You are on page 1of 21

Drugs Aging 2005; 22 (1): 1-21

THERAPY IN PRACTICE 1170-229X/05/0001-0001/$34.95/0

© 2005 Adis Data Information BV. All rights reserved.

Management of Glaucoma: Focus on


Pharmacological Therapy
Robert E. Marquis and Jess T. Whitson
Department of Ophthalmology, The University of Texas Southwestern Medical Center at Dallas,
Dallas, Texas, USA

Contents
Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
1. Physiology of Aqueous Humour and Intraocular Pressure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2. Medical Treatment of Glaucoma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3. Cholinergics (Acetylcholine Receptor Agonists) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3.1 Direct-Acting Cholinergics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.1.1 Adverse Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.2 Indirect-Acting Cholinergics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.3 Dual-Mechanism Cholinergics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
4. Adrenoceptor Agonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
4.1 Nonselective Adrenoceptor Agonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
4.1.1 Adverse Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
4.2 Selective α2-Adrenoceptor Agonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.2.1 Adverse Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
5. Carbonic Anhydrase Inhibitors (CAIs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5.1 Systemic CAIs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5.1.1 Adverse Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5.2 Topical CAIs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5.2.1 Adverse Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
6. β-Adrenoceptor Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
6.1 Nonselective β-Adrenoceptor Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
6.1.1 Adverse Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
6.2 Selective β-Adrenoceptor Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
6.2.1 Adverse Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
7. Prostaglandin Analogues . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
8. Combination Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
9. Other Treatment Options . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
9.1 Laser Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
9.2 Surgical Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
10. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17

Abstract Glaucoma represents a major cause of vision loss throughout the world.
Primary open-angle glaucoma, the most common form of glaucoma, is a chronic,
progressive disease often, though not always, accompanied by elevated intraocu-
lar pressure (IOP). In this disorder, retinal ganglion cell loss and excavation of the
optic nerve head produce characteristic peripheral visual field deficits. Patients
with normal-tension glaucoma present with typical visual field and optic nerve
head changes, without a documented history of elevated IOP. A variety of
secondary causes, such as pigment dispersion syndrome and ocular trauma, can
result in glaucoma as well. Treatment of all forms of glaucoma consists of
2 Marquis & Whitson

reducing IOP. With proper treatment, progression of this disease can often be
delayed or prevented.
Treatment options for glaucoma include medications, laser therapy and inci-
sional surgery. Laser techniques for the reduction of IOP include argon laser
trabeculoplasty and selective laser trabeculoplasty. Both techniques work by
increasing outflow of aqueous humour through the trabecular meshwork. Surgical
options for glaucoma treatment include trabeculectomy, glaucoma drainage tube
implantation and ciliary body cyclodestruction. While each of these types of
procedures is effective at lowering IOP, therapy usually begins with medications.
Medications lower IOP either by reducing the production or by increasing the rate
of outflow of aqueous humour within the eye.
Currently, there are five major classes of drugs used for the treatment of
glaucoma: (i) cholinergics (acetylcholine receptor agonists); (ii) adrenoceptor
agonists; (iii) carbonic anhydrase inhibitors (CAIs); (iv) β-adrenoceptor antago-
nists; and (v) prostaglandin analogues (PGAs). Treatment typically begins with
the selection of an agent for IOP reduction. Although β-adrenoceptor antagonists
are still commonly used by many clinicians, the PGAs are playing an increasingly
important role in the first-line therapy of glaucoma. Adjunctive agents, such as
α-adrenoceptor agonists and CAIs are often effective at providing additional
reduction in IOP for patients not controlled on monotherapy. As with any chronic
disease, effective treatment depends on minimising the adverse effects of therapy
and maximising patient compliance. The introduction of a variety of well tolerated
and potent medications over the past few years now allows the clinician to choose
a treatment regimen on an individual patient basis and thereby treat this disorder
more effectively.

Glaucoma is a heterogeneous family of ocular which there is a characteristic acquired loss of reti-
disorders that share common characteristics includ- nal ganglion cells and atrophy of the optic nerve.[6]
ing excavation of the optic nerve head and peripher- Risk factors for the development of POAG include
al visual field loss. Retinal ganglion cell death, elevated intraocular pressure (IOP), family history
which is thought to occur through apoptosis,[1] re- of the disease, advanced age and African heritage
sults in progressive optic nerve dysfunction and (table I). While elevated IOP is considered an im-
visual impairment. According to the WHO, portant risk factor for POAG, it is not essential for
glaucoma accounts for 13.5% of global blindness
(behind cataracts and trachoma that account for Table I. Risk factors for the development and progression of
41.8% and 15.5% of global blindness, respective- glaucoma
ly[2]). In the US, glaucoma is the second leading Elevated IOP
cause of blindness overall and the leading cause Diurnal fluctuation of IOP
among African-Americans and Hispanics.[3,4] Typi- Family history of glaucoma
cally, glaucoma is asymptomatic until late in its Race (African heritage, Hispanics)
course, making early detection difficult. It is esti- Advanced age
mated that as many as one-half of those with Other
glaucoma in developed countries may be unaware increased optic nerve cup-to-disc ratio
that they have the disease.[5] compromised ocular blood flow
Primary open-angle glaucoma (POAG) is the diabetes mellitus
most common form of glaucoma throughout the myopia
world accounting for about two-thirds of cases.[2] It corneal thickness (thin corneas – greater risk)
IOP = intraocular pressure.
is defined as a multifactorial optic neuropathy in

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
Pharmacological Therapy of Glaucoma 3

its diagnosis. Visual loss and progressive optic neu- often difficult clinically to distinguish corticoste-
ropathy can occur in patients who do not demon- roid-induced glaucoma from inflammatory glauco-
strate elevations in IOP.[7] Patients who develop ma.
typical optic nerve head and visual field changes in Ocular trauma may lead to glaucoma by a variety
the absence of documented high IOP are said to have of mechanisms. In the acute post-traumatic period,
normal-tension glaucoma (NTG). NTG is believed
markedly elevated IOP may result from hyphaema
to account for about 30% of glaucoma cases in
(anterior chamber haemorrhage), inflammation and
Western countries[8] and over two-thirds of cases in
cellular debris that clog the TM. Normal red blood
Japan.[9] Some investigators believe that NTG may
represent a variant of POAG,[10,11] whereas others cells (RBCs) may filter through the TM readily in
feel that the mechanisms of the two disorders are most cases; however, if RBCs denature and swell,
different.[12,13] they may become lodged in the TM and cause an
elevation in IOP. Blunt anterior segment trauma
Ocular hypertensive subjects are those who
may also directly damage the TM and result in
demonstrate consistently elevated IOP without any
angle-recession glaucoma. Finally, inflammation af-
obvious cause and have normal visual fields and
optic discs. This condition is frequently encountered ter trauma may cause scar-like adhesions between
by clinicians and may occur as much as ten times Table II. The secondary glaucomas
more often than POAG.[14] Patients with ocular
Open-angle
hypertension are at risk for developing POAG as a
Pigmentary (associated with pigment dispersion syndrome)
result of their elevated IOP. Recently, the OHTS
Pseudoexfoliation
(Ocular Hypertension Treatment Study) has demon-
Corticosteroid-induced
strated that lowering IOP with medical therapy in Uveitic (e.g. HLA-B27+, arthritic, sarcoidosis, idiopathic)
patients with ocular hypertension can significantly Haemolytic/ghost cell (after vitreous haemorrhage)
reduce the risk of developing POAG.[15] Elevated episcleral venous pressure (e.g. carotid-cavernous
While POAG accounts for a majority of cases of fistula, Sturge-Weber syndrome)
glaucoma in the US, several other prevalent forms of Post-traumatic
the disease exist as well (table II). Secondary causes Acute hyphaema
of open-angle glaucoma (OAG) result from either an Lens dislocation into anterior chamber
increased resistance to aqueous humour flow at the Haemolytic/ghost cell
trabecular meshwork (TM) or from elevated epis- Angle recession

cleral venous pressure. Pigmentary glaucoma is Lens-associated


Phacolytic (leakage of protein from hypermature cataract)
associated with pigment dispersion syndrome which
Lens particle (after cataract surgery or penetrating trauma)
occurs when iris pigment accumulates in the anterior
Phacoantigenic (formerly phacoanaphylatic)
chamber and deposits on the TM and in some cases
Other OAG associated with uveitis
the corneal endothelium. The pigment is liberated
Posner-Schlossman syndrome (glaucomatocyclitic crisis)
from the posterior iris surface as it makes contact
Fuchs’ heterochromic iridocyclitis
with the zonular fibres that support the lens.[16]
Postsurgical (e.g. after corneal transplant)
Another form of secondary OAG is seen in Closed-angle
pseudoexfoliation syndrome. Deposition of a fibril- Plateau iris syndrome
lar material on anterior segment structures, includ- Phacomorphic (swollen or large lens pushing iris forward)
ing the TM, may result in increased IOP and Uveal effusion or ciliary body anterior rotation
glaucoma. A third form of secondary OAG occurs in Posterior segment tumour
some patients receiving corticosteroid therapy either Neovascularisation of the angle (e.g. with retinal ischaemia)
topically or (less frequently) systemically. Patients Peripheral anterior synechiae (e.g. with chronic uveitis)
with intraocular inflammation are typically treated ICE syndromes (essential iris atrophy, Chandler syndrome and
with corticosteroids. Because intraocular inflamma- Cogan-Reese syndrome)
tion (uveitis) itself may cause elevated IOP, either HLA = human leukocyte antigen; ICE = iridocorneal endothelial;
OAG = open-angle glaucoma.
through blockage or inflammation of the TM, it is

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
4 Marquis & Whitson

the peripheral iris and cornea, which can impede 1. Physiology of Aqueous Humour and
egress of aqueous humour by blocking outflow Intraocular Pressure
through the TM. If these adhesions, termed periph- IOP is determined by the balance that exists
eral anterior synechiae (PAS), are extensive, angle- between the rate of aqueous humour production
closure glaucoma may result. (inflow) and the rate of fluid exit from the eye
The angle-closure glaucomas are a group of con- (outflow). Aqueous humour is produced by the cilia-
ditions caused by several mechanisms with the com- ry body and leaves the eye through either the TM
mon anatomical result of peripheral iridocorneal (conventional) or uveoscleral (unconventional)
apposition and TM blockage (table II). Hyperopic pathway. The uveoscleral pathway is poorly under-
(far-sighted) eyes with short axial length, shallow stood. In glaucoma, elevated IOP is typically caused
by decreased facility of outflow through the TM
anterior chamber depth and narrow anatomical an-
pathway. Episcleral venous pressure can also influ-
gles are predisposed to angle-closure glaucoma. An ence IOP through its effect on TM outflow. The
acute attack of angle-closure glaucoma may cause relationship of IOP, aqueous humour production,
markedly elevated IOP and symptoms, such as outflow and episcleral venous pressure can be de-
headache, nausea, emesis and blurred vision from scribed as follows:
corneal oedema. If the anatomical blockade of the
F−U
TM is not relieved by peripheral iridectomy (either IOP = + Pv
C
by laser peripheral iridectomy or incisional surgery),
where F is the aqueous humour formation (μL/min),
extensive visual loss may result. Chronic angle-
U is the uveoscleral outflow (μL/min), C is the
closure glaucoma is much less common in Cauca-
trabecular meshwork outflow facility (μL/min/
sians than POAG,[17] but is a significant cause of mm Hg) and Pv is the episcleral venous pressure
glaucoma-related blindness among Asians and Es- (mm Hg).
kimos.[18] Aqueous humour is thought to be derived from
Neovascularisation of the iris (rubeosis iridis) plasma within the capillary network of the ciliary
and iridocorneal angle (NVA) can lead to chronic body (figure 1). Following ultrafiltration of plasma
angle-closure (neovascular) glaucoma if the aetiolo- into the extracellular space or stroma of the ciliary
gy of the neovascularisation is left untreated. Chron- processes, active transport and secretion of this fluid
ic uveitis and retinal ischaemia, often seen with into the posterior chamber of the eye by the nonpig-
mented epithelium of the ciliary body occurs at a
proliferative diabetic retinopathy and retinal vascu-
rate of 1.4–4.3 μL/min.[19] Aqueous humour produc-
lar occlusion, are common causes of rubeosis iridis tion decreases with age.[20] Patients with diabetes
and NVA that can result in neovascular glaucoma. mellitus[21] and inflammation of the eye (irido-
The main goal of treatment for all forms of cyclitis)[22] also experience decreased rates of aque-
glaucoma is the preservation of visual function. The ous humour formation. A large fluid intake
cornerstone of therapy to achieve this goal is the (1000mL of water) has been shown to significantly
reduction of IOP. This report discusses the efficacy, increase aqueous humour production after 90 min-
safety and basic pharmacology of currently availa- utes.[23] Carbonic anhydrase, adenosine triphos-
ble agents that can be used as monotherapy or in phatases and adrenoceptors located in the nonpig-
mented ciliary epithelium are thought to play an
combination for the medical treatment of glaucoma.
important role in aqueous humour formation. Phar-
The physiology of aqueous humour and IOP is re- macological modulation of these enzymes and re-
viewed in order to understand better the mechanism ceptors can reduce aqueous humour formation and
of action of drugs used in the treatment of this lower IOP.
disorder. Finally, a brief description of other treat- Following transport of aqueous humour into the
ment modalities for glaucoma, including laser ther- posterior chamber, it flows through the pupil into the
apy and surgery, are provided. anterior chamber (figure 1). Once there, it maintains

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
Pharmacological Therapy of Glaucoma 5

a physiological IOP and also nourishes the avascular Safety and tolerability are important considera-
cornea and lens. It may aid in the metabolism of the tions when selecting glaucoma therapy as well.
vitreous and retina as well.[24] As mentioned earlier, Glaucoma drops can produce a variety of local ad-
aqueous humour exits the eye through one of two verse effects ranging from induced miosis by
pathways. Most of the aqueous humour (about 80%) cholinergics (acetylcholine receptor agonists) to
leaves the eye through the TM pathway. This path- lash growth by PGAs (see section 7). Topical drugs
way consists of TM, a sieve-like structure which can also produce important systemic adverse effects.
consists of a connective tissue core surrounded by Once a glaucoma medication is instilled into the eye,
endothelium, and Schlemm’s canal, a 360° collector much of it (about 80%) passes through the naso-
channel for aqueous humour that leads to the epis- lacrimal canal and is absorbed into the systemic
cleral venous system. A small amount of aqueous circulation. Such absorption avoids hepatic first-
humour (about 20%) flows directly through the iris pass metabolism, so with some agents, systemic
root and interstitial spaces of the ciliary muscle into adverse effects can be significant.[29] Eyelid closure
the suprachoroidal space (uveoscleral pathway). The and punctal occlusion following drop instillation
relative percentages of aqueous humour which exit have been shown to significantly decrease the sys-
the anterior chamber through these two pathways is temic absorption of topical medications.[30]
variable and can change with age and presence of
disease.[20,25,26] Pharmacological modulation of ad- 3. Cholinergics (Acetylcholine
renoceptors and prostanoid receptors located in the Receptor Agonists)
TM or ciliary body can increase aqueous humour
The cholinergics, also known as parasympath-
outflow through one or both pathways and lead to a
omimetics or miotics, were the first class of agents
reduction in IOP.
used for the treatment of glaucoma, introduced over
2. Medical Treatment of Glaucoma 100 years ago.[31] They can be divided into two
categories: the direct-acting agents work directly on
The cornerstone of glaucoma therapy is the re- the parasympathetic receptors in the eye, whereas
duction of IOP to a level which safely halts progres- the indirect-acting agents inhibit acetylcholinester-
sive visual loss. For most patients, initial treatment
begins with medication. Currently, there are five Schemm's canal
main categories of medication for the treatment of Trabecular
glaucoma (table III). Typically, treatment begins meshwork Ciliary body
with the selection of an agent as monotherapy. β- Cornea
Adrenoceptor antagonists have been a mainstay of
the medical treatment of glaucoma for many years
and are still often used as first-line therapy, particu-
larly in Europe and Latin America. Recently, the Lens
prostaglandin analogues (PGAs) have emerged as a
popular choice for the first-line treatment of Iris
glaucoma.[27] Following a therapeutic trial, the med- Anterior
ication is either continued or switched to a different chamber
class of medication based on its effect. If expected
reduction in IOP is achieved, but additional pressure Posterior
reduction is required, another agent can be added to chamber
the regimen. The past few years have seen an enor- Fig. 1. Schematic diagram of the anterior segment of an eye in
mous increase of new available medications for cross section. The dotted line indicates the flow of aqueous humour
glaucoma therapy (table III). A recent review esti- from its source in the posterior chamber (the ciliary body), through
the pupil, into the anterior chamber and toward the trabecular
mated that there are now over 56 000 possible com- meshwork. A majority of aqueous humour traverses the trabacular
binations of medications for the treatment of meshwork, collects in Schlemm’s canal, then drains into the venous
glaucoma.[28] system (not shown).

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
6 Marquis & Whitson

Table III. Drugs used for the treatment of glaucoma


Drug class/drug Strength/dose Usual dosage
Cholinergics (acetylcholine receptor agonists)
Pilocarpine ophthalmic solution (%) 0.5, 1, 2, 4, 6, 8 qid
Pilocarpine ophthalmic gel (%) 4 hs
Carbachol (%) 0.75, 1.5, 3 tid
Ecothiophate iodide (%) 0.25 bid

Adrenoceptor agonists
Nonselective
epinephrine (adrenaline) [%] 0.5, 1, 2 bid
dipivefrine (%) 0.1 bid
Selective α2
apraclonidine (%) 0.5, 1.0 tid
brimonidine (%) 0.2 tid
brimonidine P (%) 0.15 tid

Carbonic anhydrase inhibitors


Systemic
acetazolamide (mg) 125, 250 qid
acetazolamide sustained release (mg) 500 bid
methazolamide (mg) 25, 50 bid to tid
Topical
brinzolamide suspension (%) 1 tid
dorzolamide solution (%) 2 tid

β-Adrenoceptor antagonists
Nonselective
carteolol (%) 1 bid
levobunolol (%) 0.25, 0.5 bid
metipranolol (%) 0.3 bid
timolol (%) 0.25, 0.5 bid
timolol GFS (%) 0.25, 0.5 qd
Selective
betaxolol suspension (%) 0.25 bid
betaxolol solution (%) 0.5 bid

Prostaglandin analogues
Bimatoprost (%) 0.03 qd
Latanoprost (%) 0.005 qd
Travoprost (%) 0.004 qd
Unoprostone (%) 0.15 bid

Combination agents
Dorzolamide/timolol (%) 2.0/0.5 bid

Hyperosmotic agents
IV mannitol 0.5–2 g/kg
IV urea 0.5–2 g/kg
PO glycerin 1–1.5 g/kg
bid = twice daily; GFS = gel-forming solution; hs = at bedtime; IV = intravenous; PO = oral; qd = every day; qid = four times daily; tid =
three times daily.

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
Pharmacological Therapy of Glaucoma 7

ase, the enzyme responsible for the degradation of paralysis can occur in patients treated with indirect-
acetylcholine. The cholinergics lower IOP by in- acting cholinergics during general anaesthesia if
creasing outflow of aqueous humour through the suxamethonium chloride is used as a muscle relax-
TM. ant.[35] A cataractogenic effect has also been de-
scribed with chronic use of ecothiophate iodide for
3.1 Direct-Acting Cholinergics the treatment of glaucoma.[36] The indirect agents
are still often used for the treatment of glaucomas in
Pilocarpine is the most commonly prescribed
aphakia or pseudophakia across much of Europe and
cholinergic compound. It works by directly stimu-
Latin America.
lating the muscarinic receptors of the ciliary muscle
that widens the anterior chamber angle, resulting in 3.3 Dual-Mechanism Cholinergics
increased outflow of aqueous humour through the
TM. Pilocarpine is available in multiple concentra- Carbachol works by directly stimulating musca-
tions ranging from 0.5% to 8%. It is a short-acting rinic receptors within the eye as well as indirectly by
agent and must be applied four times daily. It is also inhibiting acetylcholinesterase.[37] It is available in
available as a 4% gel, which is usually applied at concentrations ranging from 0.75% to 3.0% and is
bedtime. Pilocarpine typically reduces IOP by about typically applied three times daily. Carbachol re-
20–30%.[32] It has been shown to decrease uveos- quires an adjuvant, such as benzalkonium chloride
cleral outflow,[33] which may have a significant to effectively penetrate the cornea and achieve ef-
clinical effect in eyes with compromised TM out- fective levels in aqueous humour. Carbachol is more
flow. potent than pilocarpine,[38] but is also more likely to
produce browache and accommodative spasm.[38] It
3.1.1 Adverse Effects
is no longer commonly used in current medical
Ocular adverse effects of pilocarpine are related practice.
to its effect on the ciliary and pupillary sphincter
muscle. Visual acuity is often diminished as a result 4. Adrenoceptor Agonists
of pupillary constriction and accommodative spasm.
Other local adverse events include browache and,
4.1 Nonselective Adrenoceptor Agonists
rarely, retinal detachment. Systemic adverse effects
from pilocarpine are not common but can include Epinephrine (adrenaline) stimulates both α- and
increased salivation and sweating, diarrhoea, vomit- β-adrenoceptors within the eye. IOP reduction is
ing and tachycardia. While pilocarpine is an inex- achieved primarily by increased aqueous humour
pensive and effective IOP-lowering agent, it is not outflow through both the TM and uveoscleral path-
as commonly used today as in previous years be- ways.[39] An acute decrease in aqueous humour pro-
cause of its local adverse effects and multiple daily duction from vasoconstriction following instillation
dosage requirements. has been described,[40] but is not thought to be
important clinically. Epinephrine is available in con-
3.2 Indirect-Acting Cholinergics centrations ranging from 0.5% to 2.0% and is ap-
The indirect-acting cholinergics include ecothio- plied twice daily, although it is rarely used in mod-
phate iodide and demecarium bromide. These agents ern clinical practice. Reductions in IOP ranging
work by inhibiting acetylcholinesterase, the enzyme from 15% to 25% have been reported.[41]
responsible for the degradation of acetylcholine. 4.1.1 Adverse Effects
Like pilocarpine, the indirect-acting miotics are Local adverse effects of epinephrine include pu-
available in multiple concentrations. They are typi- pillary dilation, conjunctival hyperaemia and ocular
cally used twice daily. The reduction in IOP is irritation.[42] Cystoid macular oedema (CMO) has
comparable to that seen with pilocarpine.[34] In addi- been described in patients treated with topical epi-
tion to the potential adverse effects seen with pilo- nephrine following cataract surgery.[43] Long-term
carpine, these agents deplete systemic cholinester- administration of epinephrine can result in adre-
ase and pseudocholinesterase. Prolonged respiratory nochrome deposits in the conjunctiva[44] and cor-

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
8 Marquis & Whitson

nea.[45] Systemic adverse effects associated with epi- for prevention of postoperative IOP elevations fol-
nephrine therapy include headache, palpitations, lowing anterior segment laser surgery. The original
high blood pressure and anxiety. 0.2% formulation is approved for three times daily
Dipivefrine 0.1% is a pro-drug molecule of epi- administration in the US, although it is often used
nephrine with two ester groups added. It was de- twice daily by most clinicians It has received formal
signed to enhance corneal penetration and reduce approval for twice daily administration in other
systemic and local adverse effects. Following drop countries. Brimonidine 0.2% has been shown to be
instillation, corneal esterases cleave the molecule equivalent to timolol at reducing IOP at peak (hour 2
into an active metabolite, which then passes into the after administration), but is significantly less effec-
anterior chamber.[46] IOP reduction with dipivefrine tive at trough.[53,54] Brimonidine 0.2% is significant-
is comparable to that seen with epinephrine, while ly more effective at reducing IOP than the cardiose-
both local and systemic adverse effects are seen less lective β-adrenoceptor antagonist, betaxolol, at peak
frequently.[47] effect (5.8mm Hg vs 3.8mm Hg, p = 0.004).[55] The
trough reduction in IOP; however, is comparable
4.2 Selective α2-Adrenoceptor Agonists between the two agents (3.9mm Hg
vs 3.2mm Hg).[55]
Clonidine is a relatively selective α2-adrenocep-
tor agonist with some α1-adrenoceptor agonistic 4.2.1 Adverse Effects
activity. It lowers IOP by decreasing production of Local adverse effects associated with apracloni-
aqueous humour.[48] It is a very lipophilic molecule dine include allergic blepharoconjunctivitis
and readily crosses the blood-brain barrier. Systemic (15–48%), tearing and foreign body sensation.[56]
hypotension can be seen following topical instilla- Eyelid retraction and pupillary mydriasis, through
tion of clonidine. It is still used clinically in parts of α1-adrenoceptor stimulation, can be seen as well.
Europe for the treatment of glaucoma. Systemic adverse effects include dry mouth, and
Apraclonidine, or para-aminoclonidine hydro- nose, headache and fatigue.[57] Unlike clonidine,
chloride, is a para-amino derivative of clonidine cardiovascular adverse effects, such as systemic hy-
with relatively selective α2-adrenoceptor agonistic potension are rarely seen with apraclonidine.[58]
activity. It is a much more hydrophilic molecule
Brimonidine is devoid of many of the α1-adre-
than clonidine and less likely to penetrate the blood-
brain barrier. Apraclonidine lowers IOP by reducing noceptor mediated adverse effects seen with
aqueous humour production and increasing outflow apraclonidine, such as pupillary mydriasis, eyelid
through the TM pathway.[49] It is available in 0.5% retraction and conjunctival blanching. The most
and 1.0% concentrations. Mean reductions in IOP common ocular adverse event seen with brimoni-
for both concentrations range from 20% to 27%.[50] dine is allergic blepharoconjunctivitis, occurring in
The 0.5% concentration is typically used for short- 12–15% of patients after several months of ther-
term adjunctive therapy (<3 months) in patients not apy.[53,54] Systemic adverse effects reported with
controlled with other agents, whereas the 1.0% con- brimonidine use include dry mouth (33%), fatigue
centration is indicated for the prevention of postop- (16%) and headache (18.7%).[53,54] Brimonidine
erative IOP elevation following anterior segment should be avoided in small children because of
laser surgery. Apraclonidine is rarely used for long- associated CNS and respiratory depression.
term therapy because of its high rate of allergic A new formulation, brimonidine P, has recently
blepharoconjunctivitis.[51] been introduced that contains a lower concentration
Brimonidine is a highly selective α2-adre- of brimonidine (0.15%) and a different vehicle with
noceptor agonist. It lowers IOP by decreasing aque- stabilised oxychloro complex as a preservative in
ous humour production and increasing uveoscleral place of benzalkonium chloride. Comparable reduc-
outflow.[52] It is used as mono- or, more typically, tion in IOP to the original 0.2% solution has been
adjunctive therapy for the long-term management of reported along with a significantly lower rate of
glaucoma or ocular hypertension. It is also effective allergy (9.2% vs 15.7%, p = 0.007).[59] Lower rates

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
Pharmacological Therapy of Glaucoma 9

of fatigue, dry mouth and conjunctival hyperaemia 5.2 Topical CAIs


were also found with the new formulation.[59]
Dorzolamide was the first topical CAI to be used
5. Carbonic Anhydrase Inhibitors (CAIs) for glaucoma therapy, introduced into the US mar-
ket in 1994. It is available as a 2.0% ophthalmic
The carbonic anhydrase inhibitors (CAIs) are solution with a relatively acidic pH of 5.5 and is
sulfonamide drugs that lower IOP by reducing aque- approved for three times daily administration. At
ous humour formation. In the ciliary epithelium, this dosage it lowers IOP by 18–22%.[63] It has been
carbonic anhydrase isoenzyme II catalyses the con- shown to be less effective at lowering IOP than oral
version of CO2 and H2O to HCO3– and H+, a acetazolamide or 0.5% timolol used twice daily, but
process important for the production of aqueous comparable to 0.5% betaxolol used twice daily.[63] It
humour. By inhibiting this conversion, aqueous hu- is additive in hypotensive efficacy to timolol[64] and
mour formation is decreased. CAIs are available as latanoprost.[65] Comparisons between 2% dorzo-
both oral (systemic) and topical agents. lamide and 0.2% brimonidine used as monotherapy
have shown similar efficacy at both peak and
5.1 Systemic CAIs trough.[66,67]
Brinzolamide was introduced into the US market
Acetazolamide was first used for glaucoma ther- in 1998. It is available as a 1.0% ophthalmic suspen-
apy in 1954.[60] It is currently available in 125mg sion with a more physiological pH of 7.4, roughly
and 250mg tablets, and typically administered four equivalent to that of human tears. Like dorzolamide,
times daily. Sustained release capsules of 500mg are brinzolamide is approved for three times daily ad-
also available and are used twice daily. Acetazo- ministration. It has been shown to lower IOP equally
lamide produces reduction in IOP of about as well as dorzolamide.[68] Brinzolamide has been
20–30%.[61] Methazolamide, a slightly weaker CAI, reported to be better tolerated than dorzolamide by
is available in 25mg and 50mg tablets. It can be most patients, presumably as a result of its more
administered two or three times daily. It is slightly physiological pH.[69]
less effective than acetazolamide, but is sometimes
better tolerated by patients. 5.2.1 Adverse Effects
When introduced in the mid 1990s, the topical
5.1.1 Adverse Effects CAIs represented a major advance over the oral
The oral CAIs are effective IOP-lowering agents agents for the medical management of glaucoma.
but are limited in their clinical usefulness by their Thus far, they appear to be devoid of many of the
numerous and often severe adverse effects. Paraes- significant systemic adverse effects seen with the
thesias of the hands and feet, nausea, vomiting, oral CAIs. Bitter taste has been reported in about
fatigue and weight loss are all common complica- 25% of patients.[70] Local adverse events seen with
tions of chronic oral CAI therapy. Less commonly, dorzolamide include stinging (12%), burning (19%)
renal stones may develop with long-term use. Sys- and itching (12%).[63] Irreversible corneal decom-
temic acidosis, hypokalaemia, and hyponatraemia pensation has been reported in patients with marked
often result from inhibition of carbonic anhydrase in endothelial compromise.[71] In a large, multicentre,
the kidney. Although uncommon, bone marrow de- comparative trial, brinzolamide caused less ocular
pression resulting in thrombocytopenia, agranulocy- discomfort than dorzolamide, with reported rates of
tosis and aplastic anaemia has been described.[62] burning and stinging of 3.0% and 16.4%, respective-
Rarely, Stevens-Johnson syndrome may result from ly.[72] Other ocular adverse events associated with
oral CAI therapy. Because of their poor tolerability brinzolamide include foreign body sensation
in most patients, the oral CAIs are typically reserved (1.8%), itching (1.2%) and dry eyes (1.2%).[72] Like
for short-term use in patients on maximal medical the oral CAIs, dorzolamide and brinzolamide are
therapy, often as a temporising measure before sur- sulfonamides and should be avoided in patients with
gery. sulfonamide hypersensitivity.

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
10 Marquis & Whitson

6. β-Adrenoceptor Antagonists available as 0.5% preparations. Once daily 0.5%


Timoptic XE® lowers mean IOP as effectively as
β-Adrenoceptor antagonists are a commonly pre- timolol 0.5% solution used twice daily.[80] Timolol
scribed class of agents for the treatment of Maleate Gel-Forming Solution® and 0.5% Timoptic
glaucoma. Since their introduction for ophthalmic
XE® produce clinically equivalent reductions in
use in 1979, they have been a mainstay of glaucoma
IOP.[81] Timolol remains the US FDA’s ‘gold stan-
treatment and a frequent choice for first-line ther-
dard’ drug for glaucoma therapy against which all
apy. Their popularity has declined somewhat in
recent years, however, with the introduction of more new medications must be compared prior to approv-
effective, safer drugs, in particular, the PGAs.[27] al.
Most topical ophthalmic β-adrenoceptor antagonists Levobunolol is available in 0.25% and 0.5% so-
used today are nonselective agents, inhibiting both lutions and is typically administered twice daily.
β1- and β2-adrenoreceptors. These include timolol, Most patients can maintain adequate IOP control
levobunolol, metipranolol and carteolol. A β1-selec- with once daily therapy as a result of its active
tive agent, betaxolol, is also available for use. β- metabolite, dihydrolevobunolol, which is also effec-
adrenoceptor antagonists lower IOP by reducing tive at lowering IOP.[82] Like timolol, levobunolol
aqueous humour formation, most likely through the lowers IOP by decreasing aqueous humour forma-
inhibition of catecholamine-stimulated synthesis of tion.[83] Its efficacy is equivalent to that of
cyclic adenosine monophosphate in the ciliary epi- timolol.[84]
thelium.[73] While β-adrenoceptor antagonists are
very effective IOP-reducing agents, tachyphylaxis, Carteolol is available as a 1% solution and, like
or ‘drift’ may occur after long-term therapy.[74] A timolol and levobunolol, it is administered twice
recent study has shown that after 2 years, as many as daily. Its efficacy is comparable to timolol with
50% of patients treated initially with a β-adre- average reported IOP reductions of 20–32%.[85,86]
noceptor antagonist as monotherapy will require a Carteolol demonstrates intrinsic sympathomimetic
different or additional medication for IOP con- activity (ISA) which produces an early, transient β-
trol.[75] adrenoceptor agonist response not seen with other
topical β-adrenoceptor antagonists. It was hoped
6.1 Nonselective that the ISA of carteolol might protect against some
β-Adrenoceptor Antagonists of the systemic adverse effects seen with other β-
adrenoceptor antagonists, such as reduced pulse and
Timolol was the first ophthalmic β-adrenoceptor blood pressure. While this has not proven to be the
antagonist approved for the treatment of glaucoma.
case, carteolol has been shown to have fewer delete-
Like other β-adrenoceptor antagonists, it lowers IOP
rious effects on serum lipids than timolol. In a
by decreasing aqueous humour formation.[73] It is
comparative study involving 58 normolipidemic
most effective during waking hours, having little to
no effect on aqueous humour production during adult men, topical carteolol therapy was associated
sleep and producing less reduction in IOP at with a 3.3% decrease in plasma high-density lipo-
night.[73,76] Timolol reduces IOP by 20–35% on av- protein-cholesterol (HDL-C) and a 4% increase in
erage.[77,78] As a result of its binding to iris melanin, total cholesterol/HDL-C ratio, whereas timolol pro-
timolol is often less efficacious in patients with dark duced changes of 8% and 10%, respectively.[87] Me-
irides.[79] It is available in 0.25% and 0.5% concen- tipranolol 0.3% solution is used twice daily. Like the
trations, which are typically used twice daily. Once other topical β-adrenoceptor antagonists, it lowers
daily gel-forming solutions (Timoptic-XE®, 1 IOP by reducing aqueous humour production.[88]
Merck & Co., Inc., West Point, PA, USA and Studies have shown comparable efficacy to timolol
Timolol Maleate Gel-Forming Solution®, Alcon and levobunolol with average IOP reductions of
Laboratories, Inc., Fort Worth, TX, USA) are also 21–33%.[89,90]

1 The use of trade names is for product identification purposes only and does not imply endorsement.

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
Pharmacological Therapy of Glaucoma 11

6.1.1 Adverse Effects 6.2.1 Adverse Effects


In general, topical nonselective β-adrenoceptor Except for occasional transient stinging upon in-
antagonists are effective and well tolerated by most stillation, betaxolol is typically well tolerated local-
patients. Reported local adverse effects include con- ly. It was designed to be specific for β1-adre-
junctival hyperaemia, stinging, superficial punctuate noceptors, thereby minimising the potential for pul-
monary (β2-adrenoceptor mediated) adverse effects.
keratitis and worsening dry eye symptoms.[91] Sev-
In most patients with restrictive airway disease,
eral reports of reversible anterior granulomatous
betaxolol does not exacerbate breathing
uveitis following long-term use of metipranolol
problems.[106] However, exceptions have been re-
have been published since the early 1990s.[92,93] ported, so caution must be used when prescribing
These agents can exert serious systemic adverse betaxolol for patients with respiratory ail-
effects from inhibition of β1-adrenoceptors of the ments.[107,108] While cardiovascular adverse effects,
heart and β2-adrenoceptors of the lung. Bradycardia, such as bradycardia and congestive heart failure
arrhythmia, cardiac block, congestive heart failure have been reported with betaxolol therapy, they
and bronchospasm have been reported following the appear to be less common than those which are seen
use of topical nonselective β-adrenoceptor antago- with the nonselective agents.[109,110] CNS adverse
nists.[94,95] These agents should, therefore, be avoid- effects are also less common with betaxolol than
ed in patients with severe heart disease, asthma or with nonselective β-adrenoceptor antagonists.[111]
chronic obstructive pulmonary disease. They should
also be used with caution in patients with diabe- 7. Prostaglandin Analogues
tes,[96] due to their masking of hypoglycaemia, and
Since their introduction into the US market in
in patients with myasthenia gravis.[97] CNS adverse
1996, the PGAs have emerged as a popular choice
effects, including depression, anxiety, impotence, for the first-line treatment of glaucoma.[27] The
fatigue and hallucinations have also been described once-daily PGAs, latanoprost, travoprost and bi-
following the use of topical nonselective β-adre- matoprost, are typically the most potent class of
noceptor antagonists.[94,95,98] IOP-reducing drugs available. They also offer excel-
lent control of diurnal IOP fluctuation that occurs
6.2 Selective β-Adrenoceptor Antagonists commonly among glaucoma patients and increases
their risk of visual field progression over the long
Betaxolol is a cardioselective β1-adrenoceptor term.[112,113] Finally, except for minor cosmetic is-
sues, such as mild conjunctival hyperaemia and
antagonist that lowers IOP by reducing aqueous
eyelash growth, they pose few, if any, systemic
humour formation.[99] It is a less effective IOP-
safety concerns. A fourth PGA, unoprostone, is also
lowering agent than timolol and the other nonselec-
available, although it is less effective than other
tive agents with average reductions in IOP ranging
members of this class and must be used twice daily.
from 18% to 26%.[100,101] Despite its weaker IOP-
The PGAs are all multicarbon-chain, lipophylic
lowering efficacy, two reports have shown trends molecules, derived from arachidonic acid and shar-
for better visual field preservation with long-term ing similar structural features to PGF2α (figure 2).
betaxolol therapy than with timolol therapy.[102,103] They lower IOP by increasing outflow of aqueous
Although increasing ocular perfusion has not yet humour, primarily through the uveoscleral path-
been proven to be beneficial for the treatment of way.[114] Their exact mechanism of action is still not
glaucoma, several investigators have speculated that clear, but they are thought to exert their effect by
enhanced retinal and optic nerve head blood flow binding to the prostanoid FP receptors of the ciliary
from the calcium channel blocking properties of body, upregulating production of various metal-
betaxolol may account for this observed enhanced loproteinases. These metalloproteinases then re-
visual field preservation.[104,105] Betaxolol is availa- model the surrounding extracellular matrix making
ble as a 0.25% suspension and a 0.5% solution that it more permeable to aqueous humour, which leads
are each administered twice daily. to enhanced outflow of aqueous humour and reduc-

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
12 Marquis & Whitson

O
O O
O
HO HO

O CF3
HO HO
OH OH

PGF2α isopropyl ester Travoprost

O O
O
O
HO HO

HO HO
OH O

Latanoprost Unoprostone

NH
O
HO

HO
OH

Bimatoprost
Fig. 2. Chemical structures of prostaglandin F2α (PGF2α) and commercially available prostaglandin analogues.

tion of IOP.[115] PGAs have also been shown to relax occurred with bimatoprost compared with lata-
the ciliary muscle, further facilitating uveoscleral noprost (p = 0.001).
outflow.[33] Some increased outflow may occur Latanoprost was introduced into the US market
through the TM pathway as well.[116] Only one pub- in 1996. It has since become the most widely pre-
lished report has compared the safety and efficacy of scribed drug for the treatment of glaucoma in the US
all three once-daily PGAs (latanoprost, travoprost and in available countries around the world. Lata-
and bimatoprost) in a single, large, prospective, noprost has been compared with timolol in several
multicentre studies involving over 800 pa-
multicentre trial. This study, which involved more
tients.[118-120] Once-daily latanoprost administered in
than 400 patients with glaucoma or ocular hyperten- the evening was significantly more effective at low-
sion, showed statistically equivalent reduction in ering IOP than twice daily timolol. Mean IOP reduc-
IOP among the three agents after 3 months of ther- tion was 6.7 ± 3.4mm Hg for latanoprost compared
apy.[117] Significantly more conjunctival hyperaemia with 4.9 ± 2.9mm Hg for timolol after 6 months of

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
Pharmacological Therapy of Glaucoma 13

treatment.[118] Unlike timolol, which decreases offers excellent diurnal IOP control and long dura-
aqueous humour production during waking hours tion of action, with significant reduction in IOP
only, latanoprost has been found to be equally as (approximately 6mm Hg) remaining for at least 84
effective at reducing IOP during the evening as hours after its final dose.[131] It is a very stable
during the day.[121] Other studies have shown lata- compound and does not require refrigeration or pro-
noprost to be significantly more effective at reduc- tection from sunlight.[132] Travoprost is available as
ing IOP than dorzolamide[122] and brimonidine.[123] a 0.004% solution and, like latanoprost, is adminis-
A long-term study involving over 300 patients with tered once daily in the evening.
glaucoma or ocular hypertension has shown no loss Bimatoprost was also introduced into the US
of efficacy after 5 years of use of latanoprost as an market in the spring of 2001. It is termed a pros-
adjunctive agent.[124] At this time, latanoprost is the tamide because of its unique structural presence of
only PGA to have received a formal first-line usage an amide rather than an ester group at the carboxy-
approval from the FDA. It requires refrigeration for terminal end of the α carbon chain (figure 2). It is
long-term (>1 year) storage as well as protection still not clear whether bimatoprost is hydrolysed to a
from sunlight to maintain stability.[125] It does not free acid during its passage through the cornea (as
require refrigeration once opened by the patient. It is are the other members of this class of agents) ena-
available as a 0.005% solution administered once bling it to bind to the prostanoid FP receptor, or
daily, typically in the evening. whether it enters the eye as an intact molecule to
Travoprost was introduced into the US market in interact with a unique, as yet, unidentified, pros-
the spring of 2001. Similar to the other PGAs, it tamide receptor.[133,134] It lowers IOP by increasing
works by increasing outflow of aqueous humour aqueous humour outflow through both the uveos-
through the uveoscleral pathway. It differs from the cleral (pressure-insensitive) pathway as well as the
other PGAs, which exhibit partial agonist activity, in TM (pressure-sensitive) pathway.[135] In a large,
that it is a full agonist at the PGF2α receptor.[126] randomised, clinical trial involving over 1100 pa-
Like latanoprost, travoprost is thought to reduce IOP tients with glaucoma or ocular hypertension, once-
by interaction with this receptor resulting in in- daily bimatoprost was found to be significantly
creased outflow of aqueous humour.[126] In a large, more effective at lowering IOP than timolol
multicentre trial involving over 800 patients with (8.1mm Hg vs 5.6mm Hg, p < 0.001).[136] Bimato-
glaucoma or ocular hypertension, 0.004% travoprost prost was also found to be equal or superior at
was found to be equal or superior to 0.005% lata- reducing IOP compared with latanoprost after 3
noprost using pooled data, and superior to 0.5% months of therapy in 232 patients.[137] When signifi-
timolol at reducing IOP after 1 year with final IOP cant baseline IOP differences were accounted for in
values ranging from 17.7mm Hg to 19.1mm Hg for this study, both drugs produced equivalent mean
travoprost, 18.5mm Hg to 19.2mm Hg for lata- reductions in IOP. More recently, a large, randomis-
noprost and 19.4mm Hg to 20.3mm Hg for timo- ed multicentre trial involving 269 patients has
lol.[127] Travoprost and latanoprost produced clini- shown bimatoprost to produce significantly greater
cally and statistically equivalent reductions in IOP at reduction in IOP than latanoprost at all assessment
the majority of individual time points evaluated points after 6 months of treatment.[138] However, in
throughout this 1 year study. Two other multicentre this study, the mean IOP lowering effect of lata-
trials have confirmed its greater efficacy when com- noprost at 8:00am. (24.1%) was much less than that
pared with timolol.[128,129] Travoprost has also been reported in previous studies (approximately
shown to be an effective adjunctive agent offering 30–35%).[118-120] Bimatoprost has also been com-
an additional 5–7mm Hg IOP reduction in patients pared with the fixed combination 2% dorzolamide/
inadequately controlled on timolol.[130] Subgroup 0.5% timolol (Cosopt®, Merck & Co., Inc., West
analysis has revealed that travoprost is significantly Point, PA, USA). Bimatoprost produced significant-
more effective at reducing IOP (by almost 2mm Hg) ly greater reduction in IOP than Cosopt® at the early
in Blacks compared with non-Blacks.[127] morning timepoint at each visit throughout the 3-
Postmarketing studies have shown that travoprost month study.[139] Furthermore, about twice as many

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
14 Marquis & Whitson

patients had an IOP <16mm Hg at the final visit with from 1.1% to 23% have been reported with the once-
bimatoprost (31% vs 14%). However, it is important daily PGAs.[119,127,136] The incidence with unopros-
to note that Cosopt® performed less well in this tone appears to be much lower. The increase in
study than in earlier reports,[64,140] producing reduc- pigmentation is irreversible and is a result of in-
tions in IOP ranging from only 4.4mm Hg to creased production of melanin within iris melano-
5.6mm Hg versus a more typical range of about cytes, not a proliferation of melanocytes.[146] Eye-
7.5–9.0mm Hg. Bimatoprost is available as a 0.03% lash growth is also common with long-term use of
solution and is administered once daily in the eve- these agents with reported rates ranging from 26%
ning. It does not require refrigeration to maintain to 57%.[127,136] Exacerbations of anterior uveitis[147]
stability.[141] and herpetic keratitis[148] have been reported with
Unoprostone was introduced in Japan in 1994 latanoprost use. CMO has been reported with use of
and became available in the US in the summer of latanoprost after complicated cataract surgery and in
2000. It differs structurally from the other PGAs in those with a history of CMO.[149,150] The PGAs
having a 22-carbon chain backbone instead of the should probably be avoided in these patients.
typical truncated 20-carbon structure found in the
other agents (figure 2). When used as monotherapy, 8. Combination Therapy
studies have shown twice daily unoprostone to be
significantly less effective than either timolol[142] or The selection of agents for combination therapy
latanoprost.[143] It has demonstrated comparable ef- in the treatment of glaucoma is a task commonly
ficacy to brimonidine or dorzolamide when used as encountered by clinicians. In the OHTS, almost 50%
an adjunctive agent to timolol.[144] It is available as a of patients required two or more medications to
0.15% formulation and is administered twice daily. reach the required, relatively modest, target IOP
Because unoprostone is less effective at lowering reduction of 20% from baseline and a final IOP of
IOP than the other PGAs and requires more frequent <24mm Hg.[15] For many years, β-adrenoceptor an-
administered, its clinical use has been somewhat tagonists have been a mainstay of glaucoma therapy
limited. and remain a popular choice for first-line therapy
In general, the PGAs are well tolerated drugs among some ophthalmologists. Recently, however,
with typically <5% of patients discontinuing regis- the once-daily PGAs have assumed a leading role in
tration studies as a result of adverse events. Report- the initial medical treatment of glaucoma.[27] Once a
ed systemic adverse effects have been limited to first-line agent is chosen, its efficacy is evaluated
headache and upper respiratory tract symptoms. after a brief trial period. If less than expected IOP
Specifically, latanoprost has been shown to have no reduction is produced, the drug is usually stopped
deleterious effect on blood pressure, pulse and pul- and an alternate class of agents is tried. If the ex-
monary function in patients with chronic obstructive pected IOP reduction occurs but a lower IOP is
pulmonary disease.[145] Local cosmetic effects, on needed for that particular patient, selection of an
the other hand, are common with these agents. Con- adjunctive agent is typically performed.
junctival hyperaemia has been reported in 3–15% of When selecting an adjunctive agent, its safety
patients treated with latanoprost.[118-120] Hyperaemia and tolerability are of paramount concern. A simple
is more commonly seen with the newer agents with dosage regimen can enhance patient compliance.
reported rates of 15–45% for bimatoprost[136,137] and Also, an agent’s ability to work well with the initial
35–50% for travoprost.[127-129] Typically, the con- drug chosen is important to consider. A drug used
junctival hyperaemia seen with these agents is fairly for the treatment of glaucoma lowers IOP by one of
mild in most patients. About 3% of patients in phase two ways, as mentioned earlier: reduction of aque-
III studies discontinued therapy with bimatoprost ous humour production or increased outflow of
and travoprost because of hyperaemia. aqueous humour (table IV). When choosing agents
An increase in iris pigmentation, most often in for combination therapy, those drugs with comple-
multicoloured irides, can occur with long-term use mentary mechanisms of action usually work well
of the PGAs. Rates of iris colour change ranging together.

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
Pharmacological Therapy of Glaucoma 15

Table IV. Mechanism of action of drugs used to treat glaucoma decreasing production of aqueous humour, they do
Decrease aqueous humour production so by different mechanisms – β-adrenoceptor antag-
α-Adrenoceptor agonists onists by inhibition of circulating catecholamines on
β-Adrenoceptor antagonists the ciliary epithelium and topical CAIs by inhibition
Carbonic anhydrase inhibitors of carbonic anhydrase. The combination drop
Increase aqueous humour outflow
Cosopt® has been shown to produce equivalent
α-Adrenoceptor agonists
reduction in IOP to its two components taken sepa-
Cholinergics (acetylcholine receptor agonists)
rately (ranging from 27% to 33% overall reduction
Prostaglandin analogues
in IOP), whereas offering patients a simpler regimen
that requires fewer bottles and daily drop instilla-
Assuming a PGA is chosen for initial monother- tions.[64] Recently, Cosopt® has been shown to pro-
apy, agents which are typically used for adjunctive duce equivalent reduction in IOP compared with a
treatment include β-adrenoceptor antagonists, regimen of 0.5% timolol and 0.2% brimonidine tak-
α2-adrenoceptor agonists (brimonidine), or topical en separately.[156] Orzalesi et al.[157] have shown that
CAIs. β-Adrenoceptor antagonists are additive to a Cosopt® and latanoprost produce equivalent reduc-
PGA, and the use of a once-daily gel-forming β- tions in IOP throughout 24 hours, while each agent
adrenoceptor antagonist in the morning combined performed significantly better than 0.2% bromoni-
with a PGA in the evening has been a popular dine during the night when compared as monother-
combination for ophthalmologists for several years. apy. Another combination product comprised 0.5%
However, recent studies which have investigated the timolol and 0.004% latanoprost (Xalacom®, Pfizer
additional IOP reduction provided by β-adre- Inc., New York, NY, USA), has been recently ap-
noceptor antagonists when added to a PGA have
proved for use in several countries across Europe,
been rather disappointing, with most reports show-
Latin America and Canada. A large multicentre
ing only about 1mm Hg additional reduction in IOP
study involving >400 patients with glaucoma or
by timolol when added to latanoprost.[151,152] The
ocular hypertension has shown that treatment with a
newly formulated 0.15% brimonidine P solution,
when added to bimatoprost, has been shown to have fixed combination of latanoprost and timolol result-
equal or better IOP-lowering efficacy compared ed in significantly (p < 0.001) lower IOP
with a timolol gel-forming solution used with lata- (19.9 ± 3.4mm Hg) than either latanoprost
noprost (mean IOP reduction at 10:00am of (20.8 ± 4.6mm Hg) or timolol (23.4 ± 5.4mm Hg)
8.5mm Hg vs 7.7mm Hg).[153] taken individually for 6 months.[152] Combination
products of timolol and pilocarpine (Timpilo®,
The topical CAIs appear especially promising for
Merck & Co., Inc., West Point, PA, USA), and
adjunctive agents used in combination with a PGA.
Recent studies have shown an overall reduction in metipranolol and pilocarpine (Normoglaucon®,
IOP of about 11mm Hg, or 40%, from baseline Bausch & Lomb Pharmaceuticals, Inc., Tampa, FL,
when dorzolamide is added to latanoprost.[65,154] A USA) have been developed and have shown similar
recent review compared the additional IOP reduc- efficacy when compared with the agents taken sepa-
tion provided by β-adrenoceptor antagonists, rately.[158,159] Although not available in the US, these
brimonidine or dorzolamide in 73 patients inade- two combination products are still commonly used
quately controlled by latanoprost monotherapy. Af- in many other parts of the world.
ter 1 year, the topical CAI, taken either two or three Several new fixed combination products have
times daily produced the most additional IOP reduc- recently been developed and are currently being
tion (3.9mm Hg) compared with β-adrenoceptor reviewed by the FDA for potential use in the US.
antagonists (2.5mm Hg) and brimonidine These include a combination 0.2% brimonidine/
(2.0mm Hg).[155] 0.5% timolol drop (Combigan®, Allergan, Inc., Ir-
If a β-adrenoceptor antagonist is used as initial vine, CA, USA), a combination 0.004% travoprost/
treatment, topical CAIs are an excellent choice for 0.5% timolol drop (Extravan®, Alcon Laboratories,
adjunctive therapy. Although both drugs work by Inc., Fort Worth, TX, USA), and a combination

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
16 Marquis & Whitson

0.03% bimatoprost/0.5% timolol drop (Allergan, damage compared with ALT.[169] SLT is also
Inc., Irvine, CA, USA). thought to lower IOP by improving outflow facility
at the TM. The mechanism of this action is not
9. Other Treatment Options understood but may involve cytokine release from
pigmented TM cells, subsequent recruitment of
Glaucoma is a chronic disease and typically re- macrophages, and removal of cellular debris.[169] In
quires long-term administration of IOP-lowering a study comparing eyes treated with ALT versus
drugs. Patients are followed up periodically with SLT, both groups achieved reductions in IOP of
assessment of IOP, optic discs and visual fields to about 22% from baseline after 6 months.[170] Eyes
exclude progressive damage and monitor the suc- with previously failed ALT had a better reduction in
cess of the chosen treatment regimen. Medications IOP with SLT than with repeat ALT
are usually added in a step-wise fashion until the (6.8 ± 2.4mm Hg vs 3.6 ± 1.8mm Hg, p = 0.01).[170]
desired pressure-lowering effect is obtained. Should SLT has similar potential complications to those of
maximal tolerated medical therapy not reach the ALT.
desired target pressure goal, or if adverse effects,
cost, or noncompliance preclude use of topical 9.2 Surgical Therapy
drugs, other treatment modalities for glaucoma, in-
cluding laser therapy or incisional surgery can be For glaucoma patients who are refractory to med-
considered. ical and laser therapy, incisional surgery can be
performed. The modern trabeculectomy technique
9.1 Laser Therapy was described over 30 years ago[171] and remains the
initial glaucoma surgical procedure for most pa-
Argon laser trabeculoplasty (ALT) was original- tients. The procedure involves creating a fistula
ly described over 20 years ago.[160] This procedure underneath a scleral flap into the anterior chamber.
involves placing evenly spaced nonpenetrating ar- Aqueous humour exits the eye through this opening
gon laser burns around the circumference of the TM. and collects in a ‘bleb’ in the subconjunctival space.
The procedure is performed at the slit lamp with the IOP is reduced by increased outflow of aqueous
aid of a gonioprism; 50–100 evenly spaced spots are humour through the surgical site. A limiting factor
applied to the TM over 180–360° of the angle. for the long-term success of filtration surgery is
Commonly used treatment parameters are 50μm scarring at the fistula site that leads to decreased
spot size, 0.1 second pulse duration and 800mW aqueous humour outflow. Antifibrotic agents, such
power. ALT lowers IOP by improving the outflow as fluorouracil and mitomycin, diminish the postop-
facility at the TM. The mechanism is not fully erative subconjunctival scarring response and have
understood, but may relate to fibrosis and traction at been shown to enhance the success rate of
each burn site which stretches and opens adjacent trabeculectomy surgery.[172,173] Complications of
meshwork.[161] Reported short-term success rates trabeculectomy surgery include hypotony, cataract
range from 65% to 97%.[162,163] Most investigators formation, choroidal effusion or haemorrhage and
report a 5-year success rate of about 50%.[164] Com- endophthalmitis. Nonpenetrating trabeculectomy
plications of ALT include transient IOP eleva- (NPT) in which the innermost layer of the TM is left
tion,[165] iritis[166] and PAS formation.[167] intact allowing aqueous humour to percolate
Selective laser trabeculoplasty (SLT) has recent- through under a scleral flap has been described.[174]
ly been described.[168] SLT selectively delivers ener- A recent study revealed that compared with standard
gy to pigmented TM cells in a process termed trabeculectomy, NPT resulted in fewer early postop-
photothermolysis. The system employs a Q- erative complications, but also produced significant-
switched, frequency-doubled Nd : YAG laser. Com- ly less reduction in mean IOP after 18 months
monly used treatment parameters are 400μm spot (25.1% vs 35.7%, p = 0.0015).[175]
size, 3 nanosecond pulse duration and 0.8mJ energy. For those patients in whom the trabeculectomy is
The theoretical advantage of targeting pigmented not successful, drainage tube implant surgery can be
cells is that nonpigmented TM cells may sustain less performed. The basic design of a glaucoma drainage

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
Pharmacological Therapy of Glaucoma 17

device consists of a silicone tube that extends from References


the anterior chamber to a plate or disc beneath the 1. Quigley HA, Nickells RW, Kerrigan LA, et al. Retinal ganglion
cell death in experimental glaucoma and after axotomy occurs
subconjunctival space. A variety of glaucoma drain- by apoptosis. Invest Ophthalmol Vis Sci 1995 Apr; 36 (5):
age implant devices have been described in the 774-86
2. Thylefors B, Negrel AD, Pararajasegaram R, et al. Global data
literature.[176] Cyclodestructive procedures, which on blindness. Bull World Health Organ 1995; 73: 115-21
lower IOP by destroying part of the ciliary body, 3. Sommer A, Tielsch JM, Katz J, et al. Racial differences in the
thus reducing aqueous humour production, are typi- cause-specific prevalence of blindness in East Baltimore. N
Engl J Med 1991 Nov; 325 (20): 1412-7
cally reserved for eyes which are refractory to all 4. Rodriguez J, Sanchez R, Munoz B, et al. Causes of blindness
other forms of therapy. These procedures include and visual impairment in a population-based sample of US
Hispanics. Ophthalmology 2002 Apr; 109 (4): 737-43
cyclocryotherapy,[177] cyclodiathermy[178] and laser 5. Quigley HA, Vitale S. Models of glaucoma prevalence and
cyclophotocoagulation.[179] incidence in the United States. Invest Ophthalmol Vis Sci 1997
Jan; 38 (1): 83-91
6. American Academy of Ophthalmology preferred practice pat-
10. Conclusion tern for primary open-angle glaucoma. San Francisco (CA):
American Academy of Ophthalmology, 2003: 3
7. Drance SM, Sweeney VP, Morgan RW, et al. Studies of factors
The medical management of glaucoma has involved in the production of low tension glaucoma. Arch
Ophthalmol 1973 Jun; 89 (6): 457-65
evolved greatly in the past few years. A variety of 8. Migdal C, Gregory W, Hitchings R. Long-term functional out-
new, effective and safer agents for the treatment of come after early surgery compared with laser and medicine in
glaucoma introduced in the mid 1990s, including the open-angle glaucoma. Ophthalmology 1994 Oct; 101 (10):
1651-6
PGAs, the α2-adrenoceptor agonists and the topical 9. Shiose Y, Kitazawa Y, Tsukahara S, et al. Epidemiology of
CAIs have largely supplanted many older agents glaucoma in Japan: a nationwide glaucoma survey. Jpn J
Ophthalmol 1991; 35 (2): 133-55
that were once mainstays of therapy, including the 10. Drance SM, Morgan RW, Sweeney VP. Shock-induced optic
oral CAIs, the cholinergics and epinephrine. While neuropathy: a cause of nonprogressive glaucoma. N Engl J
β-adrenoceptor antagonists remain a good choice for Med 1973 Feb; 288 (8): 392-8
11. Lewis RA, Hayreh SS, Phelps CD. Optic disk and visual field
both first-line and adjunctive therapy, the PGAs correlations in primary open-angle and low-tension glaucoma.
have recently gained favour among most ophthal- Am J Ophthalmol 1983 Aug; 96 (2): 148-5
12. Chumbley LC, Brubaker RF. Low-tension glaucoma. Am J
mologists. One great benefit to arise from this Ophthalmol 1976 Jun; 81 (6): 761-8
change for both clinicians and patients alike is that 13. Caprioli J, Spaeth GL. Comparison of visual field defects in the
selection of glaucoma medication can now truly be low-tension glaucomas with those in the high-tension
glaucomas. Am J Ophthalmol 1984 Jun; 97 (6): 730-7
made on an individual patient basis. Should one 14. Kahn HA, Milton RC. Alternative definitions of open-angle
class of agents prove ineffective or poorly tolerated glaucoma: effect on prevalence and associations in the
by a patient, another safe and effective drug can Framingham eye study. Arch Ophthalmol 1980 Dec; 98 (6):
2172-7
readily be chosen. Accumulating evidence in the 15. Kass MA, Heuer DK, Higginbotham EJ, et al. The Ocular
literature supports the use of adjunctive agents as Hypertension Treatment Study: a randomized trial determines
that topical ocular hypotensive medication delays or prevents
safe and effective second- or third-line drugs for use the onset of primary open-angle glaucoma. Arch Ophthalmol
in combination with PGAs or β-adrenoceptor antag- 2002 Jun; 120 (6): 701-13
onists when these are used as initial monotherapy. 16. Campbell DG. Pigmentary dispersion and glaucoma: a new
theory. Arch Ophthalmol 1979 Sep; 97 (9): 1667-72
Reduction in IOP remains a cornerstone of glauco- 17. Hollows FC, Graham PA. Intra-ocular pressure, glaucoma, and
ma therapy. Future directions of research, including glaucoma suspects in a defined population. Br J Ophthalmol
1966 Oct; 50 (10): 570-86
neuroprotection, enhanced ocular blood flow and 18. Arkell SM, Lightman DA, Sommer A, et al. The prevalence of
genetic therapy, offer exciting potential avenues for glaucoma among Eskimos of northwest Alaska. Arch
the management of this debilitating disease. Ophthalmol 1987 Apr; 105 (4): 482-5
19. McLaren JW, Trocme SD, Relf S, et al. Rate of flow of aqueous
humor determined from measurements of aqueous flare. Invest
Ophthalmol Vis Sci 1990 Feb; 31 (2): 339-46
Acknowledgements 20. Becker B. The decline in aqueous secretion and outflow facility
with age. Am J Ophthalmol 1958 Nov; 46 (5): 731-6
Supported in part by an unrestricted research grant from 21. Hayashi M, Yablonski ME, Boxrud C, et al. Decreased forma-
tion of aqueous humour in insulin-dependent diabetic patients.
Research to Prevent Blindness, Inc., New York, NY, USA. Br J Ophthalmol 1989 Aug; 73 (8): 621-3
The authors have no conflicts of interest that are directly 22. Pederson JE. Ocular hypotony. Trans Ophthalmol Soc U K
relevant to the content of this review. 1986; 105 (Pt 2): 220-6

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
18 Marquis & Whitson

23. Diestelhorst M, Krieglstein GK. The effect of the water-drink- 46. Mandell AI, Stentz F, Kitabchi AE. Dipivalyl epinephrine: a
ing test on aqueous humor dynamics in healthy volunteers. new pro-drug in the treatment of glaucoma. Ophthalmology
Graefes Arch Clin Exp Ophthalmol 1994 Mar; 232 (3): 145-7 1978 Mar; 85 (3): 268-75
24. Reddy VN. Dynamics of transport systems in the eye. Invest 47. Kass MA, Mandell AI, Goldberg I, et al. Dipivefrin and epi-
Ophthalmol Vis Sci 1979 Oct; 18 (10): 1000-18 nephrine treatment of elevated intraocular pressure: a compar-
25. Bill A. Blood circulation and fluid dynamics of the eye. Physiol ative study. Arch Ophthalmol 1979 Oct; 97 (10): 1865-6
Rev 1975 Jul; 55 (3): 383-417 48. Lee DA, Topper JE, Brubaker RF. Effect of clonidine on aque-
26. Toris CB, Pederson JE. Aqueous humor dynamics in experi- ous humor flow in normal human eyes. Exp Eye Res 1984
mental iridocyclitis. Invest Ophthalmol Vis Sci 1987 Mar; 28 Mar; 38 (3): 239-46
(3): 477-81 49. Toris CB, Tafoya ME, Camras CB, et al. Effects of
27. Soltau JB, Zimmerman TJ. Changing paradigms in the medical apraclonidine on aqueous humor dynamics in human eyes.
treatment of glaucoma. Surv Ophthalmol 2002; 47 Suppl. 1: Ophthalmology 1995 Mar; 102 (3): 456-61
S2-5 50. Robin AL. Short-term effects of unilateral 1% apraclonidine
28. Realini T, Fechtner RD. 56,000 ways to treat glaucoma. Oph- therapy. Arch Ophthalmol 1988 Jul; 106 (7): 912-5
thalmology 2002 Nov; 109 (11): 1955-6 51. Butler P, Mannschreck M, Lin S, et al. Clinical experience with
29. Diggory P, Franks W. Glaucoma: systemic side effects of topi- the long-term use of 1% apraclonidine: incidence of allergic
cal medical therapy: a common and under recognized problem. reactions. Arch Ophthalmol 1995 Mar; 113 (3): 293-6
J R Soc Med 1994 Oct; 87 (10): 575-6 52. Toris CB, Gleason ML, Camras CB, et al. Effects of
30. Whitson JT, Love R, Brown RH, et al. The effect of reduced brimonidine on aqueous humor dynamics in human eyes. Arch
eyedrop size and eyelid closure on the therapeutic index of Ophthalmol 1995 Dec; 113 (12): 1514-7
phenylephrine. Am J Ophthalmol 1993 Mar; 115 (3): 357-9 53. Schuman JS. Clinical experience with brimonidine 0.2% and
31. von Weber A. Die Ursache des Glaukoms. Albr Graefes Arch timolol 0.5% in glaucoma and ocular hypertension. Surv
Ophthalmol 1877; 23: 91-4 Ophthalmol 1996 Nov; 41 Suppl. 1: S27-37
32. Drance SM, Nash PA. The dose response of human intraocular 54. Schuman JS, Horwitz B, Choplin NT, et al. A 1-year study of
pressure to pilocarpine. Can J Ophthalmol 1971 Jan; 6 (1): brimonidine twice daily in glaucoma and ocular hypertension:
9-13 a controlled, randomized, multicenter clinical trial. Chronic
Brimonidine Study Group. Arch Ophthalmol 1997 Jul; 115
33. Crawford K, Kaufman PL. Pilocarpine antagonizes pros-
(7): 847-52
taglandin F2 alpha-induced ocular hypotension in monkeys:
evidence for enhancement of uveoscleral outflow by pros- 55. Serle JB. A comparison of the safety and efficacy of twice daily
taglandin F2 alpha. Arch Ophthalmol 1987 Aug; 105 (8): brimonidine 0.2% versus betaxolol 0.25% in subjects with
1112-6 elevated intraocular pressure: the Brimonidine Study Group
III. Surv Ophthalmol 1996 Nov; 41 Suppl. 1: S39-47
34. Barsam PC. Comparison of the effects of pilocarpine and
echothiophate on intraocular pressure and outflow facility. Am 56. Wilkerson M, Lewis RA, Shields MB. Follicular conjunctivitis
J Ophthalmol 1972 May; 73 (5): 742-9 associated with apraclonidine. Am J Ophthalmol 1991 Jan;
111 (1): 105-6
35. Ellis PP, Esterdahl M. Echothiophate iodide therapy in children:
effect upon blood cholinesterase levels. Arch Ophthalmol 57. Stewart WC, Ritch R, Shin DH, et al. The efficacy of
1967 May; 77 (5): 598-601 apraclonidine as an adjunct to timolol therapy. Apraclonidine
Adjunctive Therapy Study Group. Arch Ophthalmol 1995
36. Thoft RA. Incidence of lens changes in patients treated with Mar; 113 (3): 287-92
echothiophate iodide. Arch Ophthalmol 1968 Sep; 80 (3): 317-
58. Stewart WC, Laibovitz R, Horwitz B, et al. A 90-day study of
20
the efficacy and side effects of 0.25% and 0.5% apraclonidine
37. O’Brien CS, Swan KD. Carbaminoylcholinechloride in the vs. 0.5% timolol (Apraclonidine Primary Therapy Study
treatment of glaucoma simplex. Arch Ophthalmol 1942 Feb; Group). Arch Ophthalmol 1996 Aug; 114 (8): 938-42
27 (2): 253-7
59. Katz LJ. Twelve-month evaluation of brimonidine-purite versus
38. Reichert RW, Shields MB, Stewart WC. Intraocular pressure brimonidine in patients with glaucoma or ocular hypertension.
response to replacing pilocarpine with carbachol. Am J J Glaucoma 2002 Apr; 11 (2): 119-26
Ophthalmol 1988 Dec; 106 (6): 747-8 60. Becker B. Decrease in intraocular pressure in man by a carbonic
39. Townsend DJ, Brubaker RF. Immediate effect of epinephrine on anhydrase inhibitor (Diamox). Am J Ophthalmol 1954 Jan; 37
aqueous formation in the normal human eye as measured by (1): 13-7
fluorophotometry. Invest Ophthalmol Vis Sci 1980 Mar; 19 61. Dailey RA, Brubaker RF, Bourne WM. The effects of timolol
(3): 256 maleate and acetazolamide on the rate of aqueous formation in
40. Nagataki S, Brubaker RF. Early effect of epinephrine on aque- normal human subjects. Am J Ophthalmol 1982 Feb; 93 (2):
ous formation in the normal human eye. Ophthalmology 1981 232-7
Mar; 88 (3): 278-82 62. Fraunfelder FT, Meyer SM, Bagby Jr GC, et al. Hematologic
41. Becker B, Pettit TH, Gay AJ. Topical epinephrine therapy of reactions to carbonic anhydrase inhibitors. Am J Ophthalmol
open angle glaucoma. Arch Ophthalmol 1961 Aug; 66 (2): 1985 Jul; 100 (1): 79-81
219-25 63. Strahlman E, Tipping R, Vogel R. A double-masked, random-
42. van Alphen GW. The adrenergic receptors of the intraocular ized 1-year study comparing dorzolamide (Trusopt), timolol,
muscles of the human eye. Invest Ophthalmol 1976 Jun; 15 and betaxolol. International Dorzolamide Study Group. Arch
(6): 502-5 Ophthalmol 1995 Aug; 113 (8): 1009-16
43. Kolker AE, Becker B. Epinephrine maculopathy. Arch 64. Boyle JE, Ghosh K, Gieser DK, et al. A randomized trial
Ophthalmol 1968 May; 79 (5): 552-62 comparing the dorzolamide-timolol combination given twice
44. Cashwell LF, Shields MB, Reed JW. Adrenochrome pigmenta- daily to monotherapy with timolol and dorzolamide. Ophthal-
tion. Arch Ophthalmol 1977 Mar; 95 (3): 514-5 mology 1998 Oct; 105 (10): 1945-51
45. McCarthy RW, LeBlanc R. A ‘black cornea’ secondary to 65. Kimal AM, Topalkara A, Guier C. Additive effect of latanoprost
topical epinephrine. Can J Ophthalmol 1976 Oct; 11 (4): and dorzolamide in patients with elevated intraocular pressure.
336-40 Int Ophthalmol 1998 Jan; 22 (1): 37-42

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
Pharmacological Therapy of Glaucoma 19

66. Stewart WC, Sharpe ED, Harbin TS, et al. Brimonidine 0.2% 84. Geyer O, Lazar M, Novack GD, et al. Levobunolol compared
versus dorzolamide 2% each given three times daily to reduce with timolol: a four-year study. Br J Ophthalmol 1988 Dec; 72
intraocular pressure. Am J Ophthalmol 2000 Jun; 129 (6): (12): 892-6
723-7 85. Scoville B, Mueller B, White BG, et al. A double-masked
67. Whitson JT, Henry C, Hughes B, et al. Comparison of the safety comparison of carteolol and timolol in ocular hypertension.
and efficacy of dorzolamide 2% and brimonidine 0.2% in Am J Ophthalmol 1988 Feb; 105 (2): 150-4
patients with glaucoma or ocular hypertension. J Glaucoma 86. Stewart WC, Shields MB, Allen RC, et al. A 3-month compari-
2004 Apr; 13 (2): 168-73 son of 1% and 2% carteolol and 0.5% timolol in open-angle
68. Silver LH. The efficacy and safety of brinzolamide 1% glaucoma. Graefes Arch Clin Exp Ophthalmol 1991 Mar; 229
ophthalmic suspension (Azopt) as a primary therapy in patients (3): 258-61
with open-angle glaucoma or ocular hypertension. Brinzo- 87. Freedman SF, Freedman NJ, Shields MB, et al. Effects of ocular
lamide Primary Therapy Study Group. Surv Ophthalmol 2000 carteolol and timolol on plasma high-density lipoprotein cho-
Jan; 44 Suppl. 2: S155-62 lesterol level. Am J Ophthalmol 1993 Nov; 116 (5): 600-11
69. Barnebey H, Kwok SY. Patients’ acceptance of a switch from 88. Serle JB, Lustgarten JS, Podos SM. A clinical trial of metipra-
dorzolamide to brinzolamide for the treatment of glaucoma in nolol, a noncardioselective beta-adrenergic antagonist, in ocu-
a clinical practice setting. Clin Ther 2000 Oct; 22 (10): lar hypertension. Am J Ophthalmol 1991 Sep; 112 (3): 302-7
1204-12 89. Muller O, Knobel HR. Effectiveness and tolerance of metipra-
70. Sugrue MF, Mallorga P, Schwam H, et al. A comparison of L- nolol: results of a multi-center long-term study in Switzerland.
671,152 and MK-927, two topically effective ocular hypoten- Klin Monatsbl Augenheilkd 1986 Jan; 188 (1): 62-3
sive carbonic anhydrase inhibitors, in experimental animals. 90. Krieglstein GK, Novack GD, Voepel E, et al. Levobunolol and
Curr Eye Res 1990 Jun; 9 (6): 607-15 metipranolol: comparative ocular hypotensive efficacy, safety,
71. Konowal A, Morrison JC, Brown SVL, et al. Irreversible corne- and comfort. Br J Ophthalmol 1987 Apr; 71 (4): 250-3
al decompensation in patients treated with topical dorzo- 91. Coakes RL, Mackie IA, Seal DV. Effects of long-term treatment
lamide. Am J Ophthalmol 1999 Apr; 127 (4): 403-6 with timolol on lacrimal gland function. Br J Ophthalmol 1981
72. Silver LH. Clinical efficacy and safety of brinzolamide (Azopt), Sep; 65 (9): 603-5
a new topical carbonic anhydrase inhibitor for primary open- 92. Akingbehin T, Villada JR. Metipranolol-associated granuloma-
angle glaucoma and ocular hypertension. Am J Ophthalmol tous anterior uveitis. Br J Ophthalmol 1991 Sep; 75 (9):
1998 Sep; 126 (3): 400-8 5519-23
73. Coakes RL, Brubaker RF. The mechanism of timolol in lower- 93. Melles RB, Wong IG. Metipranolol-associated granulomatous
ing intraocular pressure in the normal eye. Arch Ophthalmol iritis. Am J Ophthalmol 1994 Dec; 118 (6): 712-5
1978 Nov; 96 (11): 2045-8 94. McMahon CD, Shaffer RN, Hoskins HDJ, et al. Adverse effects
74. Steinert RF, Thomas JV, Boger III WP. Long-term drift and experienced by patients taking timolol. Am J Ophthalmol 1979
continued efficacy after multiyear timolol therapy. Arch Oct; 88 (4): 736-8
Ophthalmol 1981 Jan; 99 (1): 100-3 95. Van Buskirk EM. Adverse reactions from timolol administra-
75. Kobelt G, Jonsson L, Gerdtham U, et al. Direct costs of tion. Ophthalmology 1980 May; 87 (5): 447-50
glaucoma management following initiation of medical ther- 96. Velde TM, Kaiser FE. Ophthalmic timolol treatment causing
apy: a simulation model based on an observational study of altered hypoglycemic response in a diabetic patient. Arch
glaucoma treatment in Germany. Graefes Arch Clin Exp Intern Med 1983 Aug; 143 (8): 1627
Ophthalmol 1998 Nov; 236 (11): 811-21 97. Coppeto JR. Timolol-associated myasthenia gravis. Am J
76. Topper JE, Brubaker RF. Effects of timolol, epinephrine, and Ophthalmol 1984 Aug; 98 (2): 244-5
acetazolamide on aqueous flow during sleep. Invest 98. Fraunfelder FT. Interim report: national registry of possible
Ophthalmol Vis Sci 1985 Oct; 26 (10): 1315-9 drug-induced ocular side effects. Ophthalmology 1980 Feb; 87
77. Zimmerman TJ, Kaufman HE. Timolol: a beta-adrenergic (2): 87-90
blocking agent for the treatment of glaucoma. Arch 99. Reiss GR, Brubaker RF. The mechanism of betaxolol, a new
Ophthalmol 1977 Apr; 95 (4): 601-4 ocular hypotensive agent. Ophthalmology 1983 Nov; 90 (11):
78. Wilson RP, Kanal N, Spaeth GL. Timolol: its effectiveness in 1369-72
different types of glaucoma. Ophthalmology 1979 Jan; 86 (1): 100. Caldwell Dr, Salisbury CR, Guzek JP. Effects of topical betax-
43-50 olol in ocular hypertensive patients. Arch Ophthalmol 1984
79. Katz IM, Berger ET. Effects of iris pigmentation on response of Apr; 102 (4): 539-40
ocular pressure to timolol. Surv Ophthalmol 1979 May; 23 (6): 101. Feghali JG, Kaufman PL. Decreased intraocular pressure in the
395-8 hypertensive human eye with betaxolol, a beta 1-adrenergic
80. Shedden A, Laurence J, Tipping R, et al. Efficacy and tolerabili- antagonist. Am J Ophthalmol 1985 Dec; 100 (6): 777-82
ty of timolol maleate ophthalmic gel-forming solution in adults 102. Collignon-Brach J. Long-term effect of ophthalmic beta-adre-
with primary open-angle glaucoma or ocular hypertension: a noceptor antagonists on intraocular pressure and retinal sensi-
six-month, double-masked, multicenter study. Clin Ther 2001 tivity in primary open-angle glaucoma. Curr Eye Res 1992
Mar; 23 (3): 440-50 Jan; 11 (1): 1-3
81. Stewart WC, Sharpe ED, Stuart JA, et al. The safety and 103. Messmer C, Flammer J, Stumpfig D. Influence of betaxolol and
efficacy of timolol 0.5% in xanthum gum versus timolol gel timolol on the visual fields of patients with glaucoma. Am J
forming solution 0.5%. Curr Eye Res 2002 May 24 (5): 387- Ophthalmol 1991 Dec; 112 (6): 678-1
391 104. Hoste AM, Sys SU. The relaxant action of betaxolol on isolated
82. Derick RJ, Robin AL, Tielsch J, et al. Once-daily versus twice- bovine retinal microarteries. Curr Eye Res 1994 Jul; 13 (7):
daily levobunolol (0.5%) therapy: a cross-over study. Ophthal- 483-7
mology 1992 Dec; 99 (3): 424-9 105. Hoste AM. Ca2+ channel blocking activity of propranolol and
83. Boozman FW, Carriker R, Foerster R, et al. Long-term evalua- betaxolol in isolated bovine retinal microartery. J Cardiovasc
tion of 0.25% levobunolol and timolol for therapy for elevated Pharmacol 1998 Sep; 32 (3): 390-6
intraocular pressure. Arch Ophthalmol 1988 May; 106 (5): 106. Schoene RB, Sharpe ED, Harbin TS, et al. Effects of topical
614-8 betaxolol, timolol, and placebo on pulmonary function in

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
20 Marquis & Whitson

asthmatic bronchitis. Am J Ophthalmol 1984 Jan; 97 (1): receptor agonist. J Ocular Pharmacol Ther 2001 Oct; 17 (5):
86-92 421-32
107. Harris LS, Greenstein SH, Bloom AF. Respiratory difficulties 127. Netland PA, Landry T, Sullivan EK, et al. Travoprost compared
with betaxolol. Am J Ophthalmol 1986 Aug; 102 (2): 274-5 with latanoprost and timolol in patients with open-angle
108. Roholt PC. Betaxolol and restrictive airway disease: case report. glaucoma or ocular hypertension. Am J Ophthalmol 2001 Oct;
Arch Ophthalmol 1987 Sep; 105 (9): 1172 132 (4): 472-84
109. Nelson WL, Kuritsky JN. Early postmarketing surveillance of 128. Fellman RL, Sullivan EK, Ratliff M, et al. Comparison of
betaxolol hydrochloride, Sept 1985-Sept 1986 [letter]. Am J travoprost 0.0015% and 0.004% with timolol 0.5% in patients
Ophthalmol 1987 Apr; 103 (4): 592 with elevated IOP: a six-month, masked, multicenter trial.
110. Ball S. Congestive heart failure from betaxolol: case report. Ophthalmology 2002 May; 109 (5): 998-1008
Arch Ophthalmol 1987 Mar; 105 (3): 320 129. Goldberg I, Cunha-Vaz J, Jakobsen JE, et al. Comparison of
111. Lynch MG, Whitson JT, Brown RH, et al. Topical beta-blocker topical travoprost eye drops given once daily and timolol 0.5%
therapy and central nervous system side effects: a preliminary given twice daily in patients with open-angle glaucoma or
study comparing betaxolol and timolol. Arch Ophthalmol 1988 ocular hypertension. J Glaucoma 2002 Oct; 10 (5): 414-22
Jul; 106 (7): 908-11 130. Orengo-Nania S, Landry T, Von Tress M, et al. Evaluation of
112. Asrani S, Zeimer R, Wilensky J, et al. Large diurnal fluctuations travoprost as adjunctive therapy in patients with uncontrolled
in IOP are an independent risk factor in patients with intraocular pressure while using timolol 0.5%. Am J
glaucoma. J Glaucoma 2000 Apr; 9 (2): 134-42 Ophthalmol 2001 Dec; 132 (6): 860-8
113. Bergea B, Bodin L, Svedbergh B. Impact of intraocular pressure 131. Dubiner HB, Sircy MD, Landry T, et al. Comparison of the
regulation on visual fields in open-angle glaucoma. Ophthal- diurnal ocular hypotensive efficacy of travoprost and lata-
mology 1999 May; 106 (5): 997-1004 noprost over a 44-hour period in patients with elevated in-
114. Mishima HK, Kiuchi Y, Takamatsu M, et al. Circadian intraocu- traocular pressure. Clin Ther 2004 Jan; 26 (1): 84-91
lar pressure management with latanoprost: diurnal and noctur- 132. Whitson JT. Travoprost-a new prostaglandin analogue for the
nal intraocular pressure reduction and increased uveoscleral treatment of glaucoma. Expert Opin Pharmacother 2002 Jul; 3
outflow. Surv Ophthalmol 1997 Feb; 41 Suppl. 2: S139-44 (7): 965-77
115. Hylton C, Robin AL. Update on prostaglandin analogues. Curr 133. Woodward DF, Krauss AH, Chen J, et al. Pharmacological
Opin Ophthalmol 2003 Apr; 14 (2): 65-9 characterization of a novel antiglaucoma agent, Bimatoprost
116. Ziai N, Dolan JW, Kacere RD, et al. The effects on aqueous (AGN 192024). J Pharmacol Exp Ther 2003 May; 305 (2):
dynamics of PhXA41, a new prostaglandin F2α analogue, 772-85
after topical application in normal and ocular hypertensive
134. Hellberg MR, Ke TL, Haggard K, et al. The hydrolysis of the
human eyes. Arch Ophthalmol 1993 Oct; 111 (1): 1351-8
prostaglandin analogue prodrug bimatoprost to 17-phenyl-tri-
117. Parrish RK, Palmberg P, Sheu WP. A comparison of lata- nor PGF2alpha by human and rabbit ocular tissue. J Ocul
noprost, bimatoprost, and travoprost in patients with elevated Pharmacol Ther 2003 Apr; 19 (2): 97-103
intraocular pressure: a 12-week, randomized, masked-evalu-
ator multicenter study. Am J Ophthalmol 2003 May; 135 (5): 135. Brubaker RF. Mechanism of action of bimatoprost (Lumigan).
688-703 Surv Ophthalmol 2001 May; 45 (4): S347-51
118. Camras CB. Comparison of latanoprost and timolol in patients 136. Sherwood M, Brandt J. Six-month comparison of bimatoprost
with ocular hypertension and glaucoma: a six-month masked, q.d. and b.i.d. with timolol b.i.d. in patients with elevated
multicenter trial in the United States. The United States Lata- intraocular pressure. Surv Ophthalmol 2001 May; 45 (4):
noprost Study Group. Ophthalmology 1996 Jan; 103 (1): 138- S361-8
47 137. Gandolfi S, Simmons ST, Sturm R, et al. Three-month compari-
119. Watson P, Stjernschantz J. A six-month, randomized, double- son of bimatoprost and latanoprost in patients with glaucoma
masked study comparing latanoprost with timolol in open- and ocular hypertension. Adv Ther 2001 May-Jun; 18 (3): 110-
angle glaucoma and ocular hypertension. The Latanoprost 21
Study Group. Ophthalmology 1996 Jan; 103 (1): 126-37 138. Noecker RS, Dirks MS, Choplin NT, et al. A six-month random-
120. Mishima HK, Masuda K, Kitazawa Y, et al. A comparison of ized clinical trial comparing the intraocular pressure-lowering
latanoprost and timolol in primary open-angle glaucoma and efficacy of bimatoprost and latanoprost in patients with ocular
ocular hypertension: a 12-week study. Arch Ophthalmol 1996 hypertension or glaucoma. Am J Ophthalmol 2003 Jan; 135
Aug; 114 (8): 929-32 (1): 55-63
121. Orzalesi N, Rossetti L, Invernizzi T, et al. Effect of timolol, 139. Coleman AL, Lerner F, Bernstein P, et al. A 3-month random-
latanoprost, and dorzolamide on circadian IOP in glaucoma or ized controlled trial of bimatoprost (LUMIGAN) versus com-
ocular hypertension. Invest Ophthalmol Vis Sci 2000 Aug; 41 bined timolol and dorzolamide (Cosopt) in patients with
(9): 2566-73 glaucoma or ocular hypertension. Ophthalmology 2003 Dec;
122. O’Donoghue EP. A comparison of latanoprost and dorzolamide 110 (12): 2362-8
in patients with glaucoma and ocular hypertension: a 3-month 140. Strohmaier K, Snyder E, DuBiner H, et al. The efficacy and
randomized study. Ireland Latanoprost Study Group. Br J safety of the dorzolamide-timolol combination versus the con-
Ophthalmol 2000 Jun; 84 (6): 579-82 comitant administration of its components. Ophthalmology
123. Dubiner HB, Mroz M, Shapiro AM, et al. A comparison of the 1998 Oct; 105 (10): 1936-44
efficacy and tolerability of brimonidine and latanoprost in 141. Cantor LB. An update on bimatoprost in glaucoma therapy.
adults with open-angle glaucoma or ocular hypertension: a Expert Opin Pharmacother 2002 Dec; 3 (12): 1753-62
three-month, multicenter, randomized, double-masked, paral- 142. Nordmann JP, Mertz B, Yannoulis NC, et al. A double-masked
lel-group trial. Clin Ther 2001 Dec; 23 (12): 1969-83 randomized comparison of the efficacy and safety of unopros-
124. Data on file, Pfizer Ophthalmics Inc., New York, 2003 tone with timolol and betaxolol in patients with primary open-
125. Morgan PV, Proniuk S, Blanchard J, et al. Effect of temperature angle glaucoma including pseudoexfoliation glaucoma or ocu-
and light on the stability of latanoprost and its clinical rele- lar hypertension: 6-month data. Am J Ophthalmol 2002 Jan;
vance. J Glaucoma 2001 Oct; 10 (5): 401-5 133 (1): 1-10
126. Hellberg MR, Sallee VL, McLaughlin MA, et al. Preclinical 143. Jampel HD, Bacharach J, Sheu WP, et al. Randomized clinical
efficacy of travoprost, a potent and selective FP prostaglandin trial of latanoprost and unoprostone in patients with elevated

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)
Pharmacological Therapy of Glaucoma 21

intraocular pressure. Am J Ophthalmol 2002 Dec; 134 (6): tion on the trabecular meshwork of human glaucomatous eyes.
863-71 Graefes Arch Clin Exp Ophthalmol 1990 Jan; 228 (1): 90-100
144. Hommer A, Kapik B, Shams N. Unoprostone as adjunctive 162. Wise JB. Long-term control of adult open angle glaucoma by
therapy to timolol: a double masked randomized study versus argon laser treatment. Ophthalmology 1981 Mar; 88 (3): 197-
brimonidine and dorzolamide. Br J Ophthalmol 2003 May; 87 202
(5): 592-8
163. Wilensky JT, Jampol LM. Laser therapy for open angle
145. Hedner J, Svedmyr N, Lunde H, et al. The lack of respiratory glaucoma. Ophthalmology 1981 Mar; 88 (3): 213-7
effects of the ocular hypotensive drug latanoprost in patients
with moderate-steroid treated asthma. Surv Ophthalmol 1997 164. Tuulonen A, Niva AK, Alanko HI. A controlled five-year fol-
Feb; 41 Suppl. 2: S111-5 low-up study of laser trabeculoplasty as primary therapy for
open-angle glaucoma. Am J Ophthalmol 1987 Oct; 104 (4):
146. Wistrand PJ, Stjernschantz J, Olsson K. The incidence and time-
334-8
course of latanoprost-induced iridial pigmentation as a func-
tion of eye color. Surv Ophthalmol 1997 Feb; 41 Suppl. 2: 165. Frucht J, Bishara S, Ticho U. Early intraocular pressure re-
S129-38 sponse following laser trabeculoplasty. Br J Ophthalmol 1985
147. Fechtner RD, Khouri AS, Zimmerman TJ, et al. Anterior uveitis Oct; 69 (10): 771-3
associated with latanoprost. Am J Ophthalmol 1998 Jul; 126 166. Thomas JV, Simmons RJ, Belcher III CD. Argon laser
(1): 37-41 trabeculoplasty in the presurgical glaucoma patient. Ophthal-
148. Dios Castro E, Maquet Dusart JA. Latanoprost-associated recur- mology 1982 Mar; 89 (3): 187-97
rent herpes simplex keratitis. Arch Soc Esp Oftalmol 2000 167. Hoskins Jr HD, Hetherington Jr J, Minckler DS, et al. Complica-
Nov; 75 (11): 775-8 tions of laser trabeculoplasty. Ophthalmology 1983 Jul; 90 (7):
149. Ayyala RS, Cruz DA, Margo CE, et al. Cystoid macular edema 796-9
associated with latanoprost in aphakic and pseudophakic eyes. 168. Latina MA, Sibayan SA, Shin DH, et al. Q-switched 532-nm
Am J Ophthalmol 1998 Oct; 126 (4): 602-4 Nd:YAG laser trabeculoplasty (selective laser trabeculoplas-
150. Callanan D, Fellman RL, Savage JA. Latanoprost-associated ty): a multicenter, pilot, clinical study. Ophthalmology 1998
cystoid macular edema. Am J Ophthalmol 1998 Jul; 126 (1): Nov; 105 (11): 2082-8
134-5
169. Kramer TR, Noecker RJ. Comparison of the morphologic
151. Bucci MG. Intraocular pressure-lowering effects of latanoprost changes after selective laser trabeculoplasty and argon laser
monotherapy versus latanoprost or pilocarpine in combination trabeculoplasty in human eye bank eyes. Ophthalmology 2001
with timolol: a randomized, observer-masked multicenter Apr; 108 (4): 773-9
study in patients with open-angle glaucoma. Italian Lata-
noprost Study Group. J Glaucoma 1999 Feb; 8 (1): 24-30 170. Damji KF, Shah KC, Rock WJ, et al. Selective laser
152. Higginbotham EJ, Feldman R, Stiles M, et al. Latanoprost and trabeculoplasty vs argon laser trabeculoplasty: a prospective
timolol combination therapy vs monotherapy. Arch randomized clinical trial. Br J Ophthalmol 1999 Jun; 83 (6):
Ophthalmol 2002 Jul; 120 (7): 915-22 718-22
153. Netland PA, Michael M, Rosner SA, et al. Brimonidine purite 171. Cairns JE. Trabeculectomy: preliminary report of a new method.
and bimatoprost compared with timolol and latanoprost in Am J Ophthalmol 1968 Oct; 66 (4): 673-9
patients with glaucoma and ocular hypertension. Adv Ther 172. Ruderman JM, Welch DB, Smith MF, et al. A randomized study
2003 Jan-Feb; 20 (1): 20-30 of 5-fluorouracil and filtration surgery. Am J Ophthalmol 1987
154. Chiselita D, Apatachioae I, Poiata I. The ocular hypotensive Sep; 104 (3): 218-24
effect of the combination of latanoprost and dorzolamide. 173. Palmer SS. Mitomycin as an adjunct chemotherapy with
Oftalmologia 1999 Jan; 46 (1): 39-45 trabeculectomy. Ophthalmology 1991 Mar; 98 (3): 317-21
155. O’Connor DJ, Martone JF, Mead A. Additive intraocular pres-
174. Zimmerman TJ, Kooner KS, Ford VJ, et al. Effectiveness of
sure lowering effect of various medications with latanoprost.
nonpenetrating trabeculectomy in aphakic patients with
Am J Ophthalmol 2002 Jun; 133 (6): 836-7
glaucoma. Ophthalmic Surg 1984 Jan; 15 (1): 44-50
156. Sall KN, Greff LJ, Johnson-Pratt LR, et al. Dorzolamide/timolol
combination versus concomitant administration of brimoni- 175. Chiselita D. Non-penetrating deep sclerectomy versus
dine and timolol: six-month comparison of efficacy and tolera- trabeculectomy in primary open angle glaucoma surgery. Eye
bility. Ophthalmology 2003 Mar; 110 (3): 615-24 2001 Apr; 15 (Pt 2): 197-201
157. Orzalesi N, Rossetti L, Bottoli A, et al. The effect of latanoprost, 176. Whitson JT. Recent developments in glaucoma drainage im-
brimonidine and a fixed combination of timolol and dorzo- plant surgery. Int Ophthalmol Clin 1999 Summer; 39 (3): 43-
lamide on circadian intraocular pressure in patients with 55
glaucoma or ocular hypertension. Arch Ophthalmol 2003 Apr; 177. deRoetth Jr A. Cryosurgery for the treatment of advanced
121 (4): 453-7 chronic simple glaucoma. Am J Ophthalmol 1966 Mar; 61 (3):
158. Airaksinen PJ, Valkonen R, Stenborg T, et al. A double-masked 443-50
study of timolol and pilocarpine combined. Am J Ophthalmol 178. Albaugh CH, Dunphy EB. Cyclodiathermy. Arch Ophthalmol
1987 Dec; 104 (6): 587-90 1942 Mar; 27 (3): 543-57
159. Scharrer A, Ober M. Metipranolol 0.1% and pilocarpine 2% as a
fixed combination compared to each substance alone in the 179. Peyman GA, Naguib KS, Gaasterland D. Transscleral applica-
treatment of glaucoma: a controlled, randomized clinical study tion of a semiconductor diode laser. Lasers Surg Med 1990; 10
comparing the intraindividual effects and tolerance. Klin (6): 569-75
Monatsbl Augenheilkd 1986 Dec; 189 (6): 450-5
160. Wise JB, Witter SL. Argon laser therapy for open-angle
glaucoma: a pilot study. Arch Ophthalmol 1979 Feb; 97 (2): Correspondence and offprints: Dr Jess T. Whitson, Depart-
319-22 ment of Ophthalmology, UT Southwestern Medical Center,
161. Babizhayev MA, Brodskaya MW, Mamedov NG, et al. Clinical, 5323 Harry Hines Blvd., Dallas, TX 75390-9057, USA.
structural and molecular phototherapy effects of laser irradia- E-mail: Jess.Whitson@UTSouthwestern.edu

© 2005 Adis Data Information BV. All rights reserved. Drugs Aging 2005; 22 (1)

You might also like