doi: 10.1016/s0161-6420(86)33526-7[][][]
  175. SCHEIE HG, FLEISCHHAUER HW. Idiopathic atrophy of the epithelial layers of the iris and ciliary body; a clinical study. AMA Arch Ophthalmol. 1958 Feb;59(2):216-28. doi: 10.1001/archopht.1958.00940030084007[][]
  176. Kaiser-Kupfer MI, Kupfer C, McCain L. Asymmetric pigment dispersion syndrome. Trans Am Ophthalmol Soc. 1983;81:310-24. https://pmc.ncbi.nlm.nih.gov/articles/instance/1312456/pdf/taos00018-0336.pdf[][]
  177. Rodrigues MM, Spaeth GL, Weinreb S, Sivalingam E. Spectrum of trabecular pigmentation in open-angle glaucoma: a clinicopathologic study. Trans Sect Ophthalmol Am Acad Ophthalmol Otolaryngol. 1976 Mar-Apr;81(2):258-76.[][][]
  178. Doyle JW, Hansen JE, Smith MF, Hamed LM, McGorray S, Sherwood MB. Use of scheimpflug photography to study iris configuration in patients with pigment dispersion syndrome and pigmentary glaucoma. J Glaucoma. 1995 Dec;4(6):398-405.[][]
  179. Mora P, Sangermani C, Ghirardini S, Carta A, Ungaro N, Gandolfi S. Ultrasound biomicroscopy and iris pigment dispersion: a case–control study. Br J Ophthalmol. 2010 Apr;94(4):428-32. doi: 10.1136/bjo.2009.162065[][]
  180. Kanadani FN, Dorairaj S, Langlieb AM, Shihadeh WA, Tello C, Liebmann JM, Ritch R. Ultrasound biomicroscopy in asymmetric pigment dispersion syndrome and pigmentary glaucoma. Arch Ophthalmol. 2006 Nov;124(11):1573-6. doi: 10.1001/archopht.124.11.1573[][][]
  181. Lord FD, Pathanapitoon K, Mikelberg FS. Keratometry and axial length in pigment dispersion syndrome: a descriptive case-control study. J Glaucoma. 2001 Oct;10(5):383-5. doi: 10.1097/00061198-200110000-00003[][]
  182. Yip LW, Sothornwit N, Berkowitz J, Mikelberg FS. A comparison of interocular differences in patients with pigment dispersion syndrome. J Glaucoma. 2009 Jan;18(1):1-5. doi: 10.1097/IJG.0b013e31816f767b[][][][]
  183. Scheie stripe in pigment dispersion syndrome. https://webeye.ophth.uiowa.edu/eyeforum/atlas/pages/Scheie-stripe/index.htm#gsc.tab=0[]
  184. Laemmer R, Mardin CY, Juenemann AG. Visualization of changes of the iris configuration after peripheral laser iridotomy in primary melanin dispersion syndrome using optical coherence tomography. J Glaucoma. 2008 Oct-Nov;17(7):569-70. doi: 10.1097/IJG.0b013e318168f02c[]
  185. Carassa RG, Bettin P, Fiori M, Brancato R. Nd:YAG laser iridotomy in pigment dispersion syndrome: an ultrasound biomicroscopic study. Br J Ophthalmol. 1998 Feb;82(2):150-3. doi: 10.1136/bjo.82.2.150[]
  186. Breingan PJ, Esaki K, Ishikawa H, Liebmann JM, Greenfield DS, Ritch R. Iridolenticular contact decreases following laser iridotomy for pigment dispersion syndrome. Arch Ophthalmol. 1999 Mar;117(3):325-8. doi: 10.1001/archopht.117.3.325[]
  187. Jampel HD. Lack of effect of peripheral laser iridotomy in pigment dispersion syndrome. Arch Ophthalmol. 1993 Dec;111(12):1606. doi: 10.1001/archopht.1993.01090120028009[]
  188. Haynes WL, Johnson AT, Alward WL. Inhibition of exercise-induced pigment dispersion in a patient with the pigmentary dispersion syndrome. Am J Ophthalmol. 1990 May 15;109(5):601-2. doi: 10.1016/s0002-9394(14)70696-8[]
  189. Haynes WL, Alward WL, Tello C, Liebmann JM, Ritch R. Incomplete elimination of exercise-induced pigment dispersion by laser iridotomy in pigment dispersion syndrome. Ophthalmic Surg Lasers. 1995 Sep-Oct;26(5):484-6.[]
  190. Gandolfi SA, Vecchi M. Effect of a YAG laser iridotomy on intraocular pressure in pigment dispersion syndrome. Ophthalmology. 1996 Oct;103(10):1693-5. doi: 10.1016/s0161-6420(96)30445-4[]
  191. Reistad CE, Shields MB, Campbell DG, Ritch R, Wang JC, Wand M; American Glaucoma Society Pigmentary Glaucoma Iridotomy Study Group. The influence of peripheral iridotomy on the intraocular pressure course in patients with pigmentary glaucoma. J Glaucoma. 2005 Aug;14(4):255-9. doi: 10.1097/01.ijg.0000169383.05605.4b[]
  192. Michelessi M, Lindsley K. Peripheral iridotomy for pigmentary glaucoma. Cochrane Database Syst Rev. 2016 Feb 12;2(2):CD005655. doi: 10.1002/14651858.CD005655.pub2[]
  193. Birt CM. Intraocular pressure spike after YAG iridotomy in patients with pigment dispersion. Can J Ophthalmol. 2004 Apr;39(3):234-9. doi: 10.1016/s0008-4182(04)80119-4[]
  194. European Glaucoma Society Terminology and Guidelines for Glaucoma, 4th Edition – Chapter 2: Classification and terminologySupported by the EGS Foundation: Part 1: Foreword; Introduction; Glossary; Chapter 2 Classification and Terminology. Br J Ophthalmol. 2017 May;101(5):73-127. doi: 10.1136/bjophthalmol-2016-EGSguideline.002[]
  195. Richter CU, Richardson TM, Grant WM. Pigmentary dispersion syndrome and pigmentary glaucoma. A prospective study of the natural history. Arch Ophthalmol. 1986 Feb;104(2):211-5. doi: 10.1001/archopht.1986.01050140065021[]
  196. Speakman JS. Pigmentary dispersion. Br J Ophthalmol. 1981 Apr;65(4):249-51. doi: 10.1136/bjo.65.4.249[]
  197. Ritch R. Nonprogressive low-tension glaucoma with pigmentary dispersion. Am J Ophthalmol. 1982 Aug;94(2):190-6. doi: 10.1016/0002-9394(82)90074-5[]
  198. Knight LSW, Ruddle JB, Taranath DA, et al. Childhood and Early Onset Glaucoma Classification and Genetic Profile in a Large Australasian Disease Registry. Ophthalmology. 2021 Nov;128(11):1549-1560. https://www.aaojournal.org/article/S0161-6420(21)00288-8/fulltext[][]
  199. Weinreb R.N., Grajewski A.L., Papadopoulos M., Grigg J.,Freedman S.F. Childhood glaucoma: the 9th consensus report of the World Glaucoma Association. Netherlands: Kugler Publications, 2013.[][][][][][][][][][][][][][][][][][]
  200. Xia Q, Zhang D, Zhuang Y, Dai Y, Jia H, Du Q, Wen T, Jiang Y. Animal Model Contributions to Primary Congenital Glaucoma. J Ophthalmol. 2022 May 26;2022:6955461. doi: 10.1155/2022/6955461[]
  201. Kaur K, Zeppieri M, Gurnani B. Primary Congenital Glaucoma. [Updated 2024 Nov 10]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK574553[][][][]
  202. Abu-Amero KK, Edward DP. Primary Congenital Glaucoma. 2004 Sep 30 [Updated 2017 Aug 17]. In: Adam MP, Feldman J, Mirzaa GM, et al., editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2025. Available from: https://www.ncbi.nlm.nih.gov/books/NBK1135[]
  203. Primary Congenital Glaucoma. https://eyewiki.org/Primary_Congenital_Glaucoma[][][][][][][]
  204. Lingham G, Thakur S, Safi S, Gordon I, Evans JR, Keel S. A systematic review of clinical practice guidelines for childhood glaucoma. BMJ Open Ophthalmol. 2022 Jan 31;7(1):e000933. doi: 10.1136/bmjophth-2021-000933[]
  205. Yeung HH, Kumar-Singh R, Walton DS. Infantile Aphakic Glaucoma: A Proposed Mechanism. J Pediatr Ophthalmol Strabismus. 2022 Jul-Aug;59(4):236-242. doi: 10.3928/01913913-20210929-02[]
  206. Jafer Chardoub AA, Zeppieri M, Blair K. Juvenile Glaucoma. [Updated 2024 Aug 8]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK562263[]
  207. Alward, W.L.M. Glaucoma: The Requisites in Ophthalmology. St. Louis: Mosby Inc; 2000.[][]
  208. Haddad A, Ait Boujmia OK, El Maaloum L, Dehbi H. Meta-analysis of CYP1B1 gene mutations in primary congenital glaucoma patients. Eur J Ophthalmol. 2021 Nov;31(6):2796-2807. doi: 10.1177/11206721211016308. Erratum in: Eur J Ophthalmol. 2024 Jul;34(4):NP71-NP72. doi: 10.1177/11206721231156301[][][]
  209. Greco, A. et al., 2022, ‘Childhood Glaucoma and Medical Treatment: An Up to Date’, in G. L. Giudice (ed.), Vision Correction and Eye Surgery, IntechOpen, London. 10.5772/intechopen.100579.[][]
  210. Feroze KB, Blair K, Patel BC. Buphthalmos. [Updated 2025 Jan 20]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK430887[][]
  211. Beck AD. Diagnosis and management of pediatric glaucoma. Ophthalmol Clin North Am. 2001 Sep;14(3):501-12. doi: 10.1016/s0896-1549(05)70248-0[]
  212. Mark HH. Buphthalmos: early glaucoma history. Acta Ophthalmol. 2011;89:591–594. doi: 10.1111/j.1755-3768.2009.01783.x[]
  213. Bouhenni RA, Ricker I, Hertle RW. Prevalence and clinical characteristics of childhood Glaucoma at a tertiary care Children’s hospital. J Glaucoma. 2019;28:655–659. doi: 10.1097/IJG.0000000000001259[]
  214. Vasileiadis GT, Frangouli O. Unilateral congenital buphthalmos. BMJ Case Rep. 2015 Jun 3;2015:bcr2015210979. doi: 10.1136/bcr-2015-210979[]
  215. Kaushik S, Dhingra D, Joshi G, Bazgain K, Thattaruthody F, Pandav SS. Acute Hydrops in the Fellow Eye of Infants with Primary Congenital Glaucoma after Intraocular Pressure Reduction in 1 Eye. Ophthalmol Glaucoma. 2020 Jul-Aug;3(4):301-305. doi: 10.1016/j.ogla.2020.03.007[]
  216. Haab striae in primary congenital glaucoma. https://eyerounds.org/atlas/pages/Haab/index.htm#gsc.tab=0[]
  217. Thau A, Lloyd M, Freedman S, Beck A, Grajewski A, Levin AV. New classification system for pediatric glaucoma: implications for clinical care and a research registry. Curr Opin Ophthalmol. 2018 Sep;29(5):385-394. doi: 10.1097/ICU.0000000000000516[]
  218. Kong L, et al. An update on progress and the changing epidemiology of causes of childhood blindness worldwide. J AAPOS. 2012;16(6):501–507. doi: 10.1016/j.jaapos.2012.09.004[]
  219. Jat NS, Tripathy K. Peters Anomaly. [Updated 2023 Aug 25]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK580540[][]
  220. Idrees F, Vaideanu D, Fraser SG, Sowden JC, Khaw PT. A review of anterior segment dysgeneses. Surv Ophthalmol. 2006 May-Jun;51(3):213-31. doi: 10.1016/j.survophthal.2006.02.006[]
  221. Weinreb RN, Aung T, Medeiros FA. The pathophysiology and treatment of glaucoma: a review. JAMA. 2014 May 14;311(18):1901-11. doi: 10.1001/jama.2014.3192[][][][][][]
  222. Wiggs JL, Pasquale LR. Genetics of glaucoma. Hum Mol Genet. 2017 Aug 1;26(R1):R21-R27. doi: 10.1093/hmg/ddx184[][]
  223. Lewis CJ, Hedberg-Buenz A, DeLuca AP, Stone EM, Alward WLM, Fingert JH. Primary congenital and developmental glaucomas. Hum Mol Genet. 2017 Aug 1;26(R1):R28-R36. doi: 10.1093/hmg/ddx205[]
  224. Shah M, Bouhenni R, Benmerzouga I. Geographical Variability in CYP1B1 Mutations in Primary Congenital Glaucoma. J Clin Med. 2022 Apr 6;11(7):2048. doi: 10.3390/jcm11072048[][][]
  225. Bejjani BA, Xu L, Armstrong D, Lupski JR, Reneker LW. Expression patterns of cytochrome P4501B1 (Cyp1b1) in FVB/N mouse eyes. Exp Eye Res. 2002 Sep;75(3):249-57. https://doi.org/10.1006/exer.2002.2025[]
  226. Teixeira LB, Zhao Y, Dubielzig RR, Sorenson CM, Sheibani N. Ultrastructural abnormalities of the trabecular meshwork extracellular matrix in Cyp1b1-deficient mice. Vet Pathol. 2015 Mar;52(2):397-403. doi: 10.1177/0300985814535613[]
  227. Williams AL, Eason J, Chawla B, Bohnsack BL. Cyp1b1 Regulates Ocular Fissure Closure Through a Retinoic Acid-Independent Pathway. Invest Ophthalmol Vis Sci. 2017 Feb 1;58(2):1084-1097. doi: 10.1167/iovs.16-20235[]
  228. Suri F, Yazdani S, Narooie-Nejhad M, Zargar SJ, Paylakhi SH, Zeinali S, Pakravan M, Elahi E. Variable expressivity and high penetrance of CYP1B1 mutations associated with primary congenital glaucoma. Ophthalmology. 2009 Nov;116(11):2101-9. doi: 10.1016/j.ophtha.2009.04.045[]
  229. Alghamdi A, Aldossary W, Albahkali S, Alotaibi B, Alrfaei BM. The loss of microglia activities facilitates glaucoma progression in association with CYP1B1 gene mutation (p.Gly61Glu). PLoS One. 2020 Nov 10;15(11):e0241902. doi: 10.1371/journal.pone.0241902[]
  230. Schlötzer-Schrehardt U, Zenkel M, Küchle M, Sakai LY, Naumann GO. Role of transforming growth factor-beta1 and its latent form binding protein in pseudoexfoliation syndrome. Exp Eye Res. 2001 Dec;73(6):765-80. doi: 10.1006/exer.2001.1084[]
  231. Hyytiäinen M, Keski-Oja J. Latent TGF-beta binding protein LTBP-2 decreases fibroblast adhesion to fibronectin. J Cell Biol. 2003 Dec 22;163(6):1363-74. doi: 10.1083/jcb.200309105[]
  232. Ali M, McKibbin M, Booth A, et al. Null mutations in LTBP2 cause primary congenital glaucoma. Am J Hum Genet. 2009 May;84(5):664-71. doi: 10.1016/j.ajhg.2009.03.017[]
  233. Suri F, Yazdani S, Elahi E. LTBP2 knockdown and oxidative stress affect glaucoma features including TGFβ pathways, ECM genes expression and apoptosis in trabecular meshwork cells. Gene. 2018 Oct 5;673:70-81. doi: 10.1016/j.gene.2018.06.038[]
  234. Sarfarazi M, Akarsu AN, Hossain A, Turacli ME, Aktan SG, Barsoum-Homsy M, Chevrette L, Sayli BS. Assignment of a locus (GLC3A) for primary congenital glaucoma (Buphthalmos) to 2p21 and evidence for genetic heterogeneity. Genomics. 1995 Nov 20;30(2):171-7. doi: 10.1006/geno.1995.9888[]
  235. Fan BJ, Wiggs JL. Glaucoma: genes, phenotypes, and new directions for therapy. J Clin Invest. 2010 Sep;120(9):3064-72. doi: 10.1172/JCI43085[]
  236. Kumar A, Han Y, Oatts JT. Genetic changes and testing associated with childhood glaucoma: A systematic review. PLoS One. 2024 Feb 22;19(2):e0298883. doi: 10.1371/journal.pone.0298883[]
  237. Yu-Wai-Man C, Arno G, Brookes J, Garcia-Feijoo J, Khaw PT, Moosajee M. Primary congenital glaucoma including next-generation sequencing-based approaches: clinical utility gene card. Eur J Hum Genet. 2018 Nov;26(11):1713-1718. doi: 10.1038/s41431-018-0227-y[][][][]
  238. Tamçelik N, Atalay E, Bolukbasi S, Çapar O, Ozkok A. Demographic features of subjects with congenital glaucoma. Indian J Ophthalmol. 2014 May;62(5):565-9. doi: 10.4103/0301-4738.126988[]
  239. Genĉík A. Epidemiology and genetics of primary congenital glaucoma in Slovakia. Description of a form of primary congenital glaucoma in gypsies with autosomal-recessive inheritance and complete penetrance. Dev Ophthalmol. 1989;16:76-115.[]
  240. deLuise VP, Anderson DR. Primary infantile glaucoma (congenital glaucoma). Surv Ophthalmol. 1983 Jul-Aug;28(1):1-19. doi: 10.1016/0039-6257(83)90174-1[][][][][]
  241. Ohtake Y, Tanino T, Suzuki Y, Miyata H, Taomoto M, Azuma N, Tanihara H, Araie M, Mashima Y. Phenotype of cytochrome P4501B1 gene (CYP1B1) mutations in Japanese patients with primary congenital glaucoma. Br J Ophthalmol. 2003 Mar;87(3):302-4. doi: 10.1136/bjo.87.3.302[]
  242. Alabdulwahhab KM, Ahmad MS. Visual Impairment and Blindness in Saudi Arabia’s School for the Blind: A Cross-Sectional Study. Clin Optom (Auckl). 2020 Oct 7;12:169-173. doi: 10.2147/OPTO.S265293[]
  243. Souma T, Tompson SW, Thomson BR, et al. Angiopoietin receptor TEK mutations underlie primary congenital glaucoma with variable expressivity. J Clin Invest. 2016 Jul 1;126(7):2575-87. doi: 10.1172/JCI85830[]
  244. Badawi AH, Al-Muhaylib AA, Al Owaifeer AM, Al-Essa RS, Al-Shahwan SA. Primary congenital glaucoma: An updated review. Saudi J Ophthalmol. 2019 Oct-Dec;33(4):382-388. doi: 10.1016/j.sjopt.2019.10.002[]
  245. Barkan O. Pathogenesis of congenital glaucoma: gonioscopic and anatomic observation of the angle of the anterior chamber in the normal eye and in congenital glaucoma. Am J Ophthalmol. 1955;40:1-11.[]
  246. Worst JGF. The pathogenesis of congential glaucoma. An embryological and goniosurgical study. Springfield, Ill: Charles C. Thomas; 1966.[]
  247. Mandal AK, Chakrabarti D. Update on congenital glaucoma. Indian J Ophthalmol. 2011 Jan;59 Suppl(Suppl1):S148-57. doi: 10.4103/0301-4738.73683[]
  248. Mandal AK, Chakrabarti D, Gothwal VK. Approach to primary congenital glaucoma: A perspective. Taiwan J Ophthalmol. 2023 Oct 19;13(4):451-460. doi: 10.4103/tjo.TJO-D-23-00104[]
  249. Anderson DR. The development of the trabecular meshwork and its abnormality in primary infantile glaucoma. Trans Am Ophthalmol Soc. 1981;79:458-85. https://pmc.ncbi.nlm.nih.gov/articles/instance/1312195/pdf/taos00020-0482.pdf[][]
  250. Tawara A, Inomata H. Developmental immaturity of the trabecular meshwork in congenital glaucoma. Am J Ophthalmol. 1981 Oct;92(4):508-25. doi: 10.1016/0002-9394(81)90644-9[][]
  251. García-Antón MT, Salazar JJ, de Hoz R, Rojas B, Ramírez AI, Triviño A, Aroca-Aguilar JD, García-Feijoo J, Escribano J, Ramírez JM. Goniodysgenesis variability and activity of CYP1B1 genotypes in primary congenital glaucoma. PLoS One. 2017 Apr 27;12(4):e0176386. doi: 10.1371/journal.pone.0176386[]
  252. Pilat AV, Proudlock FA, Shah S, Sheth V, Purohit R, Abbot J, Gottlob I. Assessment of the anterior segment of patients with primary congenital glaucoma using handheld optical coherence tomography. Eye (Lond). 2019 Aug;33(8):1232-1239. doi: 10.1038/s41433-019-0369-3[]
  253. Papadopoulos M, Cable N, Rahi J, Khaw PT; BIG Eye Study Investigators. The British Infantile and Childhood Glaucoma (BIG) Eye Study. Invest Ophthalmol Vis Sci. 2007 Sep;48(9):4100-6. doi: 10.1167/iovs.06-1350[]
  254. Sihota R, Sidhu T, Agarwal R, Sharma A, Gupta A, Sethi A, Dada T, Pandey V. Evaluating target intraocular pressures in primary congenital glaucoma. Indian J Ophthalmol. 2021 Aug;69(8):2082-2087. doi: 10.4103/ijo.IJO_3473_20[][]
  255. Mandal AK. Acute Corneal Hydrops in Children with Primary Infantile Glaucoma: A Report of 31 Cases over 23 Years at the LVPEI. PLoS One. 2016 Jun 1;11(6):e0156108. doi: 10.1371/journal.pone.0156108[]
  256. Drechsler J, Lee A, Maripudi S, Kueny L, Levin MR, Saeedi OJ, Bazemore M, Karwoski B, Birdsong R, Martinez C, Jaafar MS, Yousaf S, Ahmed ZM, Madigan WP, Alexander JL. Corneal Structural Changes in Congenital Glaucoma. Eye Contact Lens. 2022 Jan 1;48(1):27-32. doi: 10.1097/ICL.0000000000000844[]
  257. Gupta S, Mahalingam K, Singh A, Selvan H, Somarajan BI, Gupta V. Posterior corneal morphological changes in primary congenital glaucoma. Indian J Ophthalmol. 2022 Jul;70(7):2571-2577. doi: 10.4103/ijo.IJO_317_22[][]
  258. Sihota R, Mahalingam K, Maurya AK, Sharma A, Bukke AN, Dada T. Primary congenital glaucoma: An iridotrabeculodysgenesis? Indian J Ophthalmol. 2024 Mar 1;72(3):328-334. doi: 10.4103/IJO.IJO_370_23[]
  259. Walton DS. Chronic newborn primary congenital glaucoma with secondary lens subluxation. J Pediatr Ophthalmol Strabismus. 2009 Jul-Aug;46(4):200, 231. doi: 10.3928/01913913-20090706-02[]
  260. Snehi S, Singh AK, Kaushik S. Acquired Lens Zonular Loss with Bean Pot Optic Disc Cupping in Congenital Glaucoma. Ophthalmol Glaucoma. 2023 Mar-Apr;6(2):159. doi: 10.1016/j.ogla.2023.01.001[]
  261. Gupta V, James MK, Singh A, Kumar S, Gupta S, Sharma A, Sihota R, Kennedy DJ. Differences in Optic Disc Characteristics of Primary Congenital Glaucoma, Juvenile, and Adult Onset Open Angle Glaucoma Patients. J Glaucoma. 2016 Mar;25(3):239-43. doi: 10.1097/IJG.0000000000000154[]
  262. Walton DS. Diagnosis and treatment of glaucoma in childhood. In: Epstein DL, editor. Chandler and Grant’s Glaucoma. 3rd ed. Phila: Lea & Febinger; 1986.[]
  263. Morin JD, Bryars JH. Causes of loss of vision in congenital glaucoma. Arch Ophthalmol. 1980 Sep;98(9):1575-6. doi: 10.1001/archopht.1980.01020040427005[]
  264. Kiskis AA, Markowitz SN, Morin JD. Corneal diameter and axial length in congenital glaucoma. Can J Ophthalmol. 1985 Apr;20(3):93-7.[]
  265. Kessing SV, Gregersen E. The distended disc in early stages of congenital glaucoma. Acta Ophthalmol (Copenh). 1977 Jun;55(3):431-5. doi: 10.1111/j.1755-3768.1977.tb06119.x[]
  266. Quigley HA. The pathogenesis of reversible cupping in congenital glaucoma. Am J Ophthalmol. 1977 Sep;84(3):358-70. doi: 10.1016/0002-9394(77)90680-8[]
  267. Fayed MA, Chen TC. Pediatric intraocular pressure measurements: Tonometers, central corneal thickness, and anesthesia. Surv Ophthalmol. 2019 Nov-Dec;64(6):810-825. doi: 10.1016/j.survophthal.2019.05.003[]
  268. Eisenberg DL, Sherman BG, McKeown CA, Schuman JS. Tonometry in adults and children. A manometric evaluation of pneumatonometry, applanation, and TonoPen in vitro and in vivo. Ophthalmology. 1998 Jul;105(7):1173-81. doi: 10.1016/S0161-6420(98)97016-6[]
  269. Grigorian F, Grigorian AP, Olitsky SE. The use of the iCare tonometer reduced the need for anesthesia to measure intraocular pressure in children. J AAPOS. 2012 Dec;16(6):508-10. doi: 10.1016/j.jaapos.2012.07.004[]
  270. Flemmons MS, Hsiao YC, Dzau J, Asrani S, Jones S, Freedman SF. Icare rebound tonometry in children with known and suspected glaucoma. J AAPOS. 2011 Apr;15(2):153-7. doi: 10.1016/j.jaapos.2010.11.022[]
  271. McKee EC, Ely AL, Duncan JE, Dosunmu EO, Freedman SF. A comparison of Icare PRO and Tono-Pen XL tonometers in anesthetized children. J AAPOS. 2015 Aug;19(4):332-7. doi: 10.1016/j.jaapos.2015.04.004[]
  272. Sampaolesi R, Caruso R. Ocular echometry in the diagnosis of congenital glaucoma. Arch Ophthalmol. 1982 Apr;100(4):574-7. doi: 10.1001/archopht.1982.01030030576003[]
  273. Henriques MJ, Vessani RM, Reis FA, de Almeida GV, Betinjane AJ, Susanna R Jr. Corneal thickness in congenital glaucoma. J Glaucoma. 2004 Jun;13(3):185-8. doi: 10.1097/00061198-200406000-00002[]
  274. Doozandeh A, Yazdani S, Ansari S, Pakravan M, Motevasseli T, Hosseini B, Yasseri M. Corneal profile in primary congenital glaucoma. Acta Ophthalmol. 2017 Nov;95(7):e575-e581. doi: 10.1111/aos.13357[][]
  275. Zareei A, Razeghinejad MR, Salouti R. Corneal Biomechanical Properties and Thickness in Primary Congenital Glaucoma and Normal Eyes: A Comparative Study. Med Hypothesis Discov Innov Ophthalmol. 2018 Summer;7(2):68-72. https://pmc.ncbi.nlm.nih.gov/articles/PMC6146241[][]
  276. Donahue SP, Porter A. SITA visual field testing in children. J AAPOS. 2001 Apr;5(2):114-7. doi: 10.1067/mpa.2001.113840[]
  277. Salchow DJ, Oleynikov YS, Chiang MF, Kennedy-Salchow SE, Langton K, Tsai JC, Al-Aswad LA. Retinal nerve fiber layer thickness in normal children measured with optical coherence tomography. Ophthalmology. 2006 May;113(5):786-91. doi: 10.1016/j.ophtha.2006.01.036[]
  278. Ahn HC, Son HW, Kim JS, Lee JH. Quantitative analysis of retinal nerve fiber layer thickness of normal children and adolescents. Korean J Ophthalmol. 2005 Sep;19(3):195-200. doi: 10.3341/kjo.2005.19.3.195[]
  279. Hess DB, Asrani SG, Bhide MG, Enyedi LB, Stinnett SS, Freedman SF. Macular and retinal nerve fiber layer analysis of normal and glaucomatous eyes in children using optical coherence tomography. Am J Ophthalmol. 2005 Mar;139(3):509-17. doi: 10.1016/j.ajo.2004.10.047[]
  280. Rao A, Sahoo B, Kumar M, Varshney G, Kumar R. Retinal nerve fiber layer thickness in children <18 years by spectral-domain optical coherence tomography. Semin Ophthalmol. 2013 Mar;28(2):97-102. doi: 10.3109/08820538.2012.760626[]
  281. Rotruck JC, House RJ, Freedman SF, Kelly MP, Enyedi LB, Prakalapakorn SG, Lim ME, El-Dairi MA. Optical Coherence Tomography Normative Peripapillary Retinal Nerve Fiber Layer and Macular Data in Children 0-5 Years of Age. Am J Ophthalmol. 2019 Dec;208:323-330. doi: 10.1016/j.ajo.2019.06.025[]
  282. Hsu ST, Chen X, Ngo HT, House RJ, Kelly MP, Enyedi LB, Materin MA, El-Dairi MA, Freedman SF, Toth CA, Vajzovic L. Imaging Infant Retinal Vasculature with OCT Angiography. Ophthalmol Retina. 2019 Jan;3(1):95-96. doi: 10.1016/j.oret.2018.06.017[]
  283. Gouider D, Choura R, Mekni M, Sayadi J, Malek I, Nacef L. Sclerocornea: A rare ocular condition. J Fr Ophtalmol. 2021 Sep;44(7):1089-1091. doi: 10.1016/j.jfo.2021.05.001[]
  284. Gurnani B, Christy J, Narayana S, Rajkumar P, Kaur K, Gubert J. Retrospective multifactorial analysis of Pythium keratitis and review of literature. Indian J Ophthalmol. 2021 May;69(5):1095-1101. doi: 10.4103/ijo.IJO_1808_20[]
  285. Gurnani B, Narayana S, Christy J, Rajkumar P, Kaur K, Gubert J. Successful management of pediatric pythium insidiosum keratitis with cyanoacrylate glue, linezolid, and azithromycin: Rare case report. Eur J Ophthalmol. 2022 Sep;32(5):NP87-NP91. doi: 10.1177/11206721211006564[]
  286. Gurnani B, Kaur K. Pythium Keratitis. [Updated 2023 Jul 17]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK573072[]
  287. Raven ML, Rodriguez ME, Potter HD. Corneal Leukoma with Features of Both Sclerocornea and Peter’s Anomaly. Ophthalmology. 2016 Sep;123(9):1988. doi: 10.1016/j.ophtha.2016.05.011[]
  288. Altamirano F, Ortiz-Morales G, O’Connor-Cordova MA, Sancén-Herrera JP, Zavala J, Valdez-Garcia JE. Fuchs endothelial corneal dystrophy: an updated review. Int Ophthalmol. 2024 Feb 12;44(1):61. doi: 10.1007/s10792-024-02994-1[]
  289. Nusbaum L, Paul M, Maharshak I. [DEEP ORBITAL DERMOID CYSTS]. Harefuah. 2023 Feb;162(2):98-102. Hebrew.[]
  290. Balamurugan S, Gurnani B, Kaur K, Gireesh P, Narayana S. Traumatic intralenticular abscess-What is so different? Indian J Radiol Imaging. 2020 Jan-Mar;30(1):92-94. doi: 10.4103/ijri.IJRI_369_19[]
  291. Kaur K, Gurnani B, Devy N. Atypical optic neuritis – a case with a new surprise every visit. GMS Ophthalmol Cases. 2020 Feb 27;10:Doc11. doi: 10.3205/oc000138[]
  292. Coppens G, Stalmans I, Zeyen T, Casteels I. The safety and efficacy of glaucoma medication in the pediatric population. J Pediatr Ophthalmol Strabismus. 2009 Jan-Feb;46(1):12-8. doi: 10.3928/01913913-20090101-05[]
  293. Buckley MM, Goa KL, Clissold SP. Ocular betaxolol. A review of its pharmacological properties, and therapeutic efficacy in glaucoma and ocular hypertension. Drugs. 1990 Jul;40(1):75-90. doi: 10.2165/00003495-199040010-00005[]
  294. Maren TH, Haywood JR, Chapman SK, Zimmerman TJ. The pharmacology of methazolamide in relation to the treatment of glaucoma. Invest Ophthalmol Vis Sci. 1977 Aug;16(8):730-42.[]
  295. Capino AC, Dannaway DC, Miller JL. Metabolic Acidosis with Ophthalmic Dorzolamide in a Neonate. J Pediatr Pharmacol Ther. 2016 May-Jun;21(3):256-9. doi: 10.5863/1551-6776-21.3.256[]
  296. Maeda-Chubachi T, Chi-Burris K, Simons BD, Freedman SF, Khaw PT, Wirostko B, Yan E; A6111137 Study Group. Comparison of latanoprost and timolol in pediatric glaucoma: a phase 3, 12-week, randomized, double-masked multicenter study. Ophthalmology. 2011 Oct;118(10):2014-21. doi: 10.1016/j.ophtha.2011.03.010[]
  297. Kim JS, Blizzard S, Woodward JA, Leyngold IM, Liss J, Freedman SF. Prostaglandin-Associated Periorbitopathy in Children and Young Adults with Glaucoma. Ophthalmol Glaucoma. 2020 Jul-Aug;3(4):288-294. doi: 10.1016/j.ogla.2020.03.009[]
  298. El Sayed Y, Esmael A, Mettias N, El Sanabary Z, Gawdat G. Factors influencing the outcome of goniotomy and trabeculotomy in primary congenital glaucoma. Br J Ophthalmol. 2021 Sep;105(9):1250-1255. doi: 10.1136/bjophthalmol-2018-313387[]
  299. Huang H, Bao WJ, Yamamoto T, Kawase K, Sawada A. Postoperative outcome of three different procedures for childhood glaucoma. Clin Ophthalmol. 2018 Dec 17;13:1-7. doi: 10.2147/OPTH.S186929[]
  300. Mandal AK, Gothwal VK, Nutheti R. Surgical outcome of primary developmental glaucoma: a single surgeon’s long-term experience from a tertiary eye care centre in India. Eye (Lond). 2007 Jun;21(6):764-74. doi: 10.1038/sj.eye.6702324[]
  301. Khalil DH, Abdelhakim MA. Primary trabeculotomy compared to combined trabeculectomy-trabeculotomy in congenital glaucoma: 3-year study. Acta Ophthalmol. 2016 Nov;94(7):e550-e554. doi: 10.1111/aos.13018[]
  302. Jayaram H, Scawn R, Pooley F, Chiang M, Bunce C, Strouthidis NG, Khaw PT, Papadopoulos M. Long-Term Outcomes of Trabeculectomy Augmented with Mitomycin C Undertaken within the First 2 Years of Life. Ophthalmology. 2015 Nov;122(11):2216-22. doi: 10.1016/j.ophtha.2015.07.028[]
  303. Khaw PT, Chiang M, Shah P, Sii F, Lockwood A, Khalili A. Enhanced trabeculectomy: the Moorfields Safer Surgery System. Dev Ophthalmol. 2012;50:1-28. doi: 10.1159/000334776[]
  304. Low S, Hamada S, Nischal KK. Antimetabolite and releasable suture augmented filtration surgery in refractory pediatric glaucomas. J AAPOS. 2008 Apr;12(2):166-72. doi: 10.1016/j.jaapos.2007.09.012[]
  305. Beck AD, Wilson WR, Lynch MG, Lynn MJ, Noe R. Trabeculectomy with adjunctive mitomycin C in pediatric glaucoma. Am J Ophthalmol. 1998 Nov;126(5):648-57. doi: 10.1016/s0002-9394(98)00227-x[]
  306. Margeta MA, Kuo AN, Proia AD, Freedman SF. Staying away from the optic nerve: a formula for modifying glaucoma drainage device surgery in pediatric and other small eyes. J AAPOS. 2017 Feb;21(1):39-43.e1. doi: 10.1016/j.jaapos.2016.09.027[]
  307. Ishida K, Mandal AK, Netland PA. Glaucoma drainage implants in pediatric patients. Ophthalmol Clin North Am. 2005 Sep;18(3):431-42, vii. doi: 10.1016/j.ohc.2005.05.009[]
  308. Tung I, Marcus I, Thiamthat W, Freedman SF. Second glaucoma drainage devices in refractory pediatric glaucoma: failure by fibrovascular ingrowth. Am J Ophthalmol. 2014 Jul;158(1):113-7. doi: 10.1016/j.ajo.2014.03.017[]
  309. Cunliffe IA, Molteno AC. Long-term follow-up of Molteno drains used in the treatment of glaucoma presenting in childhood. Eye (Lond). 1998;12 ( Pt 3a):379-85. doi: 10.1038/eye.1998.90[]
  310. Beck AD, Freedman S, Kammer J, Jin J. Aqueous shunt devices compared with trabeculectomy with Mitomycin-C for children in the first two years of life. Am J Ophthalmol. 2003 Dec;136(6):994-1000. doi: 10.1016/s0002-9394(03)00714-1[]
  311. Tai AX, Song JC. Surgical outcomes of Baerveldt implants in pediatric glaucoma patients. J AAPOS. 2014 Dec;18(6):550-3. doi: 10.1016/j.jaapos.2014.08.003[]
  312. Mandalos A, Tailor R, Parmar T, Sung V. The Long-term Outcomes of Glaucoma Drainage Device in Pediatric Glaucoma. J Glaucoma. 2016 Mar;25(3):e189-95. doi: 10.1097/IJG.0000000000000164[]
  313. Razeghinejad MR, Kaffashan S, Nowroozzadeh MH. Results of Ahmed glaucoma valve implantation in primary congenital glaucoma. J AAPOS. 2014 Dec;18(6):590-5. doi: 10.1016/j.jaapos.2014.08.008[]
  314. Pakravan M, Esfandiari H, Yazdani S, Doozandeh A, Dastborhan Z, Gerami E, Kheiri B, Pakravan P, Yaseri M, Hassanpour K. Clinical outcomes of Ahmed glaucoma valve implantation in pediatric glaucoma. Eur J Ophthalmol. 2019 Jan;29(1):44-51. doi: 10.1177/1120672118761332[]
  315. Abdelrahman AM, El Sayed YM. Micropulse Versus Continuous Wave Transscleral Cyclophotocoagulation in Refractory Pediatric Glaucoma. J Glaucoma. 2018 Oct;27(10):900-905. doi: 10.1097/IJG.0000000000001053[]
  316. Way AL, Nischal KK. High-frequency ultrasound-guided transscleral diode laser cyclophotocoagulation. Br J Ophthalmol. 2014 Jul;98(7):992-4. doi: 10.1136/bjophthalmol-2014-305163[]
  317. Bock CJ, Freedman SF, Buckley EG, Shields MB. Transscleral diode laser cyclophotocoagulation for refractory pediatric glaucomas. J Pediatr Ophthalmol Strabismus. 1997 Jul-Aug;34(4):235-9. doi: 10.3928/0191-3913-19970701-11[]
  318. Kirwan JF, Shah P, Khaw PT. Diode laser cyclophotocoagulation: role in the management of refractory pediatric glaucomas. Ophthalmology. 2002 Feb;109(2):316-23. doi: 10.1016/s0161-6420(01)00898-3[]
  319. Autrata R, Rehurek J. Long-term results of transscleral cyclophotocoagulation in refractory pediatric glaucoma patients. Ophthalmologica. 2003 Nov-Dec;217(6):393-400. doi: 10.1159/000073068[]
  320. Sood S, Beck AD. Cyclophotocoagulation versus sequential tube shunt as a secondary intervention following primary tube shunt failure in pediatric glaucoma. J AAPOS. 2009 Aug;13(4):379-83. doi: 10.1016/j.jaapos.2009.05.006[]
  321. Neely DE, Plager DA. Endocyclophotocoagulation for management of difficult pediatric glaucomas. J AAPOS. 2001 Aug;5(4):221-9. doi: 10.1067/mpa.2001.116868[]
  322. Glaser TS, Mulvihill MS, Freedman SF. Endoscopic cyclophotocoagulation (ECP) for childhood glaucoma: a large single-center cohort experience. J AAPOS. 2019 Apr;23(2):84.e1-84.e7. doi: 10.1016/j.jaapos.2018.10.014[]
  323. Christy J, Jain N, Gurnani B, Kaur K. Twinkling Eye -A Rare Presentation in Neovascular Glaucoma. J Glaucoma. 2019 May 23. doi: 10.1097/IJG.0000000000001287[]
  324. Gurnani B, Kaur K, Sekaran S. First case of coloboma, lens neovascularization, traumatic cataract, and retinal detachment in a young Asian female. Clin Case Rep. 2021 Aug 30;9(9):e04743. doi: 10.1002/ccr3.4743[]
  325. Gurnani B, Kaur K, Gireesh P. A rare presentation of anterior dislocation of calcified capsular bag in a spontaneously absorbed cataractous eye. Oman J Ophthalmol. 2021 Jun 28;14(2):120-121. doi: 10.4103/ojo.OJO_65_2019[]
  326. Gurnani B, Kaur K, Gireesh P. Rare Coexistence of Bilateral Congenital Sutural and Cortical Blue Dot Cataracts. J Pediatr Ophthalmol Strabismus. 2020 Jan 1;57(1):68. doi: 10.3928/01913913-20191011-01[]
  327. Kaur K, Gurnani B, Kannusamy V, Yadalla D. A tale of orbital cellulitis and retinopathy of prematurity in an infant: First case report. Eur J Ophthalmol. 2022 Nov;32(6):NP20-NP23. doi: 10.1177/11206721211026098[]
  328. Baig NB, Lin AA, Freedman SF. Ultrasound evaluation of glaucoma drainage devices in children. J AAPOS. 2015 Jun;19(3):281-4. doi: 10.1016/j.jaapos.2015.02.001[]
  329. Neustein RF, Bruce BB, Beck AD. Primary Congenital Glaucoma Versus Glaucoma Following Congenital Cataract Surgery: Comparative Clinical Features and Long-term Outcomes. Am J Ophthalmol. 2016 Oct;170:214-222. doi: 10.1016/j.ajo.2016.08.012[]
  330. Biglan AW, Hiles DA. The visual results following infantile glaucoma surgery. J Pediatr Ophthalmol Strabismus. 1979 Nov-Dec;16(6):377-81. doi: 10.3928/0191-3913-19791101-10[]
  331. de Silva DJ, Khaw PT, Brookes JL. Long-term outcome of primary congenital glaucoma. J AAPOS. 2011 Apr;15(2):148-52. doi: 10.1016/j.jaapos.2010.11.025[]
  332. Shaffer RN. Prognosis of goniotomy in primary infantile glaucoma (trabeculodysgenesis). Trans Am Ophthalmol Soc. 1982;80:321-5. https://pmc.ncbi.nlm.nih.gov/articles/instance/1312272/pdf/taos00019-0346.pdf[]
  333. Kantipuly A, Pillai MR, Shroff S, Khatiwala R, Raman GV, Krishnadas SR, Lee Robin A, Ehrlich JR. Caregiver Burden in Primary Congenital Glaucoma. Am J Ophthalmol. 2019 Sep;205:106-114. doi: 10.1016/j.ajo.2019.05.003[]
  334. McLaughlin DE, Semrov A, Munshi H, Patel AJ, Rahi J, Grajewski AL; Balkan CGRN Study Group. The impact of childhood glaucoma on psychosocial functioning and quality of life: a review of the literature. Eye (Lond). 2023 Oct;37(15):3157-3173. doi: 10.1038/s41433-023-02492-1[]
  335. Mahabadi N, Zeppieri M, Tripathy K. Open Angle Glaucoma. [Updated 2024 Mar 7]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK441887[][][][][][][][][][][][][][][][][][][]
  336. Primary Open-Angle Glaucoma. https://eyewiki.org/Primary_Open-Angle_Glaucoma[][][][][][][][][][][][][][][][][][][]
  337. Veeraraghavan N. Adult Eye Conditions: Primary Open-Angle Glaucoma and Cataract. FP Essent. 2022 Aug;519:19-23.[][]
  338. Types of Glaucoma. https://www.nei.nih.gov/learn-about-eye-health/eye-conditions-and-diseases/glaucoma/types-glaucoma[]
  339. Yanoff M, Duker JS. Ophthalmology. 5th ed. Elsevier; 2018.[]
  340. Tham YC, Li X, Wong TY, Quigley HA, Aung T, Cheng CY. Global prevalence of glaucoma and projections of glaucoma burden through 2040: a systematic review and meta-analysis. Ophthalmology. 2014 Nov;121(11):2081-90. doi: 10.1016/j.ophtha.2014.05.013[]
  341. Glaucoma in the African American and Hispanic Communities: What We Know. https://www.brightfocus.org/resource/glaucoma-in-the-african-american-and-hispanic-communities-what-we-know/[][]
  342. Kang JM, Tanna AP. Glaucoma. Med Clin North Am. 2021 May;105(3):493-510. doi: 10.1016/j.mcna.2021.01.004[][][][]
  343. Stein JD, Khawaja AP, Weizer JS. Glaucoma in Adults-Screening, Diagnosis, and Management: A Review. JAMA. 2021 Jan 12;325(2):164-174. doi: 10.1001/jama.2020.21899[]
  344. About Glaucoma. https://www.cdc.gov/vision-health/about-eye-disorders/glaucoma.html[]
  345. Skaat A, De Moraes CG, Bowd C, Sample PA, Girkin CA, Medeiros FA, Ritch R, Weinreb RN, Zangwill LM, Liebmann JM; Diagnostic Innovations in Glaucoma Study and African Descent and Glaucoma Evaluation Study Groups. African Descent and Glaucoma Evaluation Study (ADAGES): Racial Differences in Optic Disc Hemorrhage and Beta-Zone Parapapillary Atrophy. Ophthalmology. 2016 Jul;123(7):1476-83. doi: 10.1016/j.ophtha.2016.03.025[]
  346. Allison K, Patel DG, Greene L. Racial and Ethnic Disparities in Primary Open-Angle Glaucoma Clinical Trials: A Systematic Review and Meta-analysis. JAMA Netw Open. 2021 May 3;4(5):e218348. doi: 10.1001/jamanetworkopen.2021.8348[]
  347. Khazaeni B, Zeppieri M, Khazaeni L. Acute Angle-Closure Glaucoma. [Updated 2023 Nov 26]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK430857[]
  348. MYOC gene. https://medlineplus.gov/genetics/gene/myoc[][][]
  349. Skarie J.M., Link B.A. The primary open-angle glaucoma gene WDR36 functions in ribosomal RNA processing and interacts with the p53 stress-response pathway. Hum. Mol. Genet. 2008;17:2474–2485. doi: 10.1093/hmg/ddn147[]
  350. Dores G.M., Curtis R.E., Toro J.R., Devesa S.S., Fraumeni J.F., Jr. Incidence of cutaneous sebaceous carcinoma and risk of associated neoplasms: Insight into Muir-Torre syndrome. Cancer. 2008;113:3372–3381. doi: 10.1002/cncr.23963[]
  351. Fuse N. Genetic bases for glaucoma. Tohoku J. Exp. Med. 2010;221:1–10. doi: 10.1620/tjem.221.1[]
  352. Hauser M.A., Allingham R.R., Linkroum K., Wang J., RaRocque-Abramson K., Figueiredo D., Santiago-Turla C., del Bono E.A., Haines J.L., Pericak-Vance M.A., et al. Distribution of WDR36 DNA sequence variants in patients with primary open-angle glaucoma. Investig. Ophthalmol. Vis. Sci. 2006;47:2542–2546. doi: 10.1167/iovs.05-1476[]
  353. Fan BJ, Wang DY, Cheng CY, Ko WC, Lam SC, Pang CP. Different WDR36 mutation pattern in Chinese patients with primary open-angle glaucoma. Mol Vis. 2009;15:646-53. https://pmc.ncbi.nlm.nih.gov/articles/PMC2664842[][]
  354. Meer E, Aleman TS, Ross AG. WDR36-Associated Neurodegeneration: A Case Report Highlights Possible Mechanisms of Normal Tension Glaucoma. Genes (Basel). 2021 Oct 15;12(10):1624. doi: 10.3390/genes12101624[]
  355. Su HA, Li SY, Yang JJ, Yen YC. An Application of NGS for WDR36 Gene in Taiwanese Patients with Juvenile-Onset Open-Angle Glaucoma. Int J Med Sci. 2017 Sep 20;14(12):1251-1256. doi: 10.7150/ijms.20729[]
  356. Frezzotti P, Pescucci C, Papa FT, Iester M, Mittica V, Motolese I, Peruzzi S, Artuso R, Longo I, Mencarelli MA, Mittica P, Motolese E, Renieri A. Association between primary open-angle glaucoma (POAG) and WDR36 sequence variance in Italian families affected by POAG. Br J Ophthalmol. 2011 May;95(5):624-6. doi: 10.1136/bjo.2009.167494[]
  357. Pasutto F, Mardin CY, Michels-Rautenstrauss K, Weber BH, Sticht H, Chavarria-Soley G, Rautenstrauss B, Kruse F, Reis A. Profiling of WDR36 missense variants in German patients with glaucoma. Invest Ophthalmol Vis Sci. 2008 Jan;49(1):270-4. doi: 10.1167/iovs.07-0500[]
  358. Weisschuh N, Wolf C, Wissinger B, Gramer E. Variations in the WDR36 gene in German patients with normal tension glaucoma. Mol Vis. 2007 May 16;13:724-9. https://pmc.ncbi.nlm.nih.gov/articles/PMC2765470[]
  359. Liu K, He W, Zhao J, Zeng Y, Cheng H. Association of WDR36 polymorphisms with primary open angle glaucoma: A systematic review and meta-analysis. Medicine (Baltimore). 2017 Jun;96(26):e7291. doi: 10.1097/MD.0000000000007291[]
  360. Monemi S, Spaeth G, DaSilva A, Popinchalk S, Ilitchev E, Liebmann J, Ritch R, Héon E, Crick RP, Child A, Sarfarazi M. Identification of a novel adult-onset primary open-angle glaucoma (POAG) gene on 5q22.1. Hum Mol Genet. 2005 Mar 15;14(6):725-33. doi: 10.1093/hmg/ddi068[]
  361. CAV1 gene. https://medlineplus.gov/genetics/gene/cav1[]
  362. Thorleifsson G, Walters GB, Hewitt AW, et al. Common variants near CAV1 and CAV2 are associated with primary open-angle glaucoma. Nat Genet. 2010 Oct;42(10):906-9. doi: 10.1038/ng.661[]
  363. Borrás T. Gene expression in the trabecular meshwork and the influence of intraocular pressure. Prog Retin Eye Res. 2003 Jul;22(4):435-63. doi: 10.1016/s1350-9462(03)00018-1[]
  364. Pasquale LR, Loomis SJ, Kang JH, et al. CDKN2B-AS1 genotype-glaucoma feature correlations in primary open-angle glaucoma patients from the United States. Am J Ophthalmol. 2013 Feb;155(2):342-353.e5. doi: 10.1016/j.ajo.2012.07.023[]
  365. Nunes HF, Ananina G, Costa VP, Zanchin NIT, de Vasconcellos JPC, de Melo MB. Investigation of CAV1/CAV2 rs4236601 and CDKN2B-AS1 rs2157719 in primary open-angle glaucoma patients from Brazil. Ophthalmic Genet. 2018 Apr;39(2):194-199. doi: 10.1080/13816810.2017.1393830[]
  366. Burdon KP, Macgregor S, Hewitt AW, Sharma S, Chidlow G, Mills RA, Danoy P, Casson R, Viswanathan AC, Liu JZ, Landers J, Henders AK, Wood J, Souzeau E, Crawford A, Leo P, Wang JJ, Rochtchina E, Nyholt DR, Martin NG, Montgomery GW, Mitchell P, Brown MA, Mackey DA, Craig JE. Genome-wide association study identifies susceptibility loci for open angle glaucoma at TMCO1 and CDKN2B-AS1. Nat Genet. 2011 Jun;43(6):574-8. doi: 10.1038/ng.824[]
  367. Shim MS, Kim KY, Noh M, Ko JY, Ahn S, An MA, Iwata T, Perkins GA, Weinreb RN, Ju WK. Optineurin E50K triggers BDNF deficiency-mediated mitochondrial dysfunction in retinal photoreceptor cell line. Biochem Biophys Res Commun. 2018 Sep 18;503(4):2690-2697. doi: 10.1016/j.bbrc.2018.08.025[]
  368. Ma L, Zeng Y, Wei J, Yang D, Ding G, Liu J, Shang J, Kang Y, Ji X. Knockdown of LOXL1 inhibits TGF-β1-induced proliferation and fibrogenesis of hepatic stellate cells by inhibition of Smad2/3 phosphorylation. Biomed Pharmacother. 2018 Nov;107:1728-1735. doi: 10.1016/j.biopha.2018.08.156[]
  369. Berner D, Zenkel M, Pasutto F, Hoja U, Liravi P, Gusek-Schneider GC, Kruse FE, Schödel J, Reis A, Schlötzer-Schrehardt U. Posttranscriptional Regulation of LOXL1 Expression Via Alternative Splicing and Nonsense-Mediated mRNA Decay as an Adaptive Stress Response. Invest Ophthalmol Vis Sci. 2017 Nov 1;58(13):5930-5940. doi: 10.1167/iovs.17-22963[]
  370. Guven Yilmaz S, Palamar M, Onay H, Ilim O, Aykut A, Ozkinay FF, Yagci A. Association of Lysyl Oxidase-like 1 Gene Polymorphism in Turkish Patients With Pseudoexfoliation Syndrome and Pseudoexfoliation Glaucoma. J Glaucoma. 2017 Jul;26(7):684-685. doi: 10.1097/IJG.0000000000000672[]
  371. Sharma S, Martin S, Sykes MJ, Dave A, Hewitt AW, Burdon KP, Ronci M, Voelcker NH, Craig JE. Biological effect of LOXL1 coding variants associated with pseudoexfoliation syndrome. Exp Eye Res. 2016 May;146:212-223. doi: 10.1016/j.exer.2016.03.013[]
  372. Gao XR, Chiariglione M, Arch AJ. Whole-exome sequencing study identifies rare variants and genes associated with intraocular pressure and glaucoma. Nat Commun. 2022 Nov 30;13(1):7376. doi: 10.1038/s41467-022-35188-3[]
  373. Mabuchi F, Mabuchi N, Sakurada Y, et al. Genetic variants associated with glaucomatous visual field loss in primary open-angle glaucoma. Sci Rep. 2022 Dec 1;12(1):20744. doi: 10.1038/s41598-022-24915-x[]
  374. Zukerman R, Harris A, Oddone F, Siesky B, Verticchio Vercellin A, Ciulla TA. Glaucoma Heritability: Molecular Mechanisms of Disease. Genes (Basel). 2021 Jul 27;12(8):1135. doi: 10.3390/genes12081135[]
  375. Michels TC, Ivan O. Glaucoma: Diagnosis and Management. Am Fam Physician. 2023 Mar;107(3):253-262. https://www.aafp.org/pubs/afp/issues/2023/0300/glaucoma.html[][][][]
  376. Wolfs RC, Klaver CC, Ramrattan RS, van Duijn CM, Hofman A, de Jong PT. Genetic risk of primary open-angle glaucoma. Population-based familial aggregation study. Arch Ophthalmol. 1998 Dec;116(12):1640-5. doi: 10.1001/archopht.116.12.1640[]
  377. Birhanu G, Tegegne AS. Predictors for elevation of Intraocular Pressure (IOP) on glaucoma patients; a retrospective cohort study design. BMC Ophthalmol. 2022 Jun 7;22(1):254. doi: 10.1186/s12886-022-02431-w[][][]
  378. Ha A, Kim CY, Shim SR, Chang IB, Kim YK. Degree of Myopia and Glaucoma Risk: A Dose-Response Meta-analysis. Am J Ophthalmol. 2022 Apr;236:107-119. doi: 10.1016/j.ajo.2021.10.007[]
  379. Ikeda Y, Mori K, Maruyama Y, Ueno M, Yoshii K, Yamamoto Y, Imai K, Omi N, Sato R, Sato F, Nakano M, Hamuro J, Tashiro K, Sotozono C, Kinoshita S. Novel Vertical Cup-to-Disc Classification to Identify Normal Eyes That Maintain Non-Glaucoma Status: A 10-Year Longitudinal Study. J Glaucoma. 2023 Feb 1;32(2):127-132. doi: 10.1097/IJG.0000000000002109[]
  380. Lee EJ, Kee HJ, Han JC, Kee C. Evidence-based understanding of disc hemorrhage in glaucoma. Surv Ophthalmol. 2021 May-Jun;66(3):412-422. doi: 10.1016/j.survophthal.2020.09.001[]
  381. Zeppieri M, Brusini P, Miglior S. Corneal thickness and functional damage in patients with ocular hypertension. Eur J Ophthalmol. 2005 Mar-Apr;15(2):196-201. doi: 10.1177/112067210501500203[]
  382. Kim KE, Oh S, Baek SU, Ahn SJ, Park KH, Jeoung JW. Ocular Perfusion Pressure and the Risk of Open-Angle Glaucoma: Systematic Review and Meta-analysis. Sci Rep. 2020 Jun 22;10(1):10056. doi: 10.1038/s41598-020-66914-w[]
  383. Leske MC, Wu SY, Hennis A, Honkanen R, Nemesure B; BESs Study Group. Risk factors for incident open-angle glaucoma: the Barbados Eye Studies. Ophthalmology. 2008 Jan;115(1):85-93. doi: 10.1016/j.ophtha.2007.03.017[]
  384. Prum BE Jr, Rosenberg LF, Gedde SJ, Mansberger SL, Stein JD, Moroi SE, Herndon LW Jr, Lim MC, Williams RD. Primary Open-Angle Glaucoma Preferred Practice Pattern(®) Guidelines. Ophthalmology. 2016 Jan;123(1):P41-P111. doi: 10.1016/j.ophtha.2015.10.053. Epub 2015 Nov 12. Erratum in: Ophthalmology. 2018 Jun;125(6):949. doi: 10.1016/j.ophtha.2018.03.048[][][][]
  385. Li Y, Mitchell W, Elze T, Zebardast N. Association Between Diabetes, Diabetic Retinopathy, and Glaucoma. Curr Diab Rep. 2021 Sep 8;21(10):38. doi: 10.1007/s11892-021-01404-5[]
  386. Matsuda A, Hara T, Miyata K, Matsuo H, Murata H, Mayama C, Asaoka R. Do pattern deviation values accurately estimate glaucomatous visual field damage in eyes with glaucoma and cataract? Br J Ophthalmol. 2015 Sep;99(9):1240-4. doi: 10.1136/bjophthalmol-2014-306019[]
  387. Gramer G, Weber BH, Gramer E. Migraine and Vasospasm in Glaucoma: Age-Related Evaluation of 2027 Patients With Glaucoma or Ocular Hypertension. Invest Ophthalmol Vis Sci. 2015 Dec;56(13):7999-8007. doi: 10.1167/iovs.15-17274[]
  388. Baneke AJ, Aubry J, Viswanathan AC, Plant GT. The role of intracranial pressure in glaucoma and therapeutic implications. Eye (Lond). 2020 Jan;34(1):178-191. doi: 10.1038/s41433-019-0681-y[]
  389. Hogden K, Mikelberg F, Sodhi M, Khosrow-Khavar F, Mansournia MA, Kezouh A, Etminan M. The association between hormonal contraceptive use and glaucoma in women of reproductive age. Br J Clin Pharmacol. 2021 Dec;87(12):4780-4785. doi: 10.1111/bcp.14920[]
  390. Moreno-Montañés J, Gándara E, Gutierrez-Ruiz I, Moreno-Galarraga L, Ruiz-Canela M, Bes-Rastrollo M, Martínez-González MÁ, Fernandez-Montero A. Healthy Lifestyle Score and Incidence of Glaucoma: The Sun Project. Nutrients. 2022 Feb 12;14(4):779. doi: 10.3390/nu14040779[]
  391. Kass MA, Heuer DK, Higginbotham EJ, Johnson CA, Keltner JL, Miller JP, Parrish RK 2nd, Wilson MR, Gordon MO. The Ocular Hypertension Treatment Study: a randomized trial determines that topical ocular hypotensive medication delays or prevents the onset of primary open-angle glaucoma. Arch Ophthalmol. 2002 Jun;120(6):701-13; discussion 829-30. doi: 10.1001/archopht.120.6.701[]
  392. Mokhles P, Schouten JS, Beckers HJ, Azuara-Blanco A, Tuulonen A, Webers CA. Glaucoma blindness at the end of life. Acta Ophthalmol. 2017 Feb;95(1):10-11. doi: 10.1111/aos.12933[]
  393. Bhat KS, Reddy MV, Pai V. Correlation of retinal nerve fiber layer thickness with perimetric staging in primary open-angle glaucoma – A cross-sectional study. Oman J Ophthalmol. 2022 Mar 2;15(1):36-42. doi: 10.4103/ojo.ojo_345_20[]
  394. Kansal V, Armstrong JJ, Pintwala R, Hutnik C. Optical coherence tomography for glaucoma diagnosis: An evidence based meta-analysis. PLoS One. 2018 Jan 4;13(1):e0190621. doi: 10.1371/journal.pone.0190621[]
  395. Tripathy K, Sharma YR, Chawla R, Basu K, Vohra R, Venkatesh P. Triads in Ophthalmology: A Comprehensive Review. Semin Ophthalmol. 2017;32(2):237-250. doi: 10.3109/08820538.2015.1045150[]
  396. Sihota R, Angmo D, Ramaswamy D, Dada T. Simplifying “target” intraocular pressure for different stages of primary open-angle glaucoma and primary angle-closure glaucoma. Indian J Ophthalmol. 2018 Apr;66(4):495-505. doi: 10.4103/ijo.IJO_1130_17[]
  397. Wilson MR. Progression of visual field loss in untreated glaucoma patients and suspects in St Lucia, West Indies. Trans Am Ophthalmol Soc. 2002;100:365-410. https://pmc.ncbi.nlm.nih.gov/articles/instance/1358971/pdf/12545702.pdf[]
  398. Primary Open-Angle Glaucoma Preferred Practice Pattern® Guidelines. Prum, Bruce E. et al. Ophthalmology, Volume 123, Issue 1, P41 – P111. https://www.aaojournal.org/article/S0161-6420(15)01276-2/fulltext[][]
  399. Leung CK, Cheung CY, Li H, et al. Dyanamic analysis of light-dark changes of the anterior chamber angle with anterior segment OCT. IOVS. 2007;48:4116-22[]
  400. Alsbirk PH. Corneal diameter in Greenland Eskimos. Anthropometric and genetic studies with special reference to primary angle-closure glaucoma. Acta Ophthalmol (Copenh). 1975;53:635–646.[]
  401. Wyatt H, Ghosh J. Behaviour of an iris model and the pupil block hypothesis. Br J Ophthalmol. 1970;54:177–185.[]
  402. Quigley HA. Angle-closure glaucoma – simpler answers to complex mechanisms: LXVI Edward Jackson Memorial Lecture. Am J Ophthalmol. 2009;148(5):657-669.e1.[]
  403. Wand M, Grant WM, Simmons RJ, Hutchinson BT. Plateau iris syndrome. Trans Sect Ophthalmol Am Acad Ophthalmol Otolaryngol. 1977;83:122–130.[]
  404. He M, Foster PJ, Johnson GJ, Khaw PT. Angle-closure glaucoma in East Asian and European people. Different diseases? Eye. 2006;20:3–12.[]
  405. Lowe RF. Aetiology of the anatomical basis for primary angle-closure glaucoma. Biometrical comparisons between normal eyes and eyes with primary angle-closure glaucoma. Br J Ophthalmol 1970;54:161-9.[]
  406. Wang N, Wu H, Fan Z. Primary angle closure glaucoma in Chinese and Western populations. Chin Med J (Engl). 2002;115:1706–1715.[]
  407. Seah SK, Foster PJ, Chew PT, et al. Incidence of acute primary angle-closure glaucoma in Singapore. An island-wide survey. Arch Ophthalmol. 1997;115:1436-40.[][]
  408. Quigley HA, Broman AT. The number of people with glaucoma worldwide in 2010 and 2020. Br J Ophthalmol. 2006;90:262–267[]
  409. Lai JS, Liu DT, Tham CC, et al. Epidemiology of acute primary angle-closure glaucoma in the Hong Kong Chinese population: prospective study. Hong Kong Med J. 2001;7:118-23.[]
  410. Alsbirk PH. Anatomical risk factors in primary angle-closure glaucoma. A ten year follow up survey based on limbal and axial anterior chamber depths in a high risk population. Int Ophthalmol. 1992;16:265-72.[]
  411. Sihota R, Sood A, Gupta V et al: A prospective longterm study of primary chronic angle closure glaucoma. Acta Ophthalmol Scand 82:209–213, 2004[][]
  412. Lowe RF. Aetiology of the anatomical basis for primary angle-closure glaucoma. Biometrical comparisons between normal eyes and eyes with primary angle-closure glaucoma. Br J Ophthalmol. 1970;54:161–169.[]
  413. Palmberg P. Shedding light on gonioscopy. Arch Ophthalmol. 2007;125:1417–1418.[]
  414. Foster PJ, Aung T, et al. Defining “occludable” angles in population surverys: drainage anger width, peripheral anterior synechiae, and glaucomatous optic neuropathy in east Asian people. Br J Ophthalmol. 2004;88(4):486-90.[]
  415. Wilensky JT, Kaufman PL, Frohlichstein D et al. Follow-up of angle-closure glaucoma suspects. Am J Ophthalmol. 1993;115:338–346.[][]
  416. Wang B, Congdon NG, et al. Dark room provocative test and extent of angle closure: an anterior segment OCT study. J Glaucoma. 2009 Jul 9 (Epub ahead of print).[]
  417. Garcia JP Jr, Rosen RB. Anterior segment imaging: optical coherence tomography versus ultrasound biomicroscopy. Ophthalmic Surg Lasers Imaging. 2008;39(6):476-84.[][]
  418. Aung T, Wong HT, Yip CC et al. Comparison of the intraocular pressure-lowering effect of latanoprost and timolol in patients with chronic angle closure glaucoma: A preliminary study. Ophthalmology. 2000;107:1178–1183[]
  419. Hung PT, Hsieh JW, Chen YF, Wei T. Efficacy of latanoprost as an adjunct to medical therapy for residual angle-closure glaucoma after iridectomy. J Ocul Pharmacol Ther. 2000;16:43–47[]
  420. Aung T, Chew PT. Review of recent advancements in the understanding of primary angle-closure glaucoma. Curr Opin Ophthalmol. 2002;13:89–93.[][]
  421. Edwards RS. Behaviour of the fellow eye in acute angle-closure glaucoma. Br J Ophthalmol. 1982;66:576–579.[][][]
  422. Lowe RF. Primary angle-closure glaucoma. Prevention and early treatment. Isr J Med Sci. 1972;8:1362–1365.[]
  423. Saw SM, Gazzard G, Friedman DS. Interventions for angle-closure glaucoma: an evidence-based update. Ophthalmology. 2003;11:1869-78.[]
  424. Alsagoff Z, Aung T, Ang LP, Chew PT. Long-term clinical course of primary angle-closure glaucoma in an Asian population. Ophthalmology. 2000;107:2300–2304.[]
  425. Ang LP, Aung T, Chew PT. Acute primary angle closure in an Asian population: long-term outcome of the fellow eye after prophylactic laser peripheral iridotomy. Ophthalmology. 2000;107:2092-6.[][]
  426. Lim AS, Tan A, Chew P et al. Laser iridoplasty in the treatment of severe acute angle closure glaucoma. Int Ophthalmol. 1993;17:33–36.[]
  427. Sun X, Liang YB, et al. Laser Peripheral Iridotomy with and without Iridoplasty for Primary Angle-Closure Glaucoma: 1-Year Results of a Randomized Pilot Study. Am J Ophthalmol. 2010 May 14. Epub ahead of print.[]
  428. Ritch R, Tham CC, Lam DS. Argon laser peripheral iridoplasty (ALPI): An update. Surv Ophthalmol. 2007;52:279–288.[]
  429. Lam DS, Leung DY, et al. Randomized trial of early phacoemulsification versus peripheral iridotomy to prevent intraocular pressure rise after acute primary angle closure. Ophthalmology. 2008;115(7):1134-40.[]
  430. Imaizumi M, Takaki Y, Yamashita H. Phacoemulsification and intraocular lens implantation for acute angle closure not treated or previously treated by laser iridotomy. J Cataract Refract Surg. 2006;32:85–90.[]
  431. Teekhasaenee C, Ritch R. Combined phacoemulsification and goniosynechialysis for uncontrolled chronic angle-closure glaucoma after acute angle-closure glaucoma. Ophthalmology. 1999;106:669–674.[]
  432. Salmon JF. The role of trabeculectomy in the treatment of advanced chronic angle closure glaucoma. J Glaucoma. 1993;2:285–190.[]
  433. Lai JS, Tham CC, Lam DS. Incisional surgery for angle closure glaucoma. Semin Ophthalmol. 2002;17:92–99.[]
  434. Trope GE, Pavlin CJ, Bau A et al. Malignant glaucoma. Clinical and ultrasound biomicroscopic features. Ophthalmology. 1994;101:1030–1035.[]
  435. Tham CC, Leung DY, et al. Effects of phacoemulsification versus combined phaco-trabeculectomy on drainage angle status in primary angle closure glaucoma (PACG). J Glaucoma. 2010;19(2):119-23.[]
  436. Tsai HY, Liu CJ, Cheng CY. Combined trabeculectomy and cataract extraction versus trabeculectomy alone in pimary angle-closure glaucoma. Br J Ophthalmol. 2009;93(7):943-8.[]
  437. Tham CC, Kwong YY, et al. Phacoemulsification versus combined phacotrabeculectomy in medically uncontrolled chronic angle closure glaucoma with cataracts. Ophthalmology. 2009;116(4):725-31, 731.e1-3.[]
  438. Tham CC, Kwong YY, et al. Phacoemulsification vs phacotrabeculectomy in chronic angle-closure glaucoma with cataract complications. Arch Ophthalmol. 2010;128(3):303-11.[]
  439. Deokule SP, Molteno AC, Bevin TH, Herbison P. Long-term results of Molteno implant insertion in cases of chronic angle closure glaucoma. Clin Experiment Ophthalmol. 2007;35:514–519.[]
  440. Spaeth GL. Gonioscopy: Uses old and new. The inheritance of occludable angles. Ophthalmology. 1978;85:222–232.[]
  441. Friedman DS, Chew PT, Gazzard G et al. Long-term outcomes in fellow eyes after acute primary angle closure in the contralateral eye. Ophthalmology. 2006;113:1087–1091.[]
  442. Aung T, Looi AL, Chew PT. The visual field following acute primary angle closure. Acta Ophthalmol Scand. 2001;79:298–300.[][][]
  443. Shen SY, Baskaran M, Fong AC et al. Changes in the optic disc after acute primary angle closure. Ophthalmology. 2006;113:924–929.[]
  444. Wong JS, Chew PT, Alsagoff Z, Poh K. Clinical course and outcome of primary acute angle-closure glaucoma in Singapore. Singapore Med J. 1997;38:16–18.[][]
  445. Saraux H, Offret H. Long-term study of patients with iridectomy for angle-closure glaucoma. Glaucoma. 1979;1:149–151.[]
  446. Nolan WP, Foster PJ, Devereux JG et al. YAG laser iridotomy treatment for primary angle closure in East Asian eyes. Br J Ophthalmol. 2000;84:1255–1259[]
  447. Aung T, Friedman DS, Chew PT et al. Long-term outcomes in Asians after acute primary angle closure. Ophthalmology. 2004;111:1464–1469.[]
  448. Dhillon B, Chew PT, Lim AS. Field loss in primary angle closure glaucoma. Asia Pac J Ophthalmol. 1990;2:85–87.[]
  449. Ang LP, Aung T, Chua WH et al. Visual field loss from primary angle-closure glaucoma: A comparative study of symptomatic and asymptomatic disease. Ophthalmology. 2004;111:1636–1540.[]
  450. Rosman M, Aung T, Ang LP et al. Chronic angle-closure with glaucomatous damage: Long-term clinical course in a North American population and comparison with an Asian population. Ophthalmology. 2002;109:2227–2231.[]
  451. See JL, Chew PT. Glaucoma in Singapore. J Glaucoma. 2004;3:417–420.[]
  452. Quigley HA, McKinnon SJ, Zack DJ, Pease ME, Kerrigan-Baumrind LA, Kerrigan DF, Mitchell RS. Retrograde axonal transport of BDNF in retinal ganglion cells is blocked by acute IOP elevation in rats. Invest Ophthalmol Vis Sci. 2000 Oct;41(11):3460-6. https://iovs.arvojournals.org/article.aspx?articleid=2123432[]
  453. Berson DM. Phototransduction in ganglion-cell photoreceptors. Pflugers Arch. 2007 Aug;454(5):849-55. doi: 10.1007/s00424-007-0242-2[]
  454. Hattar S, Kumar M, Park A, Tong P, Tung J, Yau KW, Berson DM. Central projections of melanopsin-expressing retinal ganglion cells in the mouse. J Comp Neurol. 2006 Jul 20;497(3):326-49. doi: 10.1002/cne.20970[]
  455. Heijl A, Bengtsson B, Hyman L, Leske MC; Early Manifest Glaucoma Trial Group. Natural history of open-angle glaucoma. Ophthalmology. 2009 Dec;116(12):2271-6. doi: 10.1016/j.ophtha.2009.06.042[]
  456. Reina-Torres E, Baptiste TMG, Overby DR. Segmental outflow dynamics in the trabecular meshwork of living mice. Exp Eye Res. 2022 Dec;225:109285. doi: 10.1016/j.exer.2022.109285[]
  457. Dielemans I, Vingerling JR, Wolfs RC, Hofman A, Grobbee DE, de Jong PT. The prevalence of primary open-angle glaucoma in a population-based study in The Netherlands. The Rotterdam Study. Ophthalmology. 1994 Nov;101(11):1851-5. doi: 10.1016/s0161-6420(94)31090-6[]
  458. Toris CB, Koepsell SA, Yablonski ME, Camras CB. Aqueous humor dynamics in ocular hypertensive patients. J Glaucoma. Jun 2002;11(3):253-8. doi:10.1097/00061198-200206000-00015[][][]
  459. Chapter 3. The Glaucoma Eye. https://eyerounds.org/books/glaucoma_guide/chapter3.html#gsc.tab=0[]
  460. Harizman N, Oliveira C, Chiang A, et al. The ISNT Rule and Differentiation of Normal From Glaucomatous Eyes. Arch Ophthalmol. 2006;124(11):1579–1583. doi:10.1001/archopht.124.11.1579[][][][]
  461. Maupin E, Baudin F, Arnould L, Seydou A, Binquet C, Bron AM, Creuzot-Garcher CP. Accuracy of the ISNT rule and its variants for differentiating glaucomatous from normal eyes in a population-based study. Br J Ophthalmol. 2020 Oct;104(10):1412-1417. doi: 10.1136/bjophthalmol-2019-315554[]
  462. Hollands H, Johnson D, Hollands S, Simel DL, Jinapriya D, Sharma S. Do findings on routine examination identify patients at risk for primary open-angle glaucoma? The rational clinical examination systematic review. JAMA. 2013 May 15;309(19):2035-42. doi: 10.1001/jama.2013.5099[]
  463. Susanna R Jr, Vessani RM. Staging glaucoma patient: why and how? Open Ophthalmol J. 2009 Sep 17;3:59-64. doi: 10.2174/1874364100903020059[][]
  464. Kalyani VK, Bharucha KM, Goyal N, Deshpande MM. Comparison of diagnostic ability of standard automated perimetry, short wavelength automated perimetry, retinal nerve fiber layer thickness analysis and ganglion cell layer thickness analysis in early detection of glaucoma. Indian J Ophthalmol. 2021 May;69(5):1108-1112. doi: 10.4103/ijo.IJO_2409_20[]
  465. Zeppieri M, Brusini P, Parisi L, Johnson CA, Sampaolesi R, Salvetat ML. Pulsar perimetry in the diagnosis of early glaucoma. Am J Ophthalmol. 2010 Jan;149(1):102-12. doi: 10.1016/j.ajo.2009.07.020[]
  466. Salvetat ML, Zeppieri M, Parisi L, Johnson CA, Sampaolesi R, Brusini P. Learning effect and test-retest variability of pulsar perimetry. J Glaucoma. 2013 Mar;22(3):230-7. doi: 10.1097/IJG.0b013e318237bfe7[]
  467. Salvetat ML, Zeppieri M, Parisi L, Brusini P. Rarebit perimetry in normal subjects: test-retest variability, learning effect, normative range, influence of optical defocus, and cataract extraction. Invest Ophthalmol Vis Sci. 2007 Nov;48(11):5320-31. doi: 10.1167/iovs.06-1495[]
  468. Brusini P, Salvetat ML, Parisi L, Zeppieri M. Probing glaucoma visual damage by rarebit perimetry. Br J Ophthalmol. 2005 Feb;89(2):180-4. doi: 10.1136/bjo.2003.041178[]
  469. Brusini P, Salvetat ML, Zeppieri M, Tosoni C, Parisi L, Felletti M. Visual field testing with the new Humphrey Matrix: a comparison between the FDT N-30 and Matrix N-30-F tests. Acta Ophthalmol Scand. 2006 Jun;84(3):351-6. doi: 10.1111/j.1600-0420.2006.00637.x[]
  470. Mastropasqua L, Brusini P, Carpineto P, Ciancaglini M, Di Antonio L, Zeppieri MW, Parisi L. Humphrey matrix frequency doubling technology perimetry and optical coherence tomography measurement of the retinal nerve fiber layer thickness in both normal and ocular hypertensive subjects. J Glaucoma. 2006 Aug;15(4):328-35. doi: 10.1097/01.ijg.0000212230.65545.d3[]
  471. Harwerth RS, Quigley HA. Visual field defects and retinal ganglion cell losses in patients with glaucoma. Arch Ophthalmol. 2006 Jun;124(6):853-9. doi: 10.1001/archopht.124.6.853[]
  472. Harwerth RS, Carter-Dawson L, Shen F, Smith EL 3rd, Crawford ML. Ganglion cell losses underlying visual field defects from experimental glaucoma. Invest Ophthalmol Vis Sci. 1999 Sep;40(10):2242-50.[]
  473. Shiga Y, Aizawa N, Tsuda S, Yokoyama Y, Omodaka K, Kunikata H, Yasui T, Kato K, Kurashima H, Miyamoto E, Hashimoto M, Nakazawa T. Preperimetric Glaucoma Prospective Study (PPGPS): Predicting Visual Field Progression With Basal Optic Nerve Head Blood Flow in Normotensive PPG Eyes. Transl Vis Sci Technol. 2018 Jan 23;7(1):11. doi: 10.1167/tvst.7.1.11[]
  474. Brusini P, Salvetat ML, Zeppieri M. How to Measure Intraocular Pressure: An Updated Review of Various Tonometers. J Clin Med. 2021 Aug 27;10(17):3860. doi: 10.3390/jcm10173860[]
  475. Bader J, Zeppieri M, Havens SJ. Tonometry. [Updated 2023 Dec 12]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK493225[]
  476. Zeppieri M, Gurnani B. Applanation Tonometry. [Updated 2023 Jun 11]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK582132[]
  477. Campbell P, Edgar DF, Shah R. Inter-optometrist variability of IOP measurement for modern tonometers and their agreement with Goldmann Applanation Tonometry. Clin Exp Optom. 2021 Jul;104(5):602-610. doi: 10.1080/08164622.2021.1878831[]
  478. Biswas S, Biswas P. Relationship between Diurnal Variation in Intraocular Pressure and Central Corneal Power. Optom Vis Sci. 2023 Jan 1;100(1):96-104. doi: 10.1097/OPX.0000000000001974[]
  479. Belovay GW, Goldberg I. The thick and thin of the central corneal thickness in glaucoma. Eye (Lond). 2018 May;32(5):915-923. doi: 10.1038/s41433-018-0033-3[]
  480. Foster PJ, Buhrmann R, Quigley HA, Johnson GJ. The definition and classification of glaucoma in prevalence surveys. Br J Ophthalmol. 2002 Feb;86(2):238-42. doi: 10.1136/bjo.86.2.238[]
  481. Gil P, Pires J, Matos R, Cardoso MS, Lopes N, Matias J, Mariano M. Corneal Elevation Topography in Primary Open Angle Glaucoma. J Glaucoma. 2017 Feb;26(2):e41-e45. doi: 10.1097/IJG.0000000000000535[]
  482. Brusini P, Zeppieri M, Tosoni C, Parisi L, Salvetat ML. Optic disc damage staging system. J Glaucoma. 2010 Sep;19(7):442-9. doi: 10.1097/IJG.0b013e3181ca7303[]
  483. Spaeth GL, Shields MB. The stages of glaucoma. Am J Ophthalmol. 2006 Jan;141(1):147-8. doi: 10.1016/j.ajo.2005.08.026[]
  484. Brusini P. Estimating glaucomatous anatomical damage by computerized automated perimetry. Acta Ophthalmol Scand Suppl. 1997;(224):28-9. doi: 10.1111/j.1600-0420.1997.tb00461.x[]
  485. Spaeth GL, Lopes JF, Junk AK, Grigorian AP, Henderer J. Systems for staging the amount of optic nerve damage in glaucoma: a critical review and new material. Surv Ophthalmol. 2006 Jul-Aug;51(4):293-315. doi: 10.1016/j.survophthal.2006.04.008[]
  486. Brusini P. OCT Glaucoma Staging System: a new method for retinal nerve fiber layer damage classification using spectral-domain OCT. Eye (Lond). 2018 Jan;32(1):113-119. doi: 10.1038/eye.2017.159[]
  487. Ng M, Sample PA, Pascual JP, Zangwill LM, Girkin CA, Liebmann JM, Weinreb RN, Racette L. Comparison of visual field severity classification systems for glaucoma. J Glaucoma. 2012 Oct-Nov;21(8):551-61. doi: 10.1097/IJG.0b013e31821dac66[]
  488. Medical Management of Glaucoma. https://morancore.utah.edu/section-10-glaucoma/medical-management-of-glaucoma[]
Health Jade