Open Journal of Ophthalmology, 2012, 2, 71-77
http://dx.doi.org/10.4236/ojoph.2012.23015 Published Online August 2012 (http://www.SciRP.org/journal/ojoph)
71
Childhood Glaucoma: An Overview
Parul Singh, Yogesh Kumar, Manoj Tyagi, Krishna Kuldeep, Parmeshwari Das Sharma
Department of Ophthalmology, Veer Chandra Singh Garhwali Government Medical Sciences and Research Institute, Srinagar, India.
Email: parulophtha@gmail.com
Received March 16th, 2012; revised April 27th, 2012; accepted May 28th, 2012
ABSTRACT
Several types of childhood glaucoma exist, and the terminology is based on the time of onset of disease and its potential
cause. Though childhood glaucoma occurs less commonly than adults but can lead to permanent visual damage due to
amblyopia, optic neuropathy or refractive error. A detailed evaluation should be done to establish diagnosis. Medical
therapy has a limited role and surgery remains main modality for treatment. Childhood glaucoma is a treatable disease,
if early diagnosis is established and therapeutic intervention done in time. In children with low vision efforts should be
there to maintain residual vision and visual rehabilitation with low vision aids should be done.
Keywords: Childhood Glaucoma; Goniotomy; Trabeculotomy
1. Introduction
Glaucoma is less common in children than in adults. Al-
though glaucoma can lead to permanent visual damage at
any age, the consequences of the disease are more often
severe in children due to additional damage that can
happen to the developing visual system. Associated am-
blyopia and secondary refractive errors are common.
Primary congenital glaucoma (PCG) is hereditary child-
hood glaucoma secondary to abnormal development of
the filtration angle. Nonetheless, most ophthalmologists
usually encounter the wide range of secondary forms of
glaucoma in this age group. Glaucoma surgery has dras-
tically improved the visual prognosis of children afflicted
with glaucoma. However, late diagnosis of the condition
can result in permanent and severe visual morbidity. Its
affliction of young children makes glaucoma control a
life-long goal requiring motivation and perseverance by
patients, their parents and doctors. This article is in-
tended to provide an overview of the disease at genetic
levels, newer technological tools assisting in diagnosis,
IOP lowering medications and refined surgical tech-
niques.
1.1. Definition and Terminology
Relating to age of onset
1) Congenital glaucoma: The glaucoma that exists at
birth, and usually before birth;
2) Infantile glaucoma: Glaucoma that occurs from
birth until 3 years of age;
3) Juvenile glaucoma: Occurs after the age of three to
teenage years.
Relating to deve lopment pattern
1) Developmental glaucoma: This term broadly en-
compasses all glaucomas resulting from abnormal de-
velopment of the aqueous outflow system. This may or
may not be associated with systemic anomaly;
2) Primary developmental glaucoma: This is the glau-
coma resulting from maldevelopment of aqueous outflow
system;
3) Secondary developmental glaucoma: This refers to
glaucoma resulting from damage to aqueous outflow
system due to maldevelopment of some other portion of
the eye, e.g. eye with microspherophakia or dislocated
lens.
Relating to anatomy
Hoskins classified developmental glaucoma anatomi-
cally by the structures involved:
1) Trabecular meshwork: Trabeculodysgenesis;
2) Iris and trabecular meshwork: Iridotrabeculodys-
genesis;
3) Cornea and trabecular meshwork: Corneotrabecu-
lodysgenesis [1].
1.2. Classification of Congenital and Infantile
Glaucoma
Primary congemital glaucoma (Trabeculodysgenesis)
Secondary congenital glaucoma
1) Iridocorneotrabeculodysgenesis
a) Rieger’s anamoly;
b) Axenfeld anamoly;
c) Peter’s anamoly.
2) Iridotrabeculodysgenesis
Copyright © 2012 SciRes. OJOph
Childhood Glaucoma: An Overview
72
a) Stromal defects: Hypoplasia, hyperplasia;
b) Anomalous iris vessels: Persistence of tunica vas-
culosa lenti;
c) Structural anamolies: Holes, coloboma, aniridia;
Acquired glaucomas
1) Aphakia;
2) Corticosteroid induced;
3) Retinoblastoma;
4) Traumatic;
5) Uveitis.
2. Epidemiology
Primary congenital glaucoma is hereditary with a vari-
able incidence in different populations, but an overall
occurrence of 1 in 10,000 births is seen [2]. A greater
incidence occurs in populations with higher rates of con-
sanguinity [3,4]. Boys are more commonly affected in
United States and Europe with boys to girls ratio of 3:2.
Whereas, in Japan more girls are seen having this condi-
tion [5,6]. The majority (about 75%) of PCG cases are
bilateral and asymmetric expression should be suspected
in clinically apparent unilateral cases. More than 80%
patients present within the first year of life, with 25%
diagnosed in the neonatal period and 60% within the first
6 months of life [7].
3. Genetics
The majority of PCG cases are sporadic but 10% - 40%
are familial with frequent association with consanguinity
[8]. In the most familial cases, transmission is autosomal
recessive with variable expression and penetrance of
40% - 100% [8].
Genetic heterogenicity of PCG confirmed by linkage
studies explain discrepancies like unequal sex distribu-
tion, lower than expected number of affected siblings in
familial cases, and transmission of the disease to success-
sive generations [8]. Three loci for PCG have been found
[8-10]. The initial locus on chromosome 2p21 (GLC3A)
was described in 1995 by Sarfarazi et al. who identified
significant genetic linkage to this region in 11 of 17 turk-
ish families [8,11]. Genetic heterogenicity was confirmed
when a second locus on chromosome 1p36 (GLC3B) was
found [9]. Genetic family analysis identified a third locus
GLC3C on chromosome 14q24.3 [10]. Although three
chromosomal loci have been linked to PCG, only
CYP1B1 in locus GLC3A has been identified [12]. Mu-
tations in these genes have been described as the pre-
dominant cause of PCG in Turkish and Saudi arabian
families [13]. It has been reported that 87% of familial
and 27% sporadic case are due to mutation in this gene
[8]. Approximately 45 mutations of this gene have been
identified and include deletion, insertion, point mutation,
mis-sense, non-sense, frameshift and terminator mutation
[14]. From all mutations studied, frameshift and R390C
homozygous mutations were associated with severe phe-
noltype and very poor prognosis. Primary juvenile glau-
coma can be inherited in an autosomal dominant fashion.
A mutation in the chromosome 1q23-25 region has been
linked to this disease [15,16]. Genetic studies have also
been conducted on few other glaucomas. In Axenfeld,
Rieger anomaly, the gene has been mapped to chromo-
some 6p25 [17]. In aniridia, the most common inheri-
tance pattern is autosomal dominant. The genetic locus
for aniridia has been established as the PAX6 gene, lo-
cated on 11p13 [18].
4. Pathogenesis
The exact mechanism involved in pathophysiology of
primary infantile glaucoma is not known. One theory
said that a pathologic membrane (known as Barkan’s
membrane) [19] covered and blocked the trabecular
meshwork in primary infantile glaucoma. Clinical and
histopathologic observations of the anterior chamber in
infantile glaucoma have revealed that the anatomic rela-
tionship between the iris, trabecular meshwork and
ciliary body are immature. The principal defect in pri-
mary infantile glaucoma is a failure of one or more steps
in the normal development of the anterior chamber angle.
As the genes associated with primary infantile glau-
coma are characterized further and the physiological or
developmental role of the proteins they encode become
better understood; the molecular, cellular and embryo-
logical pathophysiology of this rare disorder will become
clear [12].
Among the secondary glaucomas of childhood, the
underlying pathophysiology is as varied as that in adults.
Occurrence at or shortly after birth indicates a profound
developmental abnormality of anterior chamber angle
whereas, manifestation later usually suggests a different
process.
Secondary open angle also occur in young children.
Both corticosteroid-induced and chronic uveitic glauco-
mas are described [20]. It is difficult to classify the un-
derlying cause of glaucoma, that frequently follows pe-
diatric cataract extraction. Walton examined 65 children
in whom, pre-operative gonioscopy revealed no consis-
tent angle defect but post-operative gonioscopy revealed
filtration angle deformity [21]. Retained lens material
was one of the risk factor known another cause may be
presence of small cornea.
5. Presentation
The signs and symptoms of PCG are variable dependent
on child’s age and severity of glaucoma; and secondary
corneal abnormalities. PCG is characterized by clinical
triad of epiphora, blepharospasm and photophobia, but
Copyright © 2012 SciRes. OJOph
Childhood Glaucoma: An Overview 73
these symptoms are often missed until the more alarming
corneal edema becomes apparent. The corneal edema,
may be subtle, especially in bilateral cases; or profound
with enlarged corneal diameter and globe, breaks in De-
scemet’s membrane (Haab’s striae) and sometimes even
acute hydrops.
Myopia is typical finding in infantile glaucoma. In
older children, astigmatism and progressive axial myopia
cause symptomatic decreased uncorrected visual acuity
and refractive amblyopia.
Optic neuropathy is the most concerning consequence
from glaucoma, because the neuropathy is irreversible
once axonal death has occurred.
While cupping of the optic nerve in glaucoma is gen-
erally a gradual process in older children and adults, it
can occur rapidly in infants. Reversibility of the cupping
with normalization of the intra-ocular pressure (IOP) in
young children occurs due to suspected relative immatur-
ity and elasticity of lamina cribrosa. Less common pre-
senting signs in infantile glaucoma include conjunctivitis,
blepharitis and cellulitis.
6. Diagnosis and Ancillary Testing
It is important to do a complete ophthalmic examination
in a child suspected of glaucoma. This includes IOP
measurement, gonioscopy, optic nerve head examination
and refraction. Check for the child’s ability to fix and
follow; and for the presence of nystagmus. Examination
of the cornea is crucial with respect to size and clarity of
the cornea and the presence of Haab’s striae.
This examination can be done in clinic. With some
practice, IOP can be measured in a conscious, swaddled
infant using perkin’s tonometer or tonopen. Usually IOP
in infants with normal eyes is in the range of 11 - 14 mm
Hg using these devices. The measurement of IOP greater
than 20 mmHg in a calm, resting infant is suspicious for
glaucoma when other signs and symptoms also suggest
the disease. Measurements of IOP undertaken while a
child cries and resists efforts to hold the eye open are
invalid.
Examination of the optic nerve is attempted, because
obvious cupping confirms the diagnosis. Shaffer and
Hetherington noted a cup to disc ratio (C/D ratio) greater
than 0.3 in 68% of 126 eyes affected by primary infantile
glaucoma [22], whereas C/D ratio greater than 0.3 was
found in less than 2.6% of newborns with normal eyes
[23]. A Koeppe infant diagnostic lens offers good visu-
alization of disc using direct ophthalmoscope. Gonio-
scopy can also be performed with it even in a conscious
infant. In the normal newborn eye, the iris usually inserts
posterior to the scleral spur. In PCG, the iris commonly
inserts anteriorly directly into the trabecular meshwork.
This iris insertion is most commonly flat, although con-
cave insertion may be rarely seen.
If the diagnosis of glaucoma is confirmed or strongly
suspected based on clinical examination, an examination
under anaesthesia is required. Unfortunately, most anes-
thetic agents and sedatives have lowering effect on IOP
[24]. Tonometry should be best performed as soon as
possible after induction of anesthesia. Corneal diameters
are measured with a millimeter ruler or caliper and re-
corded. Detailed hand slit lamp examination of anterior
segment is followed by gonioscopy and fundus examina-
tion.
Visual field defects are similar to those seen in adults
with initial predilection for loss in arcuate areas, but
child should be able to follow directions for field exami-
nation (perimetry). Although the technology has been
around and algorithm is getting better and better, it is still
difficult to apply them in children as they may not follow
instructions or may not cooperate. Optical coherence
tomography may be a better option and much easier as
compared to visual field examination in young and non-
cooperative patients.
7. Differential Diagnosis
Signs and symptoms Condition
Tearing, discharge,
conjunctival injection
Nasolacrimal duct
obstruction infections
Allergic conjunctivitis
Cloudy cornea, loss of
corneal lusture, Haab’s
striae
Corneal dystrophy e.g.
CHED, PPMD Forcep
trauma Cysticercosis
Sclerocornea
Mucopolysaccharidosis
I, Is, II, III infectious keratitis
Enlarged cornea,
apparent asymmetry
of globe size
Primary megalocornea
Unilateral high myopia
Proptosis lid retraction
Contralateral microphthalmos
Enopthalmos ptosis
Photophobia,
blepharospasm
Uveitis corneal infection,
abrasion or dystrophy
Retinal cone dystrophy
High myopia Pathologic myopia
Vitreo-retinal degeneration
e.g. Stickler’s syndrome
Enlarged cup to
disc ratio
Physiologic optic nerve
cupping optic nerve
atrophy or anomaly
Copyright © 2012 SciRes. OJOph
Childhood Glaucoma: An Overview
Copyright © 2012 SciRes. OJOph
74
8. Management 8.1.1. Beta Blockers
They lower IOP by decreasing production of aqueous in
the ciliary body. The drug should be used with extreme
caution in neonates due to the possibility of broncho-
spasm, apnea and bradycardia. Cardiac abnormalities and
bronchial asthma should be specifically excluded before
its use. Use of 0.25%, rather than 0.5% is recommended
in children, in order to reduce its side-effects. In 100 eyes
with childhood glaucoma treated with timolol, 31% pa-
tients experienced a reduction in IOP [25].
The definitive treatment for primary infantile glaucoma
is surgical. Medical therapy usually provides a suppor-
tive role to reduce the IOP temporarily, to clear the cor-
nea, and to facilitate surgical intervention. Laser therapy
has limited role in developmental glaucomas. Primary
surgical treatment is usually with goniotomy or trabecu-
lotomy, although combined trabeculotomy with trabe-
culectomy may be useful in certain populations with high
risk of failure of goniotomy or trabeculotomy. Refractory
congenital glaucomas may be managed by trabeculectomy
with anti-fibrosis drugs, glaucoma drainage implants and
cyclodestructive procedures (Figure 1).
8.1.2. Alph a- 2 Ago n i sts
They also decrease aqueous production but their use in
children is limited because of central nervous system
depression. In 30 patients with mean age of 10 years,
brimodine treatment was associated with a mean reduc-
tion of IOP by 7% [26]. Two young children were tran-
siently unarousable and five other children experienced
extreme fatigue [26]. In another study involving 23 pa-
tients with mean age of 8 years, 18% had systemic ad-
verse effects that necessitated stopping of the drug [27].
8.1. Medical Management
Medications do play a limited role in the treatment of
childhood glaucoma. In primary infantile glaucoma,
medications may be used preoperatively to clear the cor-
neal edema or post-operatively if the response to surgery
is borderline and more time is needed to determine if
further surgery is required.
Con
g
enital Glaucoma Sus
p
ect
Normal Examination Congenital Glaucoma
Corneal Diameter measurement
Tonometry
Gonioscopy
Ophthalmoscopy
Slit Lamp examination
Visual field analysis (if possible)
Follow-up Goniotomy
Trabeculotomy
Trabeculectomy
Combined trabeculotomy and trabeculectomy
Successful
results
Follow-up
Unsuccessful
results
Filtration surgery with antifibrotic drugs
Glaucoma drainage implants
Cyclodestructive procedures
Figure 1. Flowchart of management of congenital glaucoma.
Childhood Glaucoma: An Overview 75
8.1.3. Carbonic Anhydrase Inhibitors (CAIs)
They suppress aqueous production and are available as
either an oral formulation or as topical drop. The oral
formulation may be more effective in IOP lowering but
also produces more side-effects of diarrhea, lethargy,
poor appetite and metabolic acidosis [28]. The mean was
reduced by 36% and 27% compared with baseline, after
treatment with acetazolamide and topical dorzolamide,
respectively. All eyes showed an increase in IOP when
switched from acetazolamide to dorzolamide with mean
increase of 3.7mmHg [29]. Topical CAI can be expected
to have additive benefit when used in conjunction with
beta-blockers [30].
8.1.4. Pros t aglandins Ana logues
These drugs reduce IOP by enhancing uveo-scleral out-
flow [31]. Their side-effects include conjunctival hy-
peremia, iris and skin pigmentation and accelerated eye-
lash growth. High non-response rate has been reported in
children with little IOP lowering in responders [32].
8.1.5. Mio t i c s
They are ineffective for PCG, but can be used pre-
operatively before angle surgery.
8.2. Surgical Treatment
Early surgical intervention is of prime importance in the
management of patients with PCG. Either goniotomy or
trabeculotomy is the procedure of choice; goniotomy
requires clear cornea while trabeculotomy may be per-
formed if cornea is hazy or opaque. When greater
chances of failure are present, trabeculotomy may be
combined with trabeculectomy.
8.2.1. Goni o to m y
A blade is inserted through the peripheral cornea 180˚
across from meshwork to be incised (usually the nasal or
the temporal portions) and is then guided across the ante-
rior chamber into the chamber angle. With the aid of go-
nioscopy lens, the trabecular meshwork is visualized, and
the blade is used to make a linear incision through the
meshwork for approximately one third of the circumfer-
ence of eye. The reported rate of success of goniotomy in
infantile glaucoma is 80% [33]. Endoscopic goniotomy
has been successfully performed utilizing a co-axial ocu-
lar endoscope in presence of corneal opacification, which
prevented standard goniotomy procedure [34].
8.2.2. Trabeculotomy
A scleral flap is created over the area of network to be
incised. Through this flap a dissection into schlemm’s
canal is created. A trabeculotome is inserted into the ca-
nal and the meshwork is then opened, as instrument is
rotated into the anterior chamber. Considering congenital
glaucoma of all grades of severity, trabeculotomy con-
trols IOP in over 90% of eyes. The efficacy of this opera-
tion compared favorably with goniotomy. The results of
trabeculotomy and goniotomy for infantile glaucoma
were compared and found equally effective and safe [35].
The significant advantage of trabeculotomy for those
cases with a cloudy cornea limiting visualization of the
angle was described [36]. A newer trabeculotomy tech-
nique with protein suture passed 360˚ through Schlemm’s
canal has been mentioned [37].
8.2.3. Trabeculectomy
A trabeculectomy involves creating a full thickness
opening in the sclera (sclerotomy) for outflow of aqueous.
A partial thickness scleral flap covers the opening and
the conjunctiva overlies the flap. Intraocular pressure can
be significantly lowered with this. Success rate in adults
is usually around 70% - 80%. In children, long term suc-
cess rate is lower being around 50% [38] due to more
aggressive wound healing response in children, which
scars down sclerotomy or scleral flap. Children are also
more prone to complications such as infections due to
eye rubbing.
8.3. Management of Refractory Pediatric
Glaucomas (Figure 1)
When the IOP is not controlled after the first surgery, the
surgical options are filtration surgery with anti-fibrosis
drugs; glaucoma drainage implants or cyclodestructive
procedures.
8.3.1. Trabeculectomy with Anti-Fibrotic Agents
Mitomycin-C and 5-Florouracil are the two most com-
monly used anti-metabolites in glaucoma surgery. Al-
though each of these medications decrease the scarring of
blebs, they also create more potential for complications
such as infection (endopthalmitis) [39-42].
8.3.2. Aqueous Drainage Implants
Implant surgery is an important treatment alternative for
PCG patients who are poor candidates for angle incision
therapy and trabeculectomy or who have proven to be
refractory to these procedures. Glaucoma drainage im-
plants may be characterized as non-restrictive devices
such as Molteno and Baerveldt implants or valved such
as krupin implants or Ahmed glaucoma valve. Reported
complications of implants include hypotony with shallow
anterior chamber, choroidal detachments, tube cornea
touch, obstructed tube or plate, endophthalmitis and reti-
nal detachment.
The surgical procedure is same for all implants [43].
The superior temporal quadrant is the preferred site. Fol-
lowing periotomy, the implant device is placed with its
anterior edge approximately 8mm from limbus. The tube
Copyright © 2012 SciRes. OJOph
Childhood Glaucoma: An Overview
76
is shortened and beveled and an incision is made into the
anterior chamber to allow entry of tube parallel to iris.
The tube is then protected with autologous sclera or
pericardium and limbal peritomy is closed.
8.3.3. Cyclode s tructive Procedures
These are selectively used for PCG which has proven
refractory to medical therapy and to conventional surgi-
cal procedures to improve aqueous outflow and work by
decreasing aqueous production. The required ciliary
epithelial ablation is produced by trans-scleral cyclocryo-
therapy or by endoscopic diode laser cyclophotocoagula-
tion. The ocular indications include a blind painful eye, a
blind eye with high pressure, rapidly deteriorating cornea
refractory to all treatment alternatives and, an eye with
anatomic defect which preclude other glaucoma proce-
dures.
8.4. Low Vision Rehabilitation
Regrettably, children with congenital glaucoma may end
up with low vision despite treatment. Various low vision
aids such as magnifiers, binoculars, telescopes are avail-
able for such patients to improve their quality of life.
Some non-optical devices such as close circuit televi-
sions, large print books exist for their help. Such patients
should receive special care and vocational training from
trained professionals, working specifically for visually
impaired patients, in order to help them develop their
abilities to maximum potential [44].
9. Conclusion
The main goal in managing primary congenital glaucoma
is early diagnosis and therefore early surgical interven-
tion. Following successful surgery, glasses should be
prescribed and care must be taken to manage amblyopia
for optimal visual rehabilitation.
REFERENCES
[1] D. H. Hoskins, R. N. Shaffers and J. Hetherington, “Ana-
tomical Classification of the Developmental Glaucomas,”
Archives of Ophthalmology, Vol. 102, No. 9, 1984, pp.
1331-1336. doi:10.1001/archopht.1984.01040031081030
[2] S. J. H. Miller, “Genetic Aspects of Glaucoma,” Transac-
tions of the Ophthalmological Society of the UK, Vol. 86,
1966, pp. 425-434.
[3] S. C. Debnath, K. D. Teichmann and K. Salamah, “Tra-
beculectomy versus Trabeculotomy in Congenital Glau-
coma,” British Journal of Ophthalmology, Vol. 73, No. 8,
1989, pp. 608-611. doi:10.1136/bjo.73.8.608
[4] A. Gencik, “Epidemiology and Genetics of Primary Con-
genital Glaucoma in Slovakia. Description of a Form of
Primary Congenital in Gypsies with Autosomal-Reces-
sive Inheritance and Complete Penetrance,” Develop-
ments in Ophthalmology, Vol. 16, 1989, pp. 76-115.
[5] V. P. DeLuise and D. R. Anderson, “Primary Infantile
Glaucoma (Congenital Glaucoma),” Survey of Ophthal-
mology, Vol. 28, No. 1, 1983, pp. 1-19.
doi:10.1016/0039-6257(83)90174-1
[6] R. N. Shaffer, “Genetics and the Congenital Glaucomas,”
American Journal of Ophthalmology, Vol. 60, 1965, pp.
981-994.
[7] A. E. Kolker and J. Hetherington, “Congenital Glau-
coma,” In: Becker-Shaffer’s Diagnosis and Therapy of
the Glaucomas, 5th Edition, CV Mosby, St Louis, 1983, p.
317
[8] M. Sarfarazi and I. Stoilov, “Molecular Genetics of Pri-
mary Congenital Glaucoma,” Eye, Vol. 14, 2000, pp. 422-
428. doi:10.1038/eye.2000.126
[9] A. N. Akarsu, M. E. Turacli, S. G. Aktan, et al., “A Sec-
ond Locus (GLC3C) for Primary Glaucoma (Buphthal-
mos) Maps to the 1p36 Region,” Human Molecular Ge-
netics, Vol. 5, No. 8, 1996, pp. 1199-1203.
doi:10.1093/hmg/5.8.1199
[10] I. R. Stoilov and M. Sarfarazzi, “The Third Genetic Locus
(GLC3C) for Primary Congenital Glaucoma (PCG) Maps
to Chromosome 14q24.3,” Investigative Ophthalmology
& Visual Science, Vol. 43, 2002, pp. 1820-1827.
[11] M. Sarfarazi, A. N. Arkarsu, A. Hossain, et al., “Assign-
ment of a Locus (GLC3A) for Primary Congenital Glau-
coma (Buphthalmos) to 2p21 and Evidence for Genetic
Heterogeneity,” Genomics, Vol. 30, No. 2, 1995, pp. 171-
178. doi:10.1006/geno.1995.9888
[12] I. Stoilov, A. Nurten and M. Sarfarazi, “Identification of
Three Different Truncating Mutations in Cytochrome
P4501B1 (CYP1B1) as the Principal Cause of Primary
Congenital Glaucoma in Families Linked to the GLC3A
on Chromosome 2p21,” Human Molecular Genetics, Vol.
6, No. 4, 1997, pp. 641-647. doi:10.1093/hmg/6.4.641
[13] B. A. Bejjani, R. A. Lewis, T. F. Tomey, et al., “Muta-
tions in CYP1B1, the Gene for Cytochrome P4501B1,
Are the Predominant Cause of Primary Congenital Glau-
coma in Saudi Arabia,” American Journal of Human Ge-
netics, Vol. 62, No. 2, 1998, pp. 325-333.
doi:10.1086/301725
[14] S. G. Panicker, A. N. Mandal, A. B. M. Reddy, V. K.
Gothwal and S. E. Hasnain, “Correlations of Genotype
with Phenotype in Indian Patients with Primary Congeni-
tal Glaucoma,” Investigative Ophthalmology & Visual
Science, Vol. 45, No. 4, 2004, pp. 1149-1156.
[15] A. T. Johnson, A. V. Drack, A. E. Kwitek, et al., “Clini-
cal Features and Linkage Analysis of a Family Autosomal
Dominant Juvenile Glaucoma,” Ophthalmology, Vol. 100,
No. 4, 1993, pp. 524-529.
[16] V. C. Sheffield, E. M. Stone, W. L. M. Alward, et al.,
“Genetic Linkage of Familial Open Angle Glaucoma to
Chromosome 1q21-q31,” Nature Genetics, Vol. 4, No. 1,
1993, pp. 47-50. doi:10.1038/ng0593-47
[17] D. B. Gould, A. J. Mears, W. G. Pearce and M. A. Walter,
“Autosomal Dominant Axenfeld-Rieger Anomaly Maps
to 6p25,” American Journal of Human Genetics, Vol. 61,
No. 3, 1997, pp. 765-768.
Copyright © 2012 SciRes. OJOph
Childhood Glaucoma: An Overview
Copyright © 2012 SciRes. OJOph
77
doi:10.1016/S0002-9297(07)64340-7
[18] T. Glaser, D. S. Walton and R. L. Mass, “Genomic Struc-
ture, Evolutionary Conservative and Aniridia Mutations
in the Human PAX6 Gene,” Nature Genetics, Vol. 2, No.
3, 1992, pp. 232-239. doi:10.1038/ng1192-232
[19] O. Barkan, “Pathogenesis of Congenital Glaucoma,”
American Journal of Ophthalmology, Vol. 40, 1955, pp.
1-11.
[20] I. Tugal-Tutkun, K. Havrlikova, W. J. Power and C. S.
Foster, “Changing Patterns in Uveitis of Childhood,”
Ophthalmology, Vol. 103, No. 3, 1996, pp. 375-383.
[21] D. S. Walton, “Pediatric Aphakic Glaucoma: A Study of
65 Patients,” Transactions of the American Ophthal-
mological Society, Vol. 93, 1995, pp. 403-413.
[22] R. N. Shaffer and J. Hetherington Jr., “The Glaucomatous
Disc in Infants. A Suggested Hypothesis for Disc Cup-
ping,” Transactions of the American Academy of Oph-
thalmology & Otolaryngology, Vol. 73, No. 5, 1969, pp.
923-935.
[23] K. T. Richardson and R. N. Shaffers, “Optic-Nerve Cup-
ping in Congenital Glaucoma,” American Journal of
Ophthalmology, Vol. 62, No. 3, 1966, pp. 507-509.
[24] R. N. Jaffar and A. K. Ghulamqadir, “Effect of Oral Cho-
ral Hydrate on the Intraocular Pressure Measurement,”
Journal of Pediatric Ophthalmology and Strabismus, Vol.
30, No. 6, 1993, pp. 372-376.
[25] H. D. Hoskins Jr., J. Hetherington Jr., S. D. Magee, R.
Naykhin and C. V. Migliazzo, “Clinical Experience with
Timolol in Childhood Glaucoma,” Archives of Ophthal-
mology, Vol. 103, No. 8, 1985, pp. 1163-1165.
doi:10.1001/archopht.1985.01050080075024
[26] L. B. Enyedi and S. F. Freedman, “Safety and Efficacy of
Brimonidine in Children with Glaucoma,” Journal of
AAPOS, Vol. 5, No. 5, 2001, pp. 281-284.
doi:10.1067/mpa.2001.117571
[27] R. J, Bowman, J, Cope and K, K. Nischal, “Ocular and
Systemic Side-Effects of Brimonidine 0.25% Eye Drops
(Alphagan in Children),” Eye, Vol. 18, 2004, pp. 24-26.
doi:10.1038/sj.eye.6700520
[28] L. I. Larsson and A. Alm, “Aqueous Humor Flow in Hu-
man Eyes Treated with Dorzolamide and Different Doses
of Acetazolamide,” Archives of Ophthalmology, Vol. 116,
No. 1, 1998, pp. 19-24.
[29] M. Portellos, E. G. Buckley and S. F. Freedman, “Topical
versus Oral Carbonic Anhydrase Inhibitor for Pediatric
Glaucoma,” Journal of AAPOS, Vol. 2, No. 1, 1998, pp.
43-47. doi:10.1016/S1091-8531(98)90109-4
[30] L. L. Wayman, L. I. Larsson, T. L. Maus and R. F. Bru-
baker, “Additive Effect of Dorzolamide on Aqueous Hu-
mor Flow in Patients Receiving Long Term Treatment
with Timolol,” Archives of Ophthalmology, Vol. 116, No.
11, 1998, pp. 1438-1440.
[31] R. K. Parrish, P. Palmberg and W. P. Sheu, “A Compari-
son of Latanoprost, Bimatoprost and Travoprost in Pa-
tients with Elevated Intraocular Pressure: A 12-Week,
Randomized, Masked-Evaluator Multicenter Study,” Ameri-
can Journal of Ophthalmology, Vol. 135, No. 5, 2003, pp.
688-703. doi:10.1016/S0002-9394(03)00098-9
[32] L. B. Enyedi, S. F. Freedman and E. G. Buckley, “The
Effectiveness of Latanoprost for the Treatment of Pediat-
ric Glaucoma,” Journal of AAPOS, Vol. 3, No. 1, 1999,
pp. 33-39. doi:10.1016/S1091-8531(99)70092-3
[33] K. S. Morgan, B. Black, F. D. Ellis and E. M. Helveston,
“Treatment of Congenital Glaucoma,” American Journal
of Ophthalmology, Vol. 92, 1981, pp. 799-803.
[34] K. M. Joos and J. H. Shen, “An Ocular Endoscope En-
ables a Goniotomy Despite a Cloudy Cornea,” Archives
of Ophthalmology, Vol. 119, No. 1, 2001, pp. 134-135.
[35] S. D. McPherson Jr. and D. MacFarland, “External Tra-
beculotomy for Developmental Glaucoma,” Ophthalmol-
ogy, Vol. 87, No. 4, 1980, pp. 302-305.
[36] S. D. McPherson Jr. and D. P. Berry, “Goniotomy vs
External Trabeculotomy for Developmental Glaucoma,”
American Journal of Ophthalmology, Vol. 95, No. 4,
1983, pp. 427-431.
[37] A. D. Beck and M. G. Lynch, “360 Degrees Trabecu-
lotomy for Congenital Glaucoma,” Archives of Ophthal-
mology, Vol. 113, No. 9, 1995, pp. 1200-1202.
doi:10.1001/archopht.1995.01100090126034
[38] G. R. Beauchamp and M. M. Parks, “Filtering Surgery in
Children: Barriers to Success,” Ophthalmology, Vol. 86,
No. 1, 1979, pp. 170-180.
[39] A. K. Mandal, D. S. Walton, T. John, et al., “Mitomycin
C Augmented Trabeculectomy in Refactory Congenital
Glaucoma,” Ophthalmology, Vol. 104, No. 6, 1997, pp.
996-1003.
[40] R. Susanna, E. W. Oltrogge, J. C. E. Carani, et al., “Mi-
tomycin as Adjunct Chemotherapy with Trabeculectomy
in Congenital and Developmental Glaucomas,” Journal of
Glaucoma, Vol. 4, No. 3, 1995, pp. 151-157.
doi:10.1097/00061198-199506000-00002
[41] S. Waheed, D. C. Ritterband and D. S. Greenfield, “Bleb-
Related Ocular Infection in Children after Trabeculec-
tomy with Mitomycin C,” Ophthalmology, Vol. 104, No.
12, 1997, pp. 2117-2120.
[42] M. Zalish, H. Leiba and M. Oliver, “Subconjunctival
Injection of 5-Fluorouracil Following Trabeculectomy for
Congenital and Infantile Glaucoma,” Ophthalmic Surgery,
Vol. 23, No. 3, 1992, pp. 203-205.
[43] M. Akimato, H. Tamihara, A. Negi and M. Nagata, “Sur-
gical Results of Trabeculotomy ab Externo for Develop-
mental Glaucoma,” Archives of Ophthalmology, Vol. 112,
No. 12, 1994, pp. 1540-1544.
[44] A. K. Mandal and D. Chakrabarti, “Update on Congenital
Glaucoma,” Indian Journal of Ophthalmology, Vol. 59,
No. 7, 2011, pp. 148-157.
doi:10.4103/0301-4738.73683