You are on page 1of 12

Oral Memantine for the Treatment of

Glaucoma
Design and Results of 2 Randomized, Placebo-Controlled,
Phase 3 Studies
Robert N. Weinreb, MD,1 Jeffrey M. Liebmann, MD,2 George A. Cioffi, MD,2
Ivan Goldberg, MBBS, FRANZCO,3 James D. Brandt, MD,4 Chris A. Johnson, PhD, DSc,5
Linda M. Zangwill, PhD,1 Susan Schneider, MD,6 Hanh Badger, PharmD,6 Marina Bejanian, PhD6

Purpose: To evaluate the effectiveness and safety of oral memantine as a potential neuroprotective agent in
open-angle glaucoma (OAG) at risk for progression.
Design: Two randomized, double-masked, placebo-controlled, parallel-group, multicenter, 48-month studies
identically designed, initiated 1 year apart, and completed in 2006. Protocol amendments included a 1-year
extension (first study) and change in primary endpoint and analysis (second study).
Participants: Patients (2298 total) with bilateral OAG; glaucomatous optic disc damage and visual field loss
in 1 eye; glaucomatous optic disc damage and/or visual field loss in the contralateral eye (at screening), topically
treated or untreated intraocular pressure (IOP) of 21 mmHg or less (at baseline); and at risk of glaucomatous
progression (per prespecified criteria).
Methods: Patients were randomized 3:2:2 to receive memantine 20 mg, memantine 10 mg, or placebo
tablets daily. Glaucomatous progression was assessed in the intent-to-treat population by full-threshold standard
automated perimetry (SAP), frequency doubling technology (FDT), and stereoscopic optic disc photographs,
standardized by quality control assessment at centralized reading centers. Safety evaluations included adverse
events (AEs), best-corrected visual acuity, biomicroscopy, IOP, and ophthalmoscopy. Efficacy data from each
study were analyzed per protocol. Pooled analyses of efficacy and safety data were also performed.
Main Outcome Measures: The predefined primary efficacy measure was glaucomatous visual field pro-
gression, as measured by SAP. Additional efficacy measures included glaucomatous progression of visual field
(FDT) and optic nerve damage (stereoscopic optic disc photographs).
Results: The proportion of patients who completed the studies was similar among groups (80%e83%).
Compared with placebo, daily treatment with memantine 10 mg or 20 mg for 48 months did not delay glau-
comatous progression significantly in the individual studies and pooled analyses. The pooled risk reduction ratio
(95% confidence interval) assessed by SAP was 0.13 (0.40, 0.09) and 0.17 (0.46, 0.07) for memantine 10
mg and 20 mg, respectively. Results were similar per FDT and stereoscopic optic disc photographs. The most
common AEs leading to treatment discontinuations were dizziness, headache, fatigue, and nausea.
Conclusions: With technologies available when the studies were conducted, daily treatment with mem-
antine over 48 months was not shown to prevent glaucomatous progression in this patient
population. Ophthalmology 2018;125:1874-1885 ª 2018 by the American Academy of Ophthalmology. This is
an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Supplemental material available at www.aaojournal.org.

Open-angle glaucoma (OAG) is an irreversible, chronic, clinical trials of glaucoma or ocular hypertension, lowering
progressive, multifactorial optic neuropathy characterized IOP did reduce the number of patients reaching a glaucoma
by progressive retinal ganglion cell death and functional endpoint but a large number of patients continued to wor-
optic nerve damage.1 Management of OAG currently relies sen.4e7 Thus, there is an unmet need for alternative treat-
on pharmacologic and surgical treatments aiming to lower ment strategies that could be used alone or in conjunction
intraocular pressure (IOP), a key risk factor.1e3 However, with IOP-lowering therapy.
lowering IOP does not always prevent disease progression. Because glaucoma is a neurodegenerative disease, patients
In each of the major National Institutes of Health-sponsored could benefit from neuroprotective therapies that would

1874 ª 2018 by the American Academy of Ophthalmology https://doi.org/10.1016/j.ophtha.2018.06.017


This is an open access article under the CC BY-NC-ND license ISSN 0161-6420/18
(http://creativecommons.org/licenses/by-nc-nd/4.0/). Published by Elsevier Inc.
Weinreb et al 
Oral Memantine in Glaucoma

prevent or slow retinal ganglion cell loss.8 Preclinical studies presents the 4-year data from both studies. Because there were no
have shown that elevated levels of the excitatory amino acid specific guidelines that could be used to develop the study protocol,
glutamate are toxic to mammalian retinal ganglion cells and scientific advice on the study design and endpoints was provided by
neurons, and that the resulting damage is mediated by the Committee for Medicinal Products for Human Use and agreed
upon by the United States Food and Drug Administration.
overstimulation (or excitotoxicity) of the N-methyl-D-
aspartate (NMDA) receptor, which in turn leads to an
excess of intracellular calcium and cell death.9e12 In addi- Study Population and Treatment
tion, increased levels of glutamate and glutamine (metabolic Patients diagnosed with bilateral OAG were eligible for enrollment
precursor) and decreased levels of the glutamate transporter in the study if they met predetermined criteria of patients at risk of
EAAT-1 have been reported in the vitreous13 and retinal glaucomatous progression (Table 1). Notably, patients were
mounts,14 respectively, of patients with glaucoma (compared excluded if they had any ocular pathology in either eye that
with healthy individuals), suggesting an association between could have interfered with the ability to obtain visual field
the excessive release of glutamate in the retina, neuronal cell assessments, or any condition or situation that could have
death, and glaucoma.15,16 confounded the study results. Patients with mild chronic
Memantine is an uncompetitive NMDA receptor antago- blepharitis, cataract, age-related macular degeneration, or back-
ground diabetic retinopathy could be enrolled at the investigator’s
nist17 that binds preferentially to NMDA receptor-operated
discretion. No washout of topical IOP-lowering medications was
cation channels; it is an open-channeleblocking NMDA required before initiation of study treatment.
antagonist and thus binds only to channels that have been At the baseline visit, unique patient numbers were assigned in
activated by glutamate binding to the receptor. Because ascending, consecutive order (without omission or reuse). An
memantine has a low to moderate affinity for the receptor and automated system was then used to randomize eligible patients to
its inhibitory effect exhibits strong dependence on membrane memantine 20 mg, memantine 10 mg, or placebo tablets daily in an
voltage,17,18 it effectively blocks excessive glutamatergic ac- uneven double-masked allocation of 3:2:2, respectively. This site-
tivity at concentrations that do not affect normal neurotrans- specific randomization scheme was based on phase 2 study results
mission. Memantine can inhibit overstimulation of the NMDA showing that the patient discontinuation rate was higher in the 20-mg
receptor and potentially provide neuroprotection by preventing memantine group than in the 10-mg memantine and placebo groups.
Although a 30-mg daily dose was deemed safe and tolerable in 2
excessive calcium influx. In support of this mechanism, results
short-term (12 weeks) phase 2 studies (with dizziness, coordina-
from preclinical studies have shown that memantine reduces tion abnormality, headache, and nausea being most frequently re-
excitotoxicity in animal models of glaucoma.19e23 ported as adverse events [AEs]; unpublished data [The first phase 2
Oral memantine was first offered for marketing by Merz study was conducted between October 1997 and December 1997,
& Co in 1982 and is currently approved in several countries while the second was conducted between February 1998 and June
for the treatment of moderate to severe Alzheimer’s and 1998] Allergan plc), it was thought that a 20-mg dose would be
Parkinson’s diseases, 2 other age-related neurodegenerative tolerated better for chronic use by most patients, consistent with the
pathologic conditions. The 2 phase 3 studies presented recommended dose in moderate to severe Alzheimer’s disease.
herein evaluated the effectiveness and safety of oral mem- Study tablets (identical in appearance) were provided in 1-week
antine administered daily in doses of either 10 mg or 20 mg, blister cards marked with the order of use to allow dose escalation
(if applicable) from 5 mg to 10 mg or 20 mg per day (Fig S1,
compared with placebo, in patients with chronic OAG who
available at www.aaojournal.org). Dosing began on a Monday
were at risk of disease progression. The clinical hypotheses (day 0), preferably between 5:00 PM and 9:00 PM (especially if
evaluated were (1) treatment with at least 1 daily dose of dizziness occurred), but could be changed to the morning
oral memantine would result in either significantly fewer (6:0010:00 AM) based on the occurrence of AEs (e.g., insomnia).
patients exhibiting glaucomatous progression or significant If treatment was discontinued before month 48 because of
delays in progression, as measured by changes in visual glaucomatous progression (confirmed by the reading center),
fields using standard automated perimetry (SAP), and (2) nonadherence to treatment, AEs, or other reasons (determined by
memantine has a favorable safety profile. the investigator), the patient remained in the study and attended
the remaining study visits. Data handling for those patients is
described in “Outcome Measures and Statistical Analyses” below.
Methods
Assessments
Study Design
Twenty visits were scheduled over 48 months (Table S1, available at
Two randomized, double-masked, placebo-controlled, parallel-group, www.aaojournal.org). Efficacy evaluations included glaucomatous
multicenter, 48-month, identically designed studies (Clinical- visual field progression by SAP (using a Humphrey field analyzer
Trials.gov identifiers, NCT00141882 and NCT00168350; registered [HFA] 24-2 full threshold program) and frequency doubling
on August 31, 2005, and September 9, 2005, respectively) were technology (FDT), as well as progression of glaucomatous optic
initiated 1 year apart and conducted in accordance with the tenets of nerve damage based on stereoscopic optic disc photographs. All
the Declaration of Helsinki, the International Conference on Har- visual field evaluations (i.e., qualification of disease status for
monisation Guidelines for Good Clinical Practice, and all privacy study entry and progression) based on SAP and FDT findings
requirements applicable at the time. Institutional review board or were standardized by undergoing quality control assessment at a
ethics committee approval was obtained, and all patients provided centralized visual field reading center (Devers Eye Institute/
written informed consent before any study-related procedure or Discoveries in Sight, Portland, Oregon). Specifically, after
changes in treatment, as well as other locally required documentation confirmation of acceptability of the baseline visual fields for each
of consent. Although the first study was extended by 1 year, this article patient (before treatment initiation), the visual field reading center

1875
Ophthalmology Volume 125, Number 12, December 2018

Table 1. Eligibility Criteria

Key Inclusion and Exclusion Criteria


Key inclusion criteria

 Topically treated or untreated IOP 21 mmHg in each eye at baseline (laser or filtering surgery performed >3 months before screening was
acceptable)
 Experience with SAP visual field tests before the screening visit (i.e., 2 tests during the past 2 yrs using the Humphrey perimeter with a 30-2 or 24-2
full-threshold algorithm, the full-threshold versions of FastPac or Swedish interactive threshold algorithm standard, or the equivalent on the Octopus
perimeter)
 Presence at the screening visit of typical glaucomatous optic disc damage and glaucomatous visual field loss in 1 eye, and either glaucomatous optic
disc damage or visual field loss, or both, in the contralateral eye, as confirmed by the visual field reading center (Devers Eye Institute/Discoveries in
Sight, Portland, Oregon) and optic disc reading center (Hamilton Glaucoma Center, University of California San Diego, La Jolla, California)
 Patient is at risk of glaucomatous progression, as determined by the documented presence of optic disc hemorrhage within 12 mos of the screening
visit in either eye, OR 2 of the following risk factors:
e Vertical cup-to-disc ratio 0.8 in one or both eyes
e Mean deviation in visual field worse than 10 dB in either eye (verified by the reading center)
e Pseudoexfoliation in either eye
e Age >70 yrs
e Family history (parent or sibling) of glaucoma
e Black African racial origin
e Systemic hypertension requiring medical treatment
e Migraine (defined by the International Headache Society24) with or without aura, Raynaud’s syndrome, or both
 BCVA of 20/32 or better (Snellen equivalent on the ETDRS chart) in each eye
Key exclusion criteria

 History of epilepsy or significant psychiatric disease, including schizophrenia, bipolar disorder, or major depression
 Clinical determination of impaired renal function
 Dizziness or lightheadedness within 1 week before the baseline visit
 Use of >2 topical (or any oral) IOP-lowering products at the baseline visit
 Any glaucoma other than chronic POAG, LTG (not defined in the protocol), pigmentary glaucoma, or pseudoexfoliative glaucoma
 Any ocular pathology in either eye that may have interfered with the ability to obtain visual field, disc imaging, or accurate IOP readings
 Any situation or condition that, in the investigator’s opinion, may have confounded the study results (e.g., interfered with visual function) or
interfered significantly with participation in the study
 Refractive error such that the spherical equivalent was worse than þ5 or 5 D, or the cylinder value was worse than þ3 or 3 D (before conversion
to spherical equivalent)
 Any active ocular disease other than OAG (e.g., uveitis, ocular infections, keratoconus, or severe dry eye) or the inability to dilate the pupils to at
least 4 mm (with medication, if necessary)
 Laser or any other intraocular surgery within 3 months before the screening visit (with a total exclusion for refractive surgery)
 Visual field mean deviation worse than 22 dB in either eye (verified by the reading center)
 Women who were pregnant, nursing, planning a pregnancy, or not using contraception

BCVA ¼ best-corrected visual acuity; D ¼ diopter; dB ¼ decibels; ETDRS ¼ Early Treatment Diabetic Retinopathy Study; IOP ¼ intraocular pressure;
LTG ¼ low-tension glaucoma; OAG ¼ open-angle glaucoma; POAG ¼ primary open-angle glaucoma; SAP ¼ standard automated perimetry.

received and evaluated all visual field tests obtained from both eyes testeretest results for glaucoma patients and sensitivity for indi-
at all visits (scheduled or unscheduled). Similarly, stereoscopic optic vidual visual field locations. Progression was declared if the same 5
disc photographs (including those obtained at the screening visit) or more visual field locations demonstrated significant reductions
were obtained by certified photographers, and progression was from baseline (P < 0.05) in a confirmatory visual field obtained
graded by independent, certified graders at a centralized optic disc within 8 weeks. Although there is still no gold standard to define
reading center (Hamilton Glaucoma Center, University of visual field progression, this procedure was found to yield 92.2%
California San Diego, La Jolla, California). sensitivity to detect the earliest change in glaucomatous visual field
Detection of progression of glaucomatous visual fields by SAP loss, and 97.2% specificity to correctly identify glaucomatous vi-
used the procedure outlined by Chauhan et al25 and was based on sual fields that have not undergone progressive change (Johnson
the glaucoma change probability26 originally described by Heijl CA, Cioffi GA, personal communication, 1999; based on longi-
et al26 and available on HFA using the STATPAC2 program tudinal data from 203 glaucoma patients with 10 visual fields
(Carl Zeiss Meditech, Dublin, CA). Baseline visual field values obtained over >5 years and verified by computer simulations).
(established by averaging data obtained at screening and baseline Glaucomatous visual field progression by FDT was measured
for each visual field location of each eye) and total deviation by certified technicians using the Humphrey Zeiss/Welch Allyn
probability maps (showing the difference between patient-specific system (Carl Zeiss, Oberkochen, Germany) with full-threshold N-
values and age-specific normal values) obtained for each visual 30 procedure (FDT software version 2.6 or higher) according to
field location at each prespecified postbaseline visit (Table S1) standardized instructions. Threshold (measured in decibels [dB])
were recorded. The change from baseline (total deviation) was and total deviation of 19 test points per eye were recorded, along
then determined for each location; a reduction greater than the with fixation, false-positive and false-negative errors. All results,
5% probability level at a single visual field location indicated a including abnormal and unreliable ones, were sent to the reading
significant change from baseline for that location, based on center, which developed an FDT glaucoma change probability

1876
Weinreb et al 
Oral Memantine in Glaucoma

program (similar to that for the HFA) to determine the progression Post hoc analyses of the first study that excluded the subgroup
status of each test point of each eye at all postbaseline visits, based of patients with low-tension glaucoma (LTG; not defined per
on testeretest results from healthy controls (1885 years of age) protocol) were performed because of the considerable number of
and glaucoma patients with various amounts of visual field dam- such patients and because LTG may involve different mechanisms
age. Specifically, total deviation probability maps were obtained (compared with non-LTG).28 Based on these and other findings of
for each visual field location and compared with baseline values (as the first study, the second study (NCT00168350) protocol and
described above). Values that indicated a change from baseline that analysis plan were amended before unmasking that study. The
was more than the 95% confidence limits (based on location and primary efficacy measure was changed to the cumulative
sensitivity) were considered significant changes (P < 0.05) from probability of developing progression by month 48 using FDT
baseline. Glaucomatous progression of an eye was declared if 2 (as verified by the reading center). The primary population was
adjacent test locations exhibited significant reductions in sensitivity changed to all randomized patients during treatment who did not
(P < 0.05) from baseline on the FDT glaucoma change probability have LTG at baseline, and comparison between memantine 20
analysis. Time to glaucomatous field progression for each patient mg and placebo was considered primary. The cumulative
was the time when the first progression event was found in either probability of developing progression by month 48 was based on
eye at any postbaseline visit. No confirmation test was required for the life table method.
this efficacy measure. For each study, a sample size of approximately 1050 patients
In addition to the methods described above, the visual field was to be enrolled to have an estimated 233 patients per treatment
readers performed clinical assessment of all visual fields (SAP and group complete the 48-month visit, or confirmed progression of
FDT) to verify that the pattern and shape of visual field loss was glaucoma (20%) by HFA 24-2 perimetry (primary efficacy var-
consistent with glaucomatous damage and was not typical of iable) while on study treatment, based on the following assump-
pathologic characteristics in other locations in the visual pathways tions: an estimated dropout rate of 45% in the 20-mg memantine
(e.g., cataract, macular disease, retinal degenerations, other optic group versus 25% in the 10-mg memantine and placebo groups
neuropathies, and chiasmal or postchiasmal disorders). (based on unpublished phase 2 study results suggesting a higher
Progression of glaucomatous optic nerve damage on stereo- rate of discontinuation with memantine 20 mg over 12 weeks); a 2-
scopic optic disc photographs was assessed by 2 graders who sided log-rank test with an a of 0.05 for equality of survival curves
compared baseline photographs with photographs taken at each of time to progression; and 85%, 92%, and 96% power to detect a
prespecified follow-up visit (Table S1) but were masked to the difference of 10%, 12.5%, and 15%, respectively, among the
temporal sequence of the photographs to determine whether there memantine and placebo groups. The nQuery Advisor software
was a change in disc excavation/cupping, rim thinning, or retinal version 1.0 (Statistical Solutions Ltd, Boston, MA) was used for
nerve fiber layer defect between the baseline photograph and the computation.
each follow-up visit photograph; if they disagreed, a third grader After the studies were completed, data were pooled and sum-
adjudicated. If photograph quality was deemed unacceptable, a marized with descriptive statistics, frequency tables, and listings.
new set was obtained within 4 weeks. The photographic sequence Categorical variables were analyzed using the Wilcoxon rank-sum
then was unmasked (after grading and adjudicating were com- test (ordinal) and Fisher exact test, Pearson chi-square test, or
plete), consistent with the procedure described in the Ocular Hy- Cochran-Mantel-Haenszel method (nominal), incorporating the
pertension Treatment Study.27 investigator site effect (to account for potential intersite variability)
Safety evaluations included AEs, as well as vital signs, clinical where applicable. Continuous variables were tested for statistical
laboratory evaluations, best-corrected visual acuity, slit-lamp bio- significance using the analysis of variance. Within-group changes
microscopy (without pupil dilation), IOP (measured by standard from baseline at each follow-up visit were analyzed using the
Goldmann applanation tonometry after the biomicroscopic exam- Wilcoxon signed-rank test (categorical variables) and paired t test
ination was completed), and ophthalmoscopy (with pupil dilation) (continuous variables). For the primary analysis, a statistical
on predetermined visits (Table S1). adjustment for multiple between-group comparisons was made
using Hochberg’s procedure to control for an overall type-I error
Outcome Measures and Statistical Analyses rate of 5%. All analyses were generated using SAS software
version 9.1 (SAS Institute, Inc., Cary, NC); P values less than 0.05
The prespecified primary efficacy measure was progression of vi- were considered statistically significant.
sual field loss, as assessed using HFA (24-2 full threshold SAP Although only pooled analyses of the demographics, baseline
program) and verified by the visual field reading center. The key characteristics, and safety data for the overall population are
secondary efficacy measures were progression of glaucomatous presented herein, the efficacy results of each individual study are
findings based on FDT visual field data and stereoscopic optic disc presented to support the change in primary endpoint during the
photographs (verified by the visual field reading center and optic course of the drug development program, along with the corre-
disc reading center, respectively). sponding pooled analyses performed after completion of the studies.
Efficacy analyses were performed using the intent-to-treat
population (i.e., all randomized patients), based on the treatment
to which patients were randomized. Patients who stayed in the Results
study and attended the remaining study visits after discontinuing
treatment were included in those analyses. A survival analysis Demographics and Baseline Characteristics
based on the KaplaneMeier method and life table method was also
carried out, from which the time to an event was estimated. Sta- In the 2 phase 3 studies, a total of 2298 patients (first study,
tistical significance was assessed using the nonparametric log-rank n ¼ 1119; second study, n ¼ 1179) from 126 centers met the
test or Cox proportional hazards model as appropriate. Key effi- eligibility criteria and were randomized (intent-to-treat population).
cacy analyses were repeated using data obtained during treatment. Notably, the proportion of patients who completed each 48-month
Safety analyses included all randomized patients who received at study was similar in the memantine 20-mg (80%), memantine
least 1 dose of study medication (as treated), using data obtained 10-mg (83%), and placebo (83%) groups (Fig 1). The proportions
during treatment, that is, data from patients on treatment and up to of patients who discontinued study treatment because of AEs were
30 days after the last dose. numerically (but not statistically) higher in the memantine groups

1877
Ophthalmology Volume 125, Number 12, December 2018

Figure 1. Flowchart showing patient disposition. aClinicalTrials.gov identifiers, NCT00141882 and NCT00168350. bAnalyzed per protocol. cAnalyzed post
hoc. dAnalyzed per amended protocol. eIncluded both low-tension glaucoma (LTG) and non-LTG patients. Patients could have discontinued treatment.
f
Glaucoma progression. gIncluded protocol violations, concomitant therapy, administrative reasons (i.e., lost to follow-up, relocation, and personal reasons),
and others. ITT ¼ intent-to-treat.

than in the placebo group, consistent with the overall proportions dB, family history, or systemic hypertension requiring medical treat-
of patients who discontinued treatment (Fig 1). ment (Table 3). Topical IOP-lowering therapy used at study entry
Pooled patient demographics and baseline characteristics were included brimonidine, brinzolamide, latanoprost, betaxolol, dorzola-
similar among treatment groups and were consistent with those of mide, timolol, fixed-combination dorzolamide plus timolol, levobu-
patients with glaucoma in general (Table 2). Of note, approximately nolol, unoprostone, carteolol, travoprost, fixed-combination
25% of patients had LTG (per investigator assessment). Overall, brimonidine plus timolol, and fixed-combination latanoprost plus
89% of patients had a vertical cup-to-disc ratio 0.8, along with timolol. Prior glaucoma surgery included trabeculectomy, trephine
another risk factor such as visual field mean deviation worse than 10 surgery, argon laser trabeculoplasty, and iridectomy.

Table 2. Patient Demographics and Baseline Characteristics (Intent-to-Treat Population)*

Memantine 20 mg (n [ 974) Memantine 10 mg (n [ 664) Placebo (n [ 660)


Age, yrs
Mean (SD) 65.0 (9.9) 64.9 (9.9) 65.1 (9.4)
Range 18e81 26e82 28e81
Sex, no. (%)
Female 566 (58) 341 (51) 353 (53)
Male 408 (42) 323 (49) 307 (47)
Race, no. (%)
Black African 141 (14) 97 (15) 93 (14)
Other 833 (86) 567 (85) 567 (86)
Glaucoma diagnosis, no. (%)
Chronic POAG 705 (72) 476 (72) 479 (73)
LTGy 241 (25) 169 (25) 160 (24)
Pigmentary glaucoma 37 (4) 17 (3) 22 (3)
Pseudoexfoliative glaucoma 43 (4) 21 (3) 22 (3)
Central corneal thickness (SD), mm 53939 53738 54036
Mean IOP (SD), mmHg
Right eye 14.6 (3.2) 14.5 (3.3) 14.5 (3.5)
Left eye 14.6 (3.2) 14.4 (3.2) 14.7 (3.3)
Mean vertical cup-to-disc ratio (SD) 0.80 (0.11) 0.80 (0.11) 0.79 (0.12)

IOP ¼ intraocular pressure; LTG ¼ low-tension glaucoma; POAG ¼ primary open-angle glaucoma; SD ¼ standard deviation.
*ClinicalTrials.gov identifiers, NCT00141882 and NCT00168350.
y
Per investigator assessment; no specific criteria were defined in the protocol.

1878
Weinreb et al 
Oral Memantine in Glaucoma

Table 3. Pooled Risk Factors for Glaucomatous Progression at Baseline (Intent-to-Treat Population)*

Memantine 20 mg (n [ 974) Memantine 10 mg (n [ 664) Placebo (n [ 660)


Vertical cup-to-disc ratio 0.8 868 (89) 589 (89) 579 (88)
Visual field mean deviation <10 dB 502 (52) 349 (53) 325 (49)
Family history 431 (44) 278 (42) 302 (46)
Systemic hypertension requiring medical treatment 405 (42) 280 (42) 259 (39)
Age >70 yrs 371 (38) 248 (37) 249 (38)
Migrainey and/or Raynaud’s syndrome 181 (19) 113 (17) 111 (17)
Optic disc hemorrhage <12 mos before screening 152 (16) 85 (13) 123 (19)
Black African racial origin 145 (15) 99 (15) 94 (14)
Pseudoexfoliation 53 (5) 29 (4) 28 (4)

dB ¼ decibels.
*ClinicalTrials.gov identifiers, NCT00141882 and NCT00168350.
y
As defined by the International Headache Society,24 with or without aura.

Efficacy higher than with placebo when analyzed by FDT, not SAP (Fig 4).
After amendment of the primary efficacy outcome, the FDT-based
In the first study (NCT00141882), the SAP-based analysis showed analysis in non-LTG patients showed that by month 48, there was a
that by month 48, the cumulative probability of glaucomatous vi- statistically significantly higher probability of glaucomatous visual
sual field progression with memantine 10 mg was statistically field progression during treatment with memantine 10 mg, whereas
significantly higher than with placebo (Fig 2A). However, in the memantine 20 mg had no effect, compared with placebo (Fig 5A).
analyses based on FDT (Fig 2B) and stereoscopic optic disc Analyses based on SAP (Fig 5B) and optic disc assessments
photographs (Fig 2C), there were no statistically significant (Fig 5C) showed no statistically significant differences in the
differences in the cumulative probability of glaucomatous visual cumulative probability of glaucomatous visual field progression
field progression and percentage of patients with progression of and percentage of patients with progression of glaucomatous
glaucomatous optic nerve damage, respectively, between optic nerve damage, respectively, among the memantine (10 or
memantine 10 mg or 20 mg and placebo groups. Interestingly, 20 mg) and placebo groups.
post hoc subgroup analysis of non-LTG patients during treatment Pooled analyses of non-LTG patients during treatment (from
showed a statistically significantly lower probability of glaucom- both studies) also showed no statistically significant differences in
atous visual field progression with memantine 20 mg compared the cumulative probability of glaucomatous visual field progression
with placebo (Fig 3). by SAP (Fig 6A) and FDT (Fig 6B), or the percentage of patients
In the second study (NCT00168350), the overall results (as with progression of glaucomatous optic nerve damage by
prespecified per protocol) were similar to those in the first study, stereoscopic optic disc photographs (Fig 6C), between the
except that the cumulative probability of glaucomatous visual field memantine 10-mg or 20-mg and placebo groups. Overall, pro-
progression with memantine 10 mg was statistically significantly gression of glaucomatous findings assessed by SAP, FDT, or

Figure 2. Bar graphs showing glaucoma progression at 48 months in the intent-to-treat population of the first study (NCT00141882) based on (A) standard
automated perimetry, (B) frequency doubling technology, and (C) stereoscopic optic disc assessments. The survival analysis using cumulative probability of
progression according to predefined intervals was performed per the life table method. For the primary analysis (standard automated perimetry), a statistical
adjustment for multiple between-group comparisons was made to control for an overall type-I error rate of 5%, using Hochberg’s procedure. yP < 0.05,
compared with placebo. aCrude rate, that is, number of patients whose disease progressed (as determined by the reading center) of the total number of
patients during the course of the study. The reading center confirmed progression if 1 or more of the following was observed: increased or new appearance of
excavation, neuroretinal rim thinning or notching, or retinal nerve fiber layer defect. mem ¼ memantine.

1879
Ophthalmology Volume 125, Number 12, December 2018

memantine 10-mg and placebo groups (Table 4). Memantine was


well tolerated and treatment-related AEs reported by at least 5%
of patients in 1 treatment group (i.e., dizziness [P < 0.001,
between-group comparison], as well as headache, fatigue, and
insomnia [P  0.204, between-group comparisons]) were mild to
moderate in severity. The most commonly reported serious AEs
(regardless of causality) included prostate cancer, coronary artery
disease, pneumonia, chest pain, atrial fibrillation, osteoarthritis,
increased IOP, and congestive cardiac failure, of which only
prostate cancer was reported in statistically significantly more pa-
tients in the memantine 20-mg per day group, compared with the
placebo group (Table 4). Notably, there were no treatment-related
deaths during the 48-month study.
Consistent with the overall frequency of AEs, dizziness was
most frequently recorded as the cause for discontinuing study
treatment (Table 5). Headache, fatigue, balance disorder, and
pneumonia, in addition to dizziness, were reported by statistically
significantly more patients in the memantine 20-mg group than
the placebo group (P  0.042; Table 5). Notably, there was no
change from baseline that was statistically significant between
treatment groups when the following parameters were analyzed:
Figure 3. Bar graph showing glaucoma progression at 48 months in biomicroscopy and ophthalmoscopy findings with 1 severity
nonelow-tension glaucoma patients of the first study (NCT00141882) grade increase, best-corrected visual acuity, mean IOP, and
during treatment, using the frequency doubling technology. The survival average central corneal thickness. In addition, analysis of standard
analysis using cumulative probability of progression according to predefined laboratory safety variables and vital signs did not reveal any
intervals was performed per the life table method. yP < 0.05, compared clinically significant treatment effects.
with placebo. mem ¼ memantine.

stereoscopic optic disc photographs did not reveal a consistent


Discussion
protective effect of memantine, and the primary endpoint was not
met in either study or in the pooled analysis. These phase 3 studies were designed to test whether treat-
ment with oral memantine (10 or 20 mg) would result in
either significantly fewer patients exhibiting glaucomatous
Safety and Tolerability progression or significant delays in progression, and thus
Dizziness was the most frequently reported AE (regardless of whether memantine would provide any neuroprotective ef-
causality) in the pooled analysis. Considering all AEs reported by fect at these doses. However, based on 24-2 SAP, FDT, and
at least 5% of patients during treatment in any group, dizziness was optic disc photograph assessments, long-term daily treat-
also the only AE reported by statistically significantly more pa- ment with memantine did not slow or prevent progression of
tients in the memantine 20-mg treatment group, compared with the visual field deterioration or glaucomatous optic nerve

Figure 4. Bar graphs showing glaucoma progression at 48 months in the intent-to-treat population of the second study (NCT00141882) based on (A)
standard automated perimetry, (B) frequency doubling technology, and (C) stereoscopic optic disc assessments (per protocol). The survival analysis using
cumulative probability of progression according to predefined intervals was performed per the life table method. For the primary analysis (standard automated
perimetry), a statistical adjustment for multiple between-group comparisons was made to control for an overall type-I error rate of 5%, using Hochberg’s
procedure. yP < 0.05, compared with placebo. aCrude rate, that is, number of patients whose disease progressed (as determined by the reading center) of the
total number of patients during the course of the study. The reading center confirmed progression if 1 or more of the following was observed: increased or
new appearance of excavation, neuroretinal rim thinning or notching, or retinal nerve fiber layer defect. mem ¼ memantine.

1880
Weinreb et al 
Oral Memantine in Glaucoma

Figure 5. Bar graphs showing glaucoma progression at 48 months in nonelow-tension glaucoma patients of the second study (NCT00168350) during
treatment, using (A) frequency doubling technology, (B) standard automated perimetry, and (C) stereoscopic optic disc assessments (per protocol
amendment). The survival analysis using cumulative probability of progression according to predefined intervals was performed per the life table method.
y
P < 0.05, compared with placebo and memantine (mem) 20 mg. aCrude rate, that is, number of patients whose disease progressed (as determined by the
reading center) of the total number of patients during the course of the study. The reading center confirmed progression if 1 or more of the following was
observed: increased or new appearance of excavation, neuroretinal rim thinning or notching, or retinal nerve fiber layer defect. mem ¼ memantine.

damage in this population of patients with glaucoma at risk increased frequency of prostate cancer in the 20-mg treat-
of progression. Although there were differences between the ment group. The finding was thus considered a spurious
20-mg and 10-mg dose groups versus placebo using result.
different endpoints in each study, overall, there were no Despite not meeting the primary endpoint, these large,
consistent or clinically meaningful outcomes. In addition, randomized, double-masked, placebo-controlled, 4-year
pooled results of visual field and optic disc analyses studies provide valuable insights regarding the selection of
revealed no consistent or beneficial effects of memantine. the study population and their risk factors, as well as the
Overall, the safety profile of memantine was mostly methodologies for future development of neuroprotective
consistent with published information.29e36 Regarding the agents in glaucoma. For example, it is possible that initiating
increased incidence of prostate cancer, additional analyses memantine treatment earlier in the course of disease or in a
were performed using relevant data from all male patients population defined by more restrictive progression risk
(including possible confounding factors), but an extensive factors (e.g., age) may have produced different effects.
investigation of these data in conjunction with preclinical Similarly, central corneal thickness, presence of disc hem-
and postmarketing information could not explain the orrhage, and type of topical IOP-lowering medications used

Figure 6. Bar graphs showing glaucoma progression at 48 months in the pooled analysis of nonelow-tension glaucoma patients during treatment based on
(A) standard automated perimetry, (B) frequency doubling technology, and (C) stereoscopic optic disc assessments. The survival analysis using cumulative
probability of progression according to predefined intervals was performed per the life table method. aCrude rate, that is, number of patients whose disease
progressed (as determined by the reading center) of the total number of patients during the course of the study. The reading center confirmed progression if
1 or more of the following was observed: increased or new appearance of excavation, neuroretinal rim thinning or notching, or retinal nerve fiber layer
defect. CI ¼ confidence interval; mem ¼ memantine; RR ¼ risk reduction.

1881
Ophthalmology Volume 125, Number 12, December 2018

Table 4. Adverse Events Reported by 5% or More of Patients During Treatment in Any Group and Serious Adverse Events Reported by
1% or More of Patients During Treatment in Any Group*

Memantine 20 mg (n [ 934)y Memantine 10 mg (n [ 648)y Placebo (n [ 640)y P Valuez


Adverse events reported by 5% of patients in 1 group during treatment, no. (%)
Dizziness 212 (23) 104 (16) 86 (13) < 0.001
IOP increased 142 (15) 97 (15) 112 (18) 0.372
Cataract 122 (13) 101 (16) 101 (16) 0.223
Headache 121 (13) 66 (10) 77 (12) 0.243
Hypertension 115 (12) 88 (14) 71 (11) 0.398
Fatigue 80 (9) 47 (7) 48 (8) 0.581
Visual acuity reduced 78 (8) 52 (8) 57 (9) 0.846
Dry eye 77 (8) 40 (6) 35 (6) 0.073
Nasopharyngitis 76 (8) 56 (9) 58 (9) 0.808
Sinusitis 73 (8) 46 (7) 50 (8) 0.845
Blepharitis 72 (8) 51 (8) 57 (9) 0.671
Vision blurred 71 (8) 46 (7) 46 (7) 0.917
Ocular hyperemia 66 (7) 32 (5) 45 (7) 0.182
Depression 64 (7) 35 (5) 35 (6) 0.382
Nausea 63 (7) 36 (6) 37 (6) 0.570
Insomnia 62 (7) 46 (7) 59 (9) 0.144
Eye pain 62 (7) 31 (5) 46 (7) 0.167
Upper respiratory tract infection 58 (6) 42 (7) 37 (6) 0.870
Bronchitis 54 (6) 34 (5) 38 (6) 0.850
Conjunctival hyperemia 51 (6) 40 (6) 43 (7) 0.578
Back pain 50 (5) 44 (7) 43 (7) 0.398
Influenza 50 (5) 41 (6) 35 (6) 0.688
Blood pressure increased 49 (5) 24 (4) 32 (5) 0.337
Optic disc hemorrhage 48 (5) 41 (6) 42 (7) 0.428
Constipation 48 (5) 27 (4) 30 (5) 0.668
Hypercholesterolemia 47 (5) 45 (7) 45 (7) 0.166
Drug hypersensitivity 47 (5) 44 (7) 39 (6) 0.325
Urinary tract infection 41 (4) 41 (6) 46 (7) 0.049
Arthritis 46 (5) 33 (5) 40 (6) 0.486
Arthralgia 39 (4) 29 (5) 43 (7) 0.057
Eye pruritus 41 (4) 27 (4) 41 (6) 0.111
Serious adverse events reported by 1.0% of patients in 1 group during treatment, no. (%)
Prostate cancer 17 (1.8) 6 (0.9) 3 (0.5) 0.039
Coronary artery disease 16 (1.7) 12 (1.9) 8 (1.3) 0.663
Pneumonia 12 (1.3) 8 (1.2) 6 (0.9) 0.806
Chest pain 11 (1.2) 6 (0.9) 5 (0.8) 0.723
Atrial fibrillation 9 (1.0) 6 (0.9) 5 (0.8) 0.928
Osteoarthritis 9 (1.0) 5 (0.8) 9 (1.4) 0.510
Intraocular pressure increased 6 (0.6) 3 (0.5) 7 (1.1) 0.409x
Cardiac failure congestive 5 (0.5) 8 (1.2) 5 (0.8) 0.310x

IOP ¼ intraocular pressure.


*ClinicalTrials.gov identifiers, NCT00141882 and NCT00168350.
y
Per protocol, safety analyses are based on data obtained during treatment (i.e., from patients on treatment and up to 30 days after the last dose). As a result,
40, 16, and 20 patients were excluded from the memantine 20-mg, memantine 10-mg, and placebo groups, respectively.
z
Pearson chi-square test, unless otherwise indicated.
x
Fisher exact test.

at baseline could potentially impact the response to treat- and analytical developments for assessing structural and
ment, and the effects of these factors should be explored in functional measures of disease severity and progression
future studies. It is also worth considering the possibility could provide much needed sensitivity, or greater speci-
that the 20-mg daily dose was too low to provide adequate ficity, and help to establish clinically applicable endpoints
neuroprotective activity and that a 30-mg dose might have that could be used in future studies of neuroprotective
been preferable, especially if administered as 15-mg tablets agents.37e43 In addition, based on the increased sensitivity,
twice daily to minimize AEs. it may be possible to determine study outcomes in a
Although the study used both functional and structural shorter time using these endpoints. However, early studies
measures of progression, additional tests (either functional exploring these novel endpoints would require confirma-
or structural) that further support the confirmation of tion of effects with more classical endpoints to assess their
progression may have been valuable. Recent technological validity.

1882
Weinreb et al 
Oral Memantine in Glaucoma

Table 5. Discontinuations Because of Adverse Events Reported by More than 0.5% of Patients in 1 Group During Treatment in Any
Group*

Adverse Event Memantine 20 mg (n [ 934)y Memantine 10 mg (n [ 648)y Placebo (n [ 640)y P Valuez


Dizziness 48 (5.1) 14 (2.2) 9 (1.4) < 0.001
Headache 19 (2.0) 5 (0.8) 5 (0.8) 0.036
Fatigue 15 (1.6) 3 (0.5) 4 (0.6) 0.042
Nausea 10 (1.1) 3 (0.5) 5 (0.8) 0.413
Depression 8 (0.9) 4 (0.6) 2 (0.3) 0.413x
Balance disorder 7 (0.7) 1 (0.2) 0 (0.0) 0.036x
Pneumonia 7 (0.7) 0 (0.0) 0 (0.0) 0.008x
Insomnia 6 (0.6) 2 (0.3) 4 (0.6) 0.714x
Constipation 6 (0.6) 1 (0.2) 1 (0.2) 0.274x
Asthenia 5 (0.5) 4 (0.6) 0 (0.0) 0.148x
Vertigo 5 (0.5) 4 (0.6) 2 (0.3) 0.803x
Death 4 (0.4) 4 (0.6) 3 (0.5) 0.928x
Stomach discomfort 1 (0.1) 1 (0.2) 4 (0.6) 0.161x

*ClinicalTrials.gov identifiers, NCT00141882 and NCT00168350.


y
Per protocol, safety analyses are based on data obtained during treatment (i.e., from patients on treatment and up to 30 days after the last dose). As a result,
40, 16, and 20 patients were excluded from the memantine 20-mg, memantine 10-mg, and placebo groups, respectively.
z
Pearson chi-square test, unless otherwise indicated.
x
Fisher exact test.

Limitations and challenges of the studies included the E.L.), respectively. The authors also thank the numerous other
fact that they were long-term studies with potentially con- team members who provided essential support throughout the
founding factors. For example, treatments for IOP lowering duration of these studies.
were based on standard of care and at the discretion of
investigator, with no requirements to measure IOP at a References
specific time of day per a predefined process, as is
customary in glaucoma studies of IOP-lowering medication.
The fact that LTG was not defined per protocol should also 1. Weinreb RN, Aung T, Medeiros FA. The pathophysiology and
treatment of glaucoma: a review. JAMA. 2014;311:
be considered, because it may have led to variations
1901e1911.
between centers in classification of these patients and may 2. American Academy of Ophthalmology. Preferred Practice
have contributed to the studies outcomes. Another limita- Pattern in Primary Open-Angle Glaucoma. San Francisco,
tion stems from the fact that patients were required to CA: Elsevier; 2015.
discontinue study treatment after HFA-based progression 3. Early Manifest Glaucoma Trial Group, Heijl A, Bengtsson B,
was declared. This would have affected the ability of other et al. Natural history of open-angle glaucoma. Ophthalmology.
measures to detect progression that may have occurred after 2009;116:2271e2276.
the discontinuation of study medication. However, as 4. The AGIS Investigators. The Advanced Glaucoma Interven-
evidenced by continued preclinical44e48 and clinical (clin- tion Study (AGIS): 7. The relationship between control of
icaltrials.gov identifier, NCT02862938) investigations, intraocular pressure and visual field deterioration. Am J Oph-
thalmol. 2000;130:429e440.
neuroprotection as a treatment strategy in glaucoma remains
5. Ocular Hypertension Treatment Study Group, Kass MA,
a viable and important concept, and methodologies,49 Heuer DK, et al. The Ocular Hypertension Treatment
biomarkers,50e55 or both that allow assessment of pro- Study: a randomized trial determines that topical ocular
gression over much shorter periods, with better sensitivity hypotensive medication delays or prevents the onset of
to detect changes, will likely benefit future studies. primary open-angle glaucoma. Arch Ophthalmol. 2002;120:
In this large population of patients with open-angle 701e713.
glaucoma at risk of progression, daily treatment with 6. Early Manifest Glaucoma Trial Group, Leske MC, Heijl A,
memantine for 48 months was not shown to prevent glau- et al. Factors for glaucoma progression and the effect of
comatous progression. treatment: the early manifest glaucoma trial. Arch Ophthalmol.
2003;121:48e56.
Acknowledgments 7. Musch DC, Gillespie BW, Lichter PR, et al. Visual field
progression in the Collaborative Initial Glaucoma Treatment
The authors thank all the investigators who participated in these Study: the impact of treatment and other baseline factors.
studies (listed in the online supplemental material available at Ophthalmology. 2009;116:200e207.
www.aaojournal.org), as well as Victor Marciano (current 8. Weinreb RN, Levin LA. Is neuroprotection a viable therapy for
employee of Allergan) and Janet Cheetham, Harry Cui, Scott glaucoma? Arch Ophthalmol. 1999;117:1540e1544.
Whitcup and Erik Lippa (employees of Allergan at the time the 9. Siliprandi R, Lipartiti M, Fadda E, et al. Activation of the
studies were conducted) for their significant roles in data glutamate metabotropic receptor protects retina against
management (V.M.), the clinical program (J.C.), statistical N-methyl-D-aspartate toxicity. Eur J Pharmacol. 1992;219:
analyses (H.C.), and evaluations of data and endpoints (S.W., 173e174.

1883
Ophthalmology Volume 125, Number 12, December 2018

10. Sucher NJ, Lei SZ, Lipton SA. Calcium channel antagonists 28. Mi XS, Yuan TF, So KF. The current research status of normal
attenuate NMDA receptor-mediated neurotoxicity of retinal tension glaucoma. Clin Interv Aging. 2014;9:1563e1571.
ganglion cells in culture. Brain Res. 1991;551:297e302. 29. Grossberg GT, Manes F, Allegri RF, et al. The safety, toler-
11. Sucher NJ, Wong LA, Lipton SA. Redox modulation of ability, and efficacy of once-daily memantine (28 mg): a
NMDA receptor-mediated Ca2þ flux in mammalian central multinational, randomized, double-blind, placebo-controlled
neurons. Neuroreport. 1990;1:29e32. trial in patients with moderate-to-severe Alzheimer’s disease
12. Sucher NJ, Lipton SA, Dreyer EB. Molecular basis of gluta- taking cholinesterase inhibitors. CNS Drugs. 2013;27:
mate toxicity in retinal ganglion cells. Vision Res. 1997;37: 469e478.
3483e3493. 30. Handforth A, Bordelon Y, Frucht SJ, Quesada A. A pilot ef-
13. Doganay S, Cankaya C, Alkan A. Evaluation of corpus gen- ficacy and tolerability trial of memantine for essential tremor.
iculatum laterale and vitreous fluid by magnetic resonance Clin Neuropharmacol. 2010;33:223e226.
spectroscopy in patients with glaucoma; a preliminary study. 31. Keck Jr PE, Hsu H-A, Papadakis K, Russo Jr J. Memantine
Eye (Lond). 2012;26:1044e1051. efficacy and safety in patients with acute mania associated with
14. Naskar R, Vorwerk CK, Dreyer EB. Concurrent down- bipolar I disorder: a pilot evaluation. Clin Neuropharmacol.
regulation of a glutamate transporter and receptor in glaucoma. 2009;32:199e204.
Invest Ophthalmol Vis Sci. 2000;41:1940e1944. 32. Lovera JF, Frohman E, Brown TR, et al. Memantine for
15. Maguire G, Simko H, Weinreb RN, Ayoub G. Transport- cognitive impairment in multiple sclerosis: a randomized
mediated release of endogenous glutamate in the vertebrate placebo-controlled trial. Mult Scler. 2010;16:715e723.
retina. Pflugers Arch. 1998;436:481e484. 33. Rao N, Chou T, Ventura D, Abramowitz W. Investigation of
16. Park CK, Cha J, Park SC, et al. Differential expression of the pharmacokinetic and pharmacodynamic interactions be-
two glutamate transporters, GLAST and GLT-1, in an tween memantine and glyburide/metformin in healthy young
experimental rat model of glaucoma. Exp Brain Res. subjects: a single-center, multiple-dose, open-label study. Clin
2009;197:101e109. Ther. 2005;27:1596e1606.
17. Chen HS, Lipton SA. Mechanism of memantine block of 34. Villoslada P, Arrondo G, Sepulcre J, et al. Memantine induces
NMDA-activated channels in rat retinal ganglion cells: un- reversible neurologic impairment in patients with MS.
competitive antagonism. J Physiol. 1997;499:27e46. Neurology. 2009;72:1630e1633.
18. Parsons CG, Danysz W, Bartmann A, et al. Amino-alkyl- 35. Wilcock G, Möbius HJ, Stöffler A. A double-blind, placebo-
cyclohexanes are novel uncompetitive NMDA receptor an- controlled multicentre study of memantine in mild to moder-
tagonists with strong voltage-dependency and fast blocking ate vascular dementia (MMM500). Int Clin Psychopharmacol.
kinetics: in vitro and in vivo characterization. Neurophar- 2002;17:297e305.
macology. 1999;38:85e108. 36. Allergan plc. Namenda XRdhighlights of prescribing informa-
19. Hare W, WoldeMussie E, Lai R, et al. Efficacy and safety of tion. https://www.allergan.com/assets/pdf/namenda_pi; 2003.
memantine, an NMDA-type open-channel blocker, for reduc- Accessed June 14, 2017.
tion of retinal injury associated with experimental glaucoma in 37. Yucel YH, Gupta N. A framework to explore the visual brain
rat and monkey. Surv Ophthalmol. 2001;45(Suppl 3): in glaucoma with lessons from models and man. Exp Eye Res.
S284eS289. 2015;141:171e178.
20. Hare WA, WoldeMussie E, Lai RK, et al. Efficacy and safety 38. Yu M, Lin C, Weinreb RN, et al. Risk of visual field pro-
of memantine treatment for reduction of changes associated gression in glaucoma patients with progressive retinal nerve
with experimental glaucoma in monkey, I: functional mea- fiber layer thinning: a 5-year prospective study. Ophthal-
sures. Invest Ophthalmol Vis Sci. 2004;45:2625e2639. mology. 2016;123:1201e1210.
21. Hare WA, WoldeMussie E, Weinreb RN, et al. Efficacy and 39. Pathak M, Demirel S, Gardiner SK. Reducing variability of
safety of memantine treatment for reduction of changes asso- perimetric global indices from eyes with progressive glaucoma
ciated with experimental glaucoma in monkey, II: structural by censoring unreliable sensitivity data. Transl Vis Sci Tech-
measures. Invest Ophthalmol Vis Sci. 2004;45:2640e2651. nol. 2017;6:11.
22. WoldeMussie E, Yoles E, Schwartz M, et al. Neuroprotective 40. Kim EK, Park H-YL, Park CK. Segmented inner plexiform
effect of memantine in different retinal injury models in rats. layer thickness as a potential biomarker to evaluate open-angle
J Glaucoma. 2002;11:474e480. glaucoma: dendritic degeneration of retinal ganglion cell.
23. Yücel YH, Zhang Q, Gupta N, et al. Loss of neurons in PLoS One. 2017;12:e0182404.
magnocellular and parvocellular layers of the lateral geniculate 41. Medeiros FA. Biomarkers and surrogate endpoints: lessons
nucleus in glaucoma. Arch Ophthalmol. 2000;118:378e384. learned from glaucoma. Invest Ophthalmol Vis Sci. 2017;58:
24. International Headache Society. International Classification of BI020eBI026.
Headache Disorders. 1st ed. http://www.ihs-headache.org/ 42. Shoji T, Zangwill LM, Akagi T, et al. Progressive macula
ichd-guidelines; 1988. Accessed June 14, 2017. vessel density loss in primary open-angle glaucoma: a longi-
25. Chauhan BC, House PH, McCormick TA, LeBlanc RP. tudinal study. Am J Ophthalmol. 2017;182:107e117.
Comparison of conventional and high-pass resolution peri- 43. De Moraes CG, Hood DC, Thenappan A, et al. 24-2 Visual
metry in a prospective study of patients with glaucoma and fields miss central defects shown on 10-2 tests in glaucoma
healthy controls. Arch Ophthalmol. 1999;117:24e33. suspects, ocular hypertensives, and early glaucoma. Ophthal-
26. Heijl A, Lindgren G, Lindgren A, et al. Extended empirical sta- mology. 2017;124:1449e1456.
tistical package for evaluation of single and multiple fields in 44. Mead B, Amaral J, Tomarev S. Mesenchymal stem cell-
glaucoma: Statpac 2. In: Mills RP, Heijl A, eds. Perimetry Update derived small extracellular vesicles promote neuroprotection
1990/1991. Amsterdam/New York: Kugler Publications; 1991. in rodent models of glaucoma. Invest Ophthalmol Vis Sci.
27. Ocular Hypertension Treatment Study Group, Parrish II RK, 2018;59:702e714.
Schiffman JC, et al. Test-retest reproducibility of optic disk 45. Chou TH, Musada GR, Romano GL, et al. Anesthetic pre-
deterioration detected from stereophotographs by masked conditioning as endogenous neuroprotection in glaucoma. Int J
graders. Am J Ophthalmol. 2005;140:762e764. Mol Sci. 2018;19.

1884
Weinreb et al 
Oral Memantine in Glaucoma

46. Akopian A, Kumar S, Ramakrishnan H, et al. Targeting 51. Oddone F, Roberti G, Micera A, et al. Exploring serum levels
neuronal gap junctions in mouse retina offers neuroprotection of brain derived neurotrophic factor and nerve growth factor
in glaucoma. J Clin Invest. 2017;127:2647e2661. across glaucoma stages. PLoS One. 2017;12:e0168565.
47. Anders F, Liu A, Mann C, et al. The small heat shock protein 52. Shahidi AM, Hudson C, Tayyari F, Flanagan JG. Retinal ox-
alpha-crystallin B shows neuroprotective properties in a ygen saturation in patients with primary open-angle glaucoma
glaucoma animal model. Int J Mol Sci. 2017;18. using a non-flash hyperspectral camera. Curr Eye Res.
48. Su W, Li Z, Jia Y, et al. microRNA-21a-5p/PDCD4 axis 2017;42:557e561.
regulates mesenchymal stem cell-induced neuroprotection in 53. Stefansson E, Olafsdottir OB, Einarsdottir AB, et al. Retinal
acute glaucoma. J Mol Cell Biol. 2017;9:289e301. oximetry discovers novel biomarkers in retinal and brain dis-
49. Vianna JR, Chauhan BC. How to detect progression in eases. Invest Ophthalmol Vis Sci. 2017;58:BI0227eBI0233.
glaucoma. In: Bagetta G, Nucci C, eds. New Trends in 54. Tanito M, Kaidzu S, Takai Y, Ohira A. Association between
Basic and Clinical Research in Glaucoma: A Neurodegen- systemic oxidative stress and visual field damage in open-
erative Disease of the Visual SystemdPart B. Amsterdam/ angle glaucoma. Sci Rep. 2016;6:25792.
Waltham: Elsevier; 2015:135e150. 55. Zhang S, Wang B, Xie Y, et al. Retinotopic changes in the
50. Cordeiro MF, Normando EM, Cardoso MJ, et al. Real-time gray matter volume and cerebral blood flow in the primary
imaging of single neuronal cell apoptosis in patients with visual cortex of patients with primary open-angle glaucoma.
glaucoma. Brain. 2017;140:1757e1767. Invest Ophthalmol Vis Sci. 2015;56:6171e6178.

Footnotes and Financial Disclosures


Originally received: January 5, 2018. L.M.Z.: Consultant e Merck (Kenilworth, New Jersey), Optovue; Financial
Final revision: May 16, 2018. support e National Eye Institute, Heidelberg Engineering, Carl Zeiss
Accepted: June 12, 2018. Meditec, Topcon (Oakland, New Jersey), Optovue.
Available online: August 3, 2018. Manuscript no. 2018-26. S.S.: Employee e Allergan plc (Irvine, California).
1
Hamilton Glaucoma Center, Shiley Eye Institute and Department of H.B.: Employee e Allergan plc (Irvine, California).
Ophthalmology, University of California San Diego, La Jolla, California.
M.B.: Employee e Allergan plc (Irvine, California).
2
Edward S. Harkness Eye Institute, Columbia University Medical Center, Supported by Allergan plc, Dublin, Ireland. The sponsor participated in the
New York, New York. design of the study, data analysis and interpretation, revision of the
3
Discipline of Ophthalmology, University of Sydney and Sydney Eye manuscript for intellectual content, and decision to submit the manuscript
Hospital, Sydney, Australia. for publication. Writing and editorial assistance was provided to the authors
4
UC Davis Eye Center, University of California, Davis, Sacramento, by Michele Jacob, PhD, CMPP, of Envision Pharma Group, Philadelphia,
California. PA, and funded by Allergan plc, Dublin, Ireland.
5
Department of Ophthalmology, University of Iowa, Iowa City, Iowa. HUMAN SUBJECTS: Human subjects were included in this study, which
6
Allergan plc, Irvine, California. was conducted in accordance with the tenets of the Declaration of Helsinki,
the International Conference on Harmonisation guidelines for Good
Financial Disclosure(s):
The author(s) have made the following disclosure(s): R.N.W.: Consultant e Clinical Practice, and all privacy requirements applicable at the time.
Aerie Pharmaceuticals (Irvine, California), Alcon (Fort Worth, Texas), Institutional Review Board/Ethics Committee approval was obtained and all
Allergan (Dublin, Ireland), Bausch & Lomb (Rochester, New York), patients provided written informed consent before any study related
procedure or changes in treatment, as well as other local required docu-
Eyenovia (Reno, Nevada), Novartis (Basel, Switzerland), Sensimed (Lau-
sanne, Switzerland), Unity (Brisbane, California), Valeant (Laval, Canada); mentation of consent.
Financial Support e Carl Zeiss Meditec (Dublin, California), Centervue No animal subjects were included in this study.
(Padova, Italy), Genentech (San Francisco, California), Heidelberg Engi- Author Contributions:
neering (Franklin, Massachusetts), Konan (Irvine, California), National Eye Conception and design: Weinreb, Cioffi, Johnson, Zangwill
Institute (Bethesda, Maryland), Optos (Marlborough, Massachusetts),
Analysis and interpretation: Weinreb, Liebmann, Cioffi, Goldberg, Brandt,
Optovue (Fremont, California), Tomey (Nagoya, Japan). Johnson, Schneider, Badger, Bejanian
J.M.L.: Consultant e Aerie Pharmaceuticals, Alcon, Allergan, Bausch & Data collection: Weinreb, Liebmann, Cioffi, Goldberg, Brandt, Johnson,
Lomb, Carl Zeiss Meditec, FORSIGHT Vision5 (Menlo Park, California), Zangwill
Heidelberg Engineering, Inotek Pharmaceuticals (Lexington, Massachu-
setts), Quark Pharmaceuticals (Ness Ziona, Israel), Reichert (Depew, New Obtained funding: N/A
York), Sustained Nano Systems (Westhampton Beach, New York); Overall responsibility: Weinreb, Liebmann, Cioffi, Johnson, Zangwill,
Financial support e Heidelberg Engineering, National Eye Institute; Equity Schneider, Badger, Bejanian
owner e Diopsys Corporation (Pine Brook, New Jersey), FORSIGHT Abbreviations and Acronyms:
Vision5, SOLX (Waltham, Massachusetts), Sustained Nano Systems. AE ¼ adverse event; FDT ¼ frequency doubling technology;
I.G.: Advisory board e Novartis, Allergan, Pfizer (New York, New York), HFA ¼ Humphrey field analyzer; IOP ¼ intraocular pressure; LTG ¼ low-
Lecturer e Mundipharma (Cambridge, United Kingdom), Financial tension glaucoma; NMDA ¼ N-methyl-D-aspartate; OAG ¼ open-angle
support e Pfizer, Novartis. glaucoma; SAP ¼ standard automated perimetry; SD ¼ standard deviation.
J.D.B.: Financial support e Forsight Vision Labs (Menlo Park, California); Correspondence:
(acquired by Allergan in 2016). Robert N. Weinreb, MD, Shiley Eye Institute, University of California San
C.A.J.: Consultant e JAEB Center (Tampa, Florida), Centervue, Haag- Diego, 9415 Campus Point Drive, La Jolla, CA 92093. E-mail: rweinreb@
Streit (Köniz, Switzerland), Aerie Pharmaceuticals, Regeneron (Tarry- ucsd.edu.
town, New York), M&S Technologies (Stokie, Illinois).

1885

You might also like