Context.— Postherpetic neuralgia (PHN) is a syndrome
of often intractable neuropathic pain following herpes zoster
(shingles) that eludes effective treatment in many patients.
Objective.— To determine the efficacy and safety of the
anticonvulsant drug gabapentin in reducing PHN pain.
Design.— Multicenter, randomized, double-blind,
placebo-controlled, parallel design, 8-week trial conducted from August
1996 through July 1997.
Setting.— Sixteen US outpatient clinical centers.
Participants.— A total of 229 subjects were randomized.
Intervention.— A 4-week titration period to a maximum dosage
of 3600 mg/d of gabapentin or matching placebo. Treatment was
maintained for another 4 weeks at the maximum tolerated dose.
Concomitant tricyclic antidepressants and/or narcotics were continued
if therapy was stabilized prior to study entry and remained constant
throughout the study.
Main Outcome Measures.— The primary efficacy measure was
change in the average daily pain score based on an 11-point Likert
scale (0, no pain; 10, worst possible pain) from baseline week to the
final week of therapy. Secondary measures included average daily sleep
scores, Short-Form McGill Pain Questionnaire (SF-MPQ), Subject Global
Impression of Change and investigator-rated Clinical Global Impression
of Change, Short Form-36 (SF-36) Quality of Life Questionnaire, and
Profile of Mood States (POMS). Safety measures included the frequency
and severity of adverse events.
Results.— One hundred thirteen patients received
gabapentin, and 89 (78.8%) completed the study; 116 received placebo,
and 95 (81.9%) completed the study. By intent-to-treat analysis,
subjects receiving gabapentin had a statistically significant reduction
in average daily pain score from 6.3 to 4.2 points compared with a
change from 6.5 to 6.0 points in subjects randomized to receive placebo
(P<.001). Secondary measures of pain as well as changes in
pain and sleep interference showed improvement with gabapentin
(P<.001). Many measures within the SF-36 and POMS also
significantly favored gabapentin (P≤.01). Somnolence,
dizziness, ataxia, peripheral edema, and infection were all more
frequent in the gabapentin group, but withdrawals were comparable in
the 2 groups (15 [13.3%] in the gabapentin group vs 11 [9.5%] in
the placebo group).
Conclusions.— Gabapentin is effective in the
treatment of pain and sleep interference associated with PHN. Mood and
quality of life also improve with gabapentin
therapy.
HERPES ZOSTER (shingles) is accompanied in the majority of
patients by intense pain that is variously described as "burning,"
"deeply aching," "tearing," "electric shocklike," and
"lancinating." Abnormalities of sensation in affected dermatomes
are common, including hyperpathia or allodynia. While herpes
zoster–associated pain tends to resolve spontaneously with
time,1 some patients suffer from chronic, debilitating
neuropathic pain that persists beyond the resolution of visible
cutaneous manifestations of the underlying viral (varicella zoster)
eruption. Postherpetic neuralgia (PHN) is most commonly
defined as pain in the area affected by herpes zoster at least 3 months
after crusting of the herpes zoster rash. Once established, PHN may
persist for many years.
In the United States, more than a million new cases of herpes
zoster arise each year.1 Approximately 10% to 15% of all
people with herpes zoster develop PHN. The age distribution of its
victims, however, includes a disproportionate number of the elderly;
nearly half of older patients (≥60 years) with herpes zoster will have
enduring neuropathic pain.1-3 Postherpetic neuralgia is
usually refractory to simple analgesic therapies, and treatment most
often is pharmacologic, including a wide variety of drugs and routes
of delivery.4,5 The most commonly used agents are oral
medications. Currently, the standard treatment for PHN is with various
tricyclic antidepressants (TCAs) (amitriptyline, desipramine, and
clomipramine) either as monotherapy6-12 or in combination
with other medications, such as carbamazepine or
opioids.13-15 Unfortunately, only about 50% of patients
treated with TCAs for PHN in clinical trials experience pain relief in
the absence of intolerable adverse effects.16
Recent reviews and meta-analyses of 11 randomized, controlled
clinical trials
for PHN concluded that TCAs appeared to be the
only agents that provided reliable pain relief.16,17
Tricyclic antidepressants, however, can also have significant adverse
effects, such as arrhythmias, postural hypotension, sedation, dry
mouth, constipation, confusion, and urinary retention. Their use is not
appropriate in many patients with cardiovascular disease, which makes
the use of these agents problematic in the 60 years and older age
group, in which PHN is most prevalent. A single nonopioid agent that
provides both substantial relief and a good safety profile is thus
needed. Gabapentin (1-[aminomethyl]-cyclohexaneacetic acid;
Neurontin, Parke-Davis, Division of Warner-Lambert Co, Morris Plains,
NJ) is a structural analog of γ-aminobutyric acid (GABA) that has
shown some promise as such an agent.
Gabapentin is lipophilic and penetrates the blood-brain barrier. Its
mechanism of action has not yet been fully elucidated, but appears not
to involve binding to GABA receptors18 and is distinct from
that of TCAs. Introduced in the United States in 1994 as an
anticonvulsant, gabapentin is used clinically to benefit patients with
epilepsy by reducing seizure frequency when added to conventional
antiepileptic drug regimens.18-21 Preclinical studies have
documented an analgesic effect of gabapentin in several rat models,
including models of chronic neuropathic pain.22-25
Gabapentin has been reported anecdotally to relieve pain in patients
with intractable neuropathic pain and reflex sympathetic dystrophy,
allowing the reduction or termination of other analgesic medications
and relieving symptoms associated with painful disease
manifestations.26-29 It also reduced spontaneous pain and
tactile allodynia in patients with peripheral or central
pain.30 We report here the results of a large, multicenter,
double-blind, placebo-controlled trial of gabapentin for the pain
of PHN.
Study Design and Subjects
Subjects from 16 clinical centers participated in this
double-blind, randomized, placebo-controlled, parallel design, 8-week
trial, which was conducted from August 26, 1996, through July 14, 1997.
The protocol and informed consents were approved by the institutional
review board of each participating center. The study period included a
1-week baseline period followed by a 4-week titration period, during
which a maximum tolerated dose (up to 3600 mg/d) was established for
each subject. This was followed by a 4-week stable dosing period.
Subjects were seen for a minimum of 5 scheduled visits: an initial
enrollment visit (screening), a randomization visit (week 0, at the end
of an intervening baseline week), and 3 subsequent visits after 2,
4, and 8 weeks of study treatment.
Inclusion criteria included the following: at least 18
years of age; pain present for more than 3 months after healing of a
herpes zoster skin rash; a pain intensity score of at least 40 mm on
the 100-mm Visual Analog Scale on the Short-Form McGill Pain
Questionnaire (SF-MPQ) at screening and at randomization; average daily
diary pain score of at least 4 (on a scale of 0-10) during the baseline
week; and discontinuance of muscle relaxants, anticonvulsants,
mexiletine, topical analgesics, and antiviral agents beginning at least
2 weeks prior to screening. Previously prescribed TCAs and/or narcotics
could be continued if therapy was stabilized prior to study entry and
remained constant throughout the study. At the screening visit,
exclusion criteria included the following: prior treatment with
gabapentin or demonstrated hypersensitivity to the drug or its
ingredients; neurolytic or neurosurgical therapy for PHN;
immunocompromised state; significant hepatic or renal insufficiency;
significant hematological disease; severe pain other than that caused
by PHN; use of experimental drugs or participation in a clinical study
within 2 months of screening; a history of illicit drug or alcohol
abuse within the last year; and any serious or unstable medical or
psychological condition.
Eligible subjects who gave informed written consent underwent
physical and neurological examinations, and blood samples were taken
for routine hematology and chemistry. Medical histories and
demographics were obtained. Subjects completed the SF-MPQ and were
instructed on the completion of daily diaries that would assess overall
pain and sleep.
At the second visit 1 week later (baseline, week 0),
physical and neurological examinations were updated, subjects again
completed the SF-MPQ, the Short Form-36 (SF-36) Quality of Life
Questionnaire, and Profile of Mood States (POMS). Diaries were
collected and reviewed. Subjects who continued to meet the inclusion
and exclusion criteria and who had completed at least 4 diaries were
randomized to receive gabapentin or placebo.
At each subsequent visit (weeks 2, 4, and 8), subjects completed
the SF-MPQ, diaries were collected, and adverse events assessed. At the
final visit (week 8), subjects again underwent physical and
neurological examinations and completed the SF-36 and POMS. Subjects
completed Global Impression of Change and investigators completed
Clinical Global Impression of Change Questionnaires. Blood samples were
taken for routine hematology and chemistry, and plasma concentration of
gabapentin was determined. In the case of early termination, week 8
assessments were completed at the last study visit.
Randomization and Treatment
Randomization was performed by producing a randomization schedule that
assigned each subject number to gabapentin or matching placebo in a 1:1
manner. Gabapentin and placebo were provided as identically appearing
capsules and were packaged in subject-specific bottles based on the
randomization schedule. As subjects enrolled at each site, they were
sequentially assigned a subject number.
Study participants who at week 0 were randomly assigned to receive
gabapentin began with an initial dose of 300 mg/d. The number of
capsules (300 mg of gabapentin per capsule) taken daily increased over
the next 4 weeks (titration period) in a step-up manner (900, 1800,
2400, and 3600 mg/d divided 3 times a day), to a maximum total dose of
3600 mg/d, regardless of whether efficacy was achieved at a lower dose,
or until the subject developed intolerable adverse effects. In the
latter event, dosage was decreased 1 level and continued at that level
for the remainder of the study. If 1800 mg/d was not tolerated, a
minimal dose of 1200 mg/d was permitted. The dose established during
the titration period was maintained throughout the remainder of the
study. Matching placebo capsules were similarly administered.
Data Collection
and Statistical Analysis
Statistical analyses for efficacy were conducted on 2
populations. The intent-to-treat population included those subjects
who, once randomized to treatment, had evidence of taking at least 1
dose of study medication and provided at least 1 follow-up efficacy
assessment. The efficacy-evaluable population consisted of those
subjects who, in addition to meeting the criteria required for the
intent-to-treat population, met strict protocol-specified criteria
regarding study medication compliance, use of concomitant medications,
and number of daily diaries returned.
The primary efficacy parameter—change in average daily pain score from
the baseline week to the final study week—was evaluated from daily
pain diaries and measured on an 11-point Likert scale having as end
points 0 ("no pain") and 10 ("worst possible pain"). The
minimum treatment group difference in change from baseline that was
considered clinically meaningful was 1.5
points. Given the assumption that the SD for
this parameter would be 3.4, a sample size of 80 evaluable patients in
each treatment was required to provide 80% power to detect this
difference with a 5% error rate for a 2-sided test.
The change from baseline week to final study week in average daily pain
score was determined by calculating, for each patient evaluated, the
average daily pain score for the baseline week and the final study
week. The change in average daily pain score was then calculated as the
difference between the 2 time periods. A secondary parameter—the
change from baseline in average daily sleep rating score—was
determined in the same manner.
Other secondary parameters were the SF-MPQ total score and the
affective and sensory subscores. Each domain of the SF-36 and POMS was
also evaluated. The change from baseline in each of the SF-MPQ, SF-36,
and POMS assessments was determined. Additional secondary parameters
were the present pain intensity (PPI) score from the SF-MPQ and the
investigator-rated Clinical Global Impression of Change and Subjects'
Global Impression of Change.
Between-treatment comparisons for all the change from baseline
parameters were accomplished by an analysis of covariance (ANCOVA)
model, including fixed terms of treatment, center, treatment by center
interaction, and baseline scoring as a covariate. A preliminary test of
normality examining change in average daily pain score in the
intent-to-treat population showed that the data for this population
were not normally distributed. Therefore, data for the dependent
variable were rank-transformed. The Cochran-Mantel-Haenszel test was
used to assess PPI at the final visit and the investigator-rated
Clinical Global Impression of Change and Subjects' Global Impression
of Change. For the evaluation of PPI, the test took baseline values
into account. All P values reported are 2-tailed and no
adjustments were made for multiple comparisons.
A summary profile of the trial is presented in Figure 1, following guidelines recommended by the
Consolidated Standards of Reporting Trials statement.31 A
total of 229 subjects were randomized during the baseline visit. Of
these, 184 (80.3%) completed the study, 15.3% discontinued the study
because of adverse events, and the remaining 4.4% discontinued because
of other reasons. The 2 treatment groups were comparable with respect
to the proportion of patients who discontinued because of adverse
events (P=.20) and the proportion of patients
who completed the study (P=.62). Of those
subjects treated with gabapentin who achieved stable dosing, 83.3%
received at least 2400 mg/d and 65.0% received 3600 mg/d.
Results for both populations (intent-to-treat and efficacy
evaluable) per treatment group were similar for all parameters
analyzed. The results presented here are for the intent-to-treat
population only, which is a more conservative approach in analyzing
these data.
Demographic and baseline clinical characteristics at
randomization are presented in Table 1. There were
no statistically significant differences in distribution of sex, age,
or race between the gabapentin and placebo treatment groups. Similarly,
there were no statistically significant differences between the
treatment groups in time since last herpes zoster eruption, baseline
average daily pain score, prior PHN medications, or concomitant
medications.
The average daily pain score was significantly reduced at the end of
the study
in the gabapentin population (33.3% reduction)
compared with the placebo population (7.7% reduction) (Table 2). At the end of week 8 (or the final week), the
gabapentin population showed an average daily pain score of 4.2
(decrease of 2.1) vs the placebo population with an average daily pain
score of 6.0 (decrease of 0.5) (P<.001). A comparison of
change in average daily pain score over the course
of the 8 weeks (Figure 2) shows this reduction was established at
week 2, with a further reduction at week 4. At week 8, pain reduction
was maintained at the week 4 level.
Secondary Efficacy Parameters
Subjects who received gabapentin reported significantly
improved average daily sleep rating scores over their placebo
counterparts (P<.001) (Table 2). Mean scores on the SF-MPQ
were also markedly improved for total pain (P<.001), as well
as for the 2 components of this measure, sensory pain
(P<.001) and affective pain (P<.001) (Table 2). The SF-MPQ ratings of PPI likewise were statistically significantly
improved among subjects treated with gabapentin (P<.01).
This included a rating of "no pain" at the final week in 16.0% of
subjects treated with gabapentin compared with 8.8% of subjects
treated with placebo.
The Subjects' Global Impression of Change Questionnaire
indicated that gabapentin had provided valuable pain relief for many
subjects. At the end of the study, 43.2% of subjects treated with
gabapentin categorized their pain as much or moderately improved
compared with 12.1% of the subjects treated with placebo (Figure 3). The majority of subjects receiving placebo
(59.5%) reported no change in their level of pain in contrast to
22.9% of the subjects treated with gabapentin. The investigator-rated
Clinical Global Impression of Change showed similar results (Figure 4).
On the SF-36, measures relating to physical functioning,
role-physical, bodily pain, vitality, and mental health all showed
gabapentin to be superior to placebo (P≤.01; Table 3). Similarly, subjects treated with gabapentin
showed significantly greater improvement than subjects treated with
placebo in the POMS assessments of depression-dejection,
anger-hostility, fatigue-inertia, and confusion-bewilderment, as well
as in total mood disturbance (P≤.01; Table 3).
Measures of frequency, nature, and severity of adverse events were
derived from a total of 229 subjects, of whom 113 had received
gabapentin and 116 placebo. Minor adverse events that were deemed
associated with the study medication were reported in a total of 62
subjects (54.9%) receiving gabapentin and 32 subjects (27.6%)
receiving placebo. No serious adverse events that were determined by
the investigator to be related to gabapentin were reported. One death
occurred in the placebo group during the study and was considered
unrelated to the study medication. Overall, the most frequently
reported adverse effects among the gabapentin group, which occurred at
higher incidences than those in the placebo group, were somnolence
(27.4% vs 5.2%), dizziness (23.9% vs 5.2%), ataxia (7.1% vs
0.0%), peripheral edema (9.7% vs 3.4%), and infection (8.0% vs
2.6%). Subjects in the older age range did not experience more of the
central nervous system–related adverse effects of dizziness,
somnolence, and ataxia than subjects in the younger age range. The most
frequently reported adverse event noted in the placebo group was pain:
10.3% compared with 4.4% of subjects in the gabapentin group.
A total of 15 (13.3%) and 11 (9.5%) of subjects in the gabapentin and
placebo treatment groups, respectively, withdrew from the study for
adverse events described as related to the study medication. Dizziness
led to withdrawal from the study of 6 subjects (5.3%) treated with
gabapentin, while somnolence led to the withdrawal of 5 subjects
(4.4%) treated with gabapentin. In the placebo group, 2 subjects
(1.7%) withdrew from the study because of somnolence, and there were
no withdrawals because of dizziness.
The results of our study clearly show that gabapentin reduces PHN pain
compared with placebo. This was demonstrated on several measures of
pain and as assessed by subjects as well as
investigators. For the primary outcome variable,
change in average daily pain score, the actual calculated power of the
study in demonstrating the predicted level of efficacy of gabapentin
approached 100%. In addition, several secondary outcome measures
also showed gabapentin as superior to placebo. Sleep, several
quality-of-life measures, and several mood state variables were
significantly improved by gabapentin therapy. Significant improvement
was evident during the titration phase (at the 2-week time period) and
continued to accrue over the course of 8 weeks of treatment. Adverse
effects of gabapentin were minor and well tolerated, consisting
primarily of somnolence and dizziness. These 2 adverse effects
accounted for most of the adverse event–related withdrawals. Despite
doses of gabapentin up to 3600 mg/d in a population with an average age
of 73 years, no serious drug-related adverse events were reported. In
clinical practice, adverse effects such as these can be managed by a
slower upward titration, dose reduction, and use of lower maximum doses
than allowed in the clinical trial protocol. Serious adverse events,
especially of a cardiovascular nature, were not evident. Overall,
gabapentin reduced PHN pain with a very acceptable adverse effect
profile.
The mechanism of action of gabapentin remains uncertain. Spinal cord
neuronal calcium channels play a potentially important role in chronic
neuropathic pain and are modulated by gabapentin. Analgesia through
GABAergic neurotransmission effects is much less certain. Despite the
uncertainty regarding the mechanism of action of gabapentin, the drug
has been shown effective in rat models of chronic neuropathic
pain.22-25 Results of treatment of PHN can likely be
predicted by testing in preclinical neuropathic pain models because PHN
is not only common, but has consistent symptomatology, a clear cause,
and consistent neuropathology.
The current standard of treatment for PHN with oral medications
are the TCAs. Nontricyclic antidepressants with better adverse effect
and safety profiles, including the selective serotonin reuptake
inhibitors, have not been proven equivalent to TCAs in terms of
efficacy.16 In the elderly population afflicted with PHN,
therapy with TCAs is frequently either contraindicated (usually for
cardiovascular reasons) or poorly tolerated because of excessive
sedation, cognitive impairment, dry mouth, constipation, sexual
dysfunction, and orthostatic lightheadedness. Other approaches with
good safety and evidence of efficacy, such as topical local
anesthetics and topical aspirin-nonsteroidal anti-inflammatory agents,
are either unproven in long-term use or not commercially
available.2 Topical capsaicin produces a modest improvement
in pain after long-term use, but has a high
rate of burning sensations that are unacceptably
severe.1 Opioids are frequently used to treat PHN in
clinical practice, but do not yet have adequate support from
placebo-controlled studies of long-term use. Many of the problematic
adverse effects of TCAs also pertain to the use of opioids, such as
sedation, cognitive impairment, and constipation. In addition, many
patients and physicians are reluctant to use medications that carry the
stigma of being addicting, despite the lack of evidence that this is a
problem in the PHN population.
From the safety and efficacy evidence in our study, a strong case can
be made for considering gabapentin as a first-line oral medication for
management of PHN pain. There are no studies directly comparing
gabapentin with TCAs. From published systematic reviews of
antidepressants and anticonvulsants for neuropathic pain by McQuay et
al,32 comparisons of safety and efficacy can be made by
calculating the number needed to treat (NNT) for both parameters. The
NNT is the reciprocal of the difference in the percentage of patients
improved or harmed by active therapy compared with control therapy,
expressed for benefit as 1/[(% improved active)−(%
improved placebo)]. For placebo-controlled TCA studies of PHN pain,
the NNT for benefit ranged from 1.9 to 4.1; for minor adverse events,
the NNT ranged from 1.7 to 8.8; and for adverse events leading to study
withdrawal, the NNT ranged from 13 to 37. From the data in Figure 4 and
the text, the gabapentin NNT for benefit is 3.2, the NNT for minor
adverse events is 3.7, and the NNT for adverse events leading to study
withdrawal is 25. From this perspective, gabapentin should be
considered at least as effective as TCAs, at least as safe, and with
fewer contraindications to use.
Tricyclic antidepressants and opioids each have a different mechanism
of action than gabapentin. Tricyclic antidepressants may relieve pain
through serotonin and norepinephrine reuptake blockade, by blockade of
α-adrenergic receptors, by sodium channel-blocking effects, and by
relief of depression. Opioids relieve pain through activation of a
family of specific receptors found in both the central and peripheral
nervous systems. Because of its straightforward pharmacokinetics and
relative lack of adverse drug interactions, multidrug regimens to
control chronic neuropathic pain can include gabapentin if gabapentin
monotherapy fails. In summary, based on the results of this 8-week
study, gabapentin can be added to the list of first-line
medications for treatment of chronic neuropathic pain syndromes such as
PHN.
1.Watson CPN, Tyler KL, Bickers DR, Millikan LE, Smith S, Coleman E. A randomized vehicle-controlled trial of topical
capsaicin in the treatment of postherpetic neuralgia.
Clin
Ther.1993;15:510-526.Google Scholar 2.Kost RG, Straus SE. Postherpetic
neuralgia—pathogenesis, treatment, and prevention.
N Engl J
Med.1996;335:32-42.Google Scholar 3.Herne K, Cirelli R, Lee P, Tyring SK. Antiviral therapy
of acute herpes zoster in older patients.
Drugs Aging.1996;8:97-112.Google Scholar 4.Johnson RW. Aspects of postherpetic neuralgia: can we
zap Z-aP?
Pain Rev.1996;3:117-135.Google Scholar 5.Watson CPN. The treatment of postherpetic neuralgia.
Neurology.1995;45(suppl 8):S58-S60.Google Scholar 6.Kishore-Kumar R, Max MB, Schafer SC.
et al. Desipramine
relieves postherpetic neuralgia.
Clin Pharmacol Ther.1990;47:305-312.Google Scholar 7.Max MB, Schafer SC, Culnane BSN, Smoller B, Dubner R, Gracely RH. Amitriptyline, but not lorazepam, relieves postherpetic
neuralgia.
Neurology.1988;38:1427-1432.Google Scholar 8.Max MB. Treatment of post-herpetic neuralgia:
antidepressants.
Ann Neurol.1994;35:S50-S53.Google Scholar 9.Takeda Y, Kobayashi H, Suzuki K, Sei Y, Nishio K, Ishizaki H. Analgesic effects of clomipramine (Anafranil) on
postherpetic neuralgia.
Skin Res.1988;30:33-36.Google Scholar 10.Watson CPN, Evans RJ, Reed K, Merskey H, Goldsmith L, Warsh J. Amitriptyline versus placebo in postherpetic neuralgia.
Neurology.1982;32:671-673.Google Scholar 11.Watson CPN, Evans RJ. A comparative trial of
amitriptyline and zimelidine in post-herpetic neuralgia.
Pain.1985;23:387-394.Google Scholar 12.Watson CP, Chipman M, Reed K, Evans RJ, Birkett N. Amitriptyline versus maprotiline in postherpetic neuralgia: a
randomized, double-blind, crossover trial.
Pain.1992;48:29-36.Google Scholar 13.Bowsher D. Postherpetic neuralgia and its treatment: a
retrospective survey of 191 patients.
J Pain Symptom
Manage.1996;12:290-299.Google Scholar 14.Göbel H, Stadler T. Treatment of post-herpes
zoster pain with tramadol: results of an open pilot study versus
clomipramine with or without levomepromazine.
Drugs.1997;53(suppl 2):34-39.Google Scholar 15.Gerson GR, Jones RB, Luscombe DK. Studies on the
concomitant use of carbamazepine and clomipramine for the relief of
post-herpetic neuralgia.
Postgrad Med J.1977;53(suppl 4):104-109.Google Scholar 16.Kingery WS. A critical review of controlled clinical
trials for peripheral neuropathic pain and complex regional pain
syndromes.
Pain.1997;73:123-139.Google Scholar 17.Volmink J, Lancaster T, Gray S, Silagy C. Treatments
for postherpetic neuralgia—a systematic review of randomized
controlled trials.
Fam Pract.1996;13:84-91.Google Scholar 18.Goa KL, Sorkin EM. Gabapentin: a review of its
pharmacological properties and clinical potential in epilepsy.
Drugs.1993;46:409-427.Google Scholar 19.Bruni J, Saunders M, Anhut H, Sauermann W. Efficacy and
safety of gabapentin (Neurontin): a multicenter, placebo-controlled,
double-blind study.
Neurology.1991;41(suppl 1):330-331.Google Scholar 20.UK Gabapentin Study Group. Gabapentin in partial
epilepsy.
Lancet.1990;335:1114-1117.Google Scholar 21.US Gabapentin Study Group. Gabapentin as add-on therapy
in refractory partial epilepsy: a double-blind, placebo-controlled,
parallel-group study.
Neurology.1993;43:2292-2298.Google Scholar 22.Gillin S, Sorkin LS. Gabapentin reverses the allodynia
produced by the administration of anti-GD2 ganglioside, an
immunotherapeutic drug.
Anesth Analg.1998;86:111-116.Google Scholar 23.Hwang JH, Yaksh TL. Effect of subarachnoid gabapentin
on tactile-evoked allodynia in a surgically induced neuropathic pain
model in the rat.
Reg Anesth.1997;22:249-256.Google Scholar 24.Singh L, Field MJ, Ferris P.
et al. The
antiepileptic agent gabapentin (Neurontin) possesses anxiolytic-like
and antinociceptive actions that are reversed by D-serine.
Psychopharmacology.1996;127:1-9.Google Scholar 25.Xiao W-H, Bennett GJ. Gabapentin has an antinociceptive
effect mediated via a spinal site of action in a rat model of painful
peripheral neuropathy.
Pain.1996;2:267-273.Google Scholar 26.Mellick GA, Mellicy LB, Mellick LB. Gabapentin in the
management of reflex sympathetic dystrophy.
J Pain Symptom
Manage.1995;10:265-266.Google Scholar 27.Rosner H, Rubin L, Kestenbaum A. Gabapentin adjunctive
therapy in neuropathic pain states.
Clin J Pain.1996;12:56-58.Google Scholar 28.Stacey BR, Owen GT, Sinclair JD, Glick RM, Tipton KD. Gabapentin for neuropathic pain states: a case series report
[Abstract].
Reg Anesth.1996;21:S65.Google Scholar 29.Rosenberg JM, Harrell C, Ristic H, Werner R, de Rosayro AM. The effect of gabapentin on neuropathic pain.
Clin J Pain.1997;13:251-255.Google Scholar 30.Attal N, Parker F, Brasseur L, Chauvin M, Bouhassira D. Efficacy of gabapentin on neuropathic pain: a pilot study. In: Program
and abstracts of the 16th Annual Scientific Meeting of the American
Pain Society; October 23-26, 1997; New Orleans, La. Abstract 653.
31.Begg C, Cho M, Eastwood S.
et al. Improving the quality
of reporting of randomized controlled trials: the CONSORT statement.
JAMA.1996;276:637-639.Google Scholar 32.McQuay H, Carroll D, Jadad AR, Wiffen P, Moore A. Anticonvulsant drugs for management of pain: a systematic review.
BMJ.1995;311:1047-1052.Google Scholar