Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Pregabalin for the treatment of postherpetic neuralgia : A

randomized, placebo-controlled trial


R.H. Dworkin, A.E. Corbin, J.P. Young, Jr., et al.
Neurology 2003;60;1274
DOI 10.1212/01.WNL.0000055433.55136.55

This information is current as of December 16, 2012

The online version of this article, along with updated information and services, is
located on the World Wide Web at:
http://www.neurology.org/content/60/8/1274.full.html

Neurology ® is the official journal of the American Academy of Neurology. Published continuously
since 1951, it is now a weekly with 48 issues per year. Copyright © 2003 by AAN Enterprises, Inc. All
rights reserved. Print ISSN: 0028-3878. Online ISSN: 1526-632X.
CME Pregabalin for the treatment
of postherpetic neuralgia
A randomized, placebo-controlled trial
R.H. Dworkin, PhD; A.E. Corbin, MS; J.P. Young Jr., MS; U. Sharma, PhD; L. LaMoreaux, MPH;
H. Bockbrader, PhD; E.A. Garofalo, MD; and R.M. Poole, MD

Abstract—Objective: To evaluate the efficacy and safety of pregabalin in the treatment of postherpetic neuralgia (PHN).
Methods: The authors conducted a multicenter, parallel-group, double-blind, placebo-controlled, 8-week, randomized
clinical trial in PHN, defined as pain for 3 or more months following herpes zoster rash healing. Patients (n ⫽ 173) were
randomized to treatment with pregabalin or placebo. Patients randomized to pregabalin received either 600 mg/day
(creatinine clearance ⬎ 60 mL/min) or 300 mg/day (creatinine clearance 30 to 60 mL/min). The primary efficacy measure
was the mean of the last seven daily pain ratings. Secondary endpoints included additional pain ratings, sleep interfer-
ence, quality of life, mood, and patient and clinician ratings of global improvement. Results: Pregabalin-treated patients
had greater decreases in pain than patients treated with placebo (endpoint mean scores 3.60 vs 5.29, p ⫽ 0.0001). Pain
was significantly reduced in the pregabalin-treated patients after the first full day of treatment and throughout the study,
and significant improvement on the McGill Pain Questionnaire total, sensory, and affective pain scores was also found.
The proportions of patients with ⱖ30% and ⱖ50% decreases in mean pain scores were greater in the pregabalin than in
the placebo group (63% vs 25% and 50% vs 20%, p ⫽ 0.001). Sleep also improved in patients treated with pregabalin
compared to placebo (p ⫽ 0.0001). Both patients and clinicians were more likely to report global improvement with
pregabalin than placebo (p ⫽ 0.001). Given the maximal dosage studied, pregabalin had acceptable tolerability compared
to placebo despite a greater incidence of side effects, which were generally mild to moderate in intensity. Conclusions:
Treatment of PHN with pregabalin is safe, efficacious in relieving pain and sleep interference, and associated with greater
global improvement than treatment with placebo.
NEUROLOGY 2003;60:1274 –1283

The most common complication of herpes zoster in of animal models of neuropathic and nociceptive
immunocompetent patients is postherpetic neuralgia pain.14,15 It has a predictable pharmacokinetic pro-
(PHN).1 PHN has been defined as pain persisting at file, few drug interactions, and a rapid onset of ac-
least 3 months after healing of the herpes zoster tion,16,17 and it has been found to be safe and
rash,2-4 and physical disability, social withdrawal, efficacious in patients with painful diabetic neuropa-
and psychological distress are common sequelae.1,2,5 thy.18 The objective of the current study was to test
Tricyclic antidepressants were the first agents to the hypothesis that pregabalin has an analgesic ef-
demonstrate efficacy in randomized controlled trials fect in patients with PHN. In addition, the effects of
in PHN and were considered first-line therapy for treatment on sleep interference, quality of life, mood,
many years.6,7 Recently, anticonvulsant,8,9 opioid,10,11 and patient and clinician ratings of the patients’
and topical analgesics12 have been found to have sig- global improvement were examined.
nificant beneficial effects. Many patients, however,
are refractory to these and other treatments because Methods. Patients. Study participants included men and
women of any race who were at least 18 years of age and had PHN
of inadequate pain relief or intolerable side effects.1,13 defined as pain present for more than 3 months after healing of a
Thus, additional safe and effective treatments are herpes zoster skin rash. Patients were considered eligible if their
needed for patients with PHN. pain was at least 40 mm on the 100 mm visual analog scale (VAS)
Pregabalin [CI-1008 or (S)-3-isobutyl GABA, (S)- of the Short-Form McGill Pain Questionnaire (SF-MPQ)19 at base-
line and randomization visits and they also completed at least
(⫹)-3-(aminomethyl)-5-methylhexanoic acid] is an four daily pain diaries and had a minimum mean daily pain rating
␣2-␦ ligand that has been found effective in a variety of 4 on an 11-point numerical pain rating scale during the base-

From the University of Rochester School of Medicine and Dentistry (Dr. Dworkin), Rochester, NY; and Pfizer Global Research and Development (A.E. Corbin,
J.P. Young, Dr. Sharma, L. LaMoreaux, Dr. Bockbrader, and Dr. Garofalo), Ann Arbor, MI, and (Dr. Poole) New London, CT.
Supported by Pfizer Global Research and Development, Ann Arbor, MI. R.H.D. received a research grant from Pfizer for participating in the clinical trial
described in this article but was not compensated for article preparation; he has received research grants, consulting fees, or speakers’ bureau honoraria in
the past year from Akros Pharma, AstraZeneca, Elan Pharmaceuticals, Endo Pharmaceuticals, GlaxoSmithKline, King Pharmaceuticals, NeurogesX,
Novartis, Ortho-McNeil Pharmaceutical, Pfizer, Reliant Pharmaceuticals, and UCB Pharma; consulting fees and speakers’ bureau honoraria in excess of
$10,000 were received from Novartis and Pfizer.
Received February 19, 2002. Accepted in final form December 18, 2002.
Address correspondence and reprint requests to Dr. Robert H. Dworkin, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box
604, Rochester, NY 14642; e-mail: robert_dworkin@urmc.rochester.edu

1274 Copyright © 2003 by AAN Enterprises, Inc.


line week preceding randomization. Women were neither preg- plied in bottles, and patients were instructed to take one capsule
nant nor lactating, and those of childbearing age had a confirmed three times daily. Study medication was packaged and labeled
negative serum pregnancy test at baseline and practiced an appro- with sequential randomization numbers according to a random-
priate method of contraception throughout the study. A normal ization schedule generated with a block size of four. An investiga-
chest X-ray within the preceding 2 years was required for partici- tor could break the blind for an individual patient if a medical
pation. All participating clinical sites received investigational re- emergency occurred and the Pfizer Clinical Research Department
view board approval of the study protocol before patient staff could not be reached. The code was to be broken only in cases
enrollment, and all patients provided written informed consent where it was necessary for proper treatment of the patient. One
before study participation. patient was hospitalized with a stroke on study day 36 and the
Patients were generally free of serious or unstable medical code was subsequently broken for this patient. No other code
conditions and were excluded if they had any other severe pain breaks occurred. The blind was maintained until after the study
that might confound assessment or self-evaluation of pain due to was completed and all decisions regarding data evaluability had
PHN or had previous neurolytic or neurosurgical therapy for been made.
PHN. Patients who had failed to respond to previous PHN treat- Efficacy measurements. The primary efficacy measure was
ment with gabapentin at dosages ⱖ1,200 mg/day were also ex- pain reduction, as recorded by patients in a daily diary using the
cluded from participation in the trial. In addition, patients were 11-point numerical pain rating scale described above. Each morn-
excluded if their baseline serum creatinine clearance was ⱕ30 ing, patients rated the intensity of their PHN pain during the
mL/min, white blood cell count was ⬍2,500/mm3, neutrophil count preceding 24-hour period.
was ⬍1,500/mm3, or platelet count was ⬍100 ⫻ 103/mm3. Patients In addition, there were several secondary measures of efficacy.
were also excluded if they had participated in any other pregaba- SF-MPQ. The SF-MPQ19 was completed by the patient at
lin clinical trial, or if they had participated in any other clinical baseline, at the start of treatment, and at the end of treatment
trial of an investigational drug within 30 days before screening. weeks 1, 3, 5, and 8. This questionnaire has three parts: the first
Certain existing medications were permitted, provided dosing assesses pain quality and yields a sensory score (sum of 11 adjec-
was stable for at least 30 days before baseline and remained tives—throbbing, shooting, stabbing, sharp, cramping, gnawing,
stable throughout the study. Permitted medications included nar- hot-burning, aching, heavy, tender, and splitting— each rated on
cotic and non-narcotic analgesics, acetaminophen (not to exceed 4 an intensity scale with 0 ⫽ none, 1 ⫽ mild, 2 ⫽ moderate, and 3 ⫽
g/day), nonsteroidal anti-inflammatory drugs, aspirin, and antide- severe), an affective score (sum of four adjectives—tiring-
pressants, including selective serotonin reuptake inhibitors. Pro- exhausting, sickening, fearful, and punishing-cruel—rated on the
hibited medications, requiring at least a 7-day washout period same intensity scale), and a total score (sum of the sensory and
before baseline, included benzodiazepines, skeletal muscle relax- affective scores). The second part of the SF-MPQ consists of a
ants, orally administered steroids, local and topical agents for 100-mm VAS of pain intensity that patients used to rate their
relief of PHN, and anticonvulsants. Patients using gabapentin pain during the preceding week. The third part of the SF-MPQ is
were required to discontinue its use at least 7 days before the a measure of present pain intensity (PPI) using a six-point scale
start of study medication. Injected local anesthetics or steroids
(0 ⫽ none, 1 ⫽ mild, 2 ⫽ discomforting, 3 ⫽ distressing, 4 ⫽
were not permitted within 1 month of baseline.
horrible, 5 ⫽ excruciating).
Study design. The study was a multicenter (29 centers),
Daily sleep interference score. This measure was used to as-
parallel-group, double-blind, randomized clinical trial of pregaba-
sess the degree to which pain interfered with sleep during the
lin vs placebo in the symptomatic treatment of PHN that con-
preceding 24-hour period on an 11-point numerical rating scale
sisted of a 1-week baseline phase and an 8-week treatment phase.
(0 ⫽ did not interfere with sleep, 10 ⫽ completely interfered with
To achieve equivalent pregabalin exposure, treatment was strati-
sleep). It was included in the daily diary and was completed by the
fied on baseline creatinine clearance. Patients with a creatinine
patient each day after awakening.
clearance ⬎60 mL/min received pregabalin 600 mg/day (200 mg
three times daily) or placebo, whereas patients with a creatinine Medical Outcomes Study (MOS) Sleep Scale. The MOS Sleep
clearance ⬎30 and ⱕ60 mL/min received pregabalin 300 mg/day Scale21 is a patient-rated questionnaire that assesses key aspects
(100 mg three times daily) or placebo. Based on pharmacokinetic of sleep and provides an overall sleep problem index; higher scores
modeling studies, this dosage adjustment results in comparable on this measure reflect worse sleep. The MOS Sleep Scale was
steady-state concentrations of pregabalin, allowing these strata to administered at the randomization and termination visits.
be combined as a single pregabalin treatment arm for the purpose SF-36 Health Survey. The SF-36 Health Survey22 is a self-
of analyzing pain relief.20 administered, 36-item measure of quality of life that assesses the
Baseline phase. During the 1-week baseline phase, patients patient’s level of functioning in eight health domains: physical
rated the intensity of their PHN pain over the previous 24 hours functioning, role-physical (role limitations caused by physical
using an 11-point numerical pain rating scale (0 ⫽ no pain, 10 ⫽ problems), social functioning, bodily pain, mental health, role-
worst possible pain). At the end of the baseline phase, those pa- emotional (role limitations caused by emotional problems), vital-
tients who had completed at least four entries in the daily pain ity, and general perception of health. This questionnaire was
diary, who had a mean daily pain score of four or greater during administered at the randomization and termination visits and
the preceding 7 days, and who met all other criteria were random- was scored in the standard manner.22 For each domain of function-
ized to treatment. ing, higher scores reflect better quality of life.
Treatment phase. Patients were randomized on a double- Profile of Mood States (POMS). The POMS23 consists of a list
blind basis to either pregabalin or placebo and titrated to a stable of 65 descriptors of mood that are each rated on a five-point scale
target dosage. All patients assigned to the pregabalin arm were by patients (0 ⫽ applies not at all, 4 ⫽ applies extremely) and that
administered 150 mg/day (50 mg three times daily) for the first 3 yield six scores—tension-anxiety, depression-dejection, anger-
days of treatment and 300 mg/day (100 mg three times daily) for hostility, vigor-activity, fatigue-inertia, and confusion-bewilder-
the remainder of the first treatment week. Beginning at the start ment—as well as an overall mood disturbance score. Except for
of the second week, those patients assigned to pregabalin who the vigor-activity scale, lower scores indicate better mood. The
were in the higher creatinine clearance group (⬎60 mL/min) were POMS was administered at the randomization and termination
administered 600 mg/day (200 mg three times daily). Clinic visits visits.
occurred at baseline, at the start of randomized treatment, and at Patient Global Impression of Change (PGIC). The PGIC24
the end of weeks 1, 3, 5, and 8. Study medication dosage remained consists of a self-evaluation by the patient of his or her overall
stable after the first week until the end of the 8-week treatment change since the beginning of the study rated on a seven-point
phase. Following this double-blind treatment phase, or after early scale (1 ⫽ very much improved; 2 ⫽ much improved; 3 ⫽ mini-
termination, patients were given the option to continue treatment mally improved; 4 ⫽ no change; 5 ⫽ minimally worse; 6 ⫽ much
with pregabalin in an open-label extension study at a starting worse; 7 ⫽ very much worse). The PGIC was administered at the
dosage of 300 mg/day. A follow-up visit was scheduled for patients termination visit.
not continuing with open-label treatment. Clinical Global Impression of Change (CGIC). The CGIC24
Study medications. All medications were administered orally consists of an evaluation by the clinician of the patient’s overall
and on a double-blind basis. Placebo capsules were identical in change since the beginning of the study, rated on the same seven-
appearance to capsules containing pregabalin. Capsules were sup- point scale as the PGIC at the termination visit.
April (2 of 2) 2003 NEUROLOGY 60 1275
Safety assessments. The safety of pregabalin treatment was specific, and validated high-performance liquid chromatographic
assessed by comparing the groups with regard to the incidence method with ultraviolet detection.29 A population pharmacokinetic
and intensity of adverse events and their relationship to study analysis was performed using the nonlinear mixed effects model-
drug as assessed by the investigator, and by comparing treatment ing program NONMEM, version V.30 The analysis assumed a 6-,
groups with respect to changes in physical and neurologic exami- 6-, and 12-hour dosing interval for the three times daily dosage
nations, vision testing (visual field screening, dilated ophthalmos- regimen. Empirical Bayesian estimates of individual pharmacoki-
copy, and visual acuity), 12-lead electrocardiogram, and clinical netic values (apparent oral plasma pregabalin clearance [CL/F]
laboratory evaluations throughout the study. Adverse events in- and apparent pregabalin volume of distribution [Vd/F]) were ob-
cluded any concurrent illness, sign, or symptom reported by the tained using the POSTHOC option in the NONMEM estimation
patient, the patient’s family, or the investigator. An adverse event step. As blood sampling for determining plasma pregabalin con-
that was not present at baseline or one that increased in intensity centrations was not performed at the same time for all patients,
or frequency relative to baseline was considered treatment emer- predicted steady-state morning trough concentrations (Cminss)
gent. All patients randomized to treatment were included in the and average steady-state concentrations (Cavg) were also esti-
safety analyses. mated using the POSTHOC option to permit comparisons among
Statistical analyses. Power analysis. Treatment effect was patients. Mean, standard error, 95% CI, and percent relative SD
estimated from the results of published studies of gabapentin in for the estimates were calculated using WinNonlin Pro version 3,
the treatment of PHN8 and painful diabetic neuropathy,25 both of a validated pharmacokinetic estimation program.31
which used the same primary efficacy measure as the current The intent-to-treat population was evaluated for safety and the
study. The difference in endpoint mean pain scores between data were analyzed using descriptive statistical methods. The
groups was 1.8 in the PHN study and 1.3 in the painful diabetic number and percentage of patients in each treatment group who
neuropathy study; in both studies, the overall SD was 2.35. As- experienced a treatment-emergent adverse event were summa-
suming similar results for the current study, and employing two- rized. Associated adverse events were also summarized, and in-
sided testing at the 0.05 level, it was determined that 76 patients clude those events the investigator considered possibly, probably,
per treatment group would provide ⬎90% power to detect a differ- or definitely related to study medication, and those for which the
ence in endpoint mean pain scores ⱖ1.3. relationship was unknown.
Statistical analysis. All analyses were carried out using the
intent-to-treat population, defined as any patient who received at Results. Patient characteristics. There were 173 patients ran-
least one dose of study medication. Patients randomized to pre- domized to treatment: 84 to placebo and 89 to pregabalin. Patients
gabalin were analyzed as a single pregabalin treatment arm. Pa- were predominantly white (95%), generally above 65 years of age
tients with no data for a given efficacy measure at baseline or at (mean age ⫽ 71.5 years; SD ⫽ 10.9; 82% ⱖ65 years), and there
the timepoint analyzed were treated as missing rather than using were slightly more women than men (53% vs 47%). Creatinine
imputed values. clearance was ⬎60 mL/min for 68% of the patients in the study.
The primary efficacy analysis was conducted on the endpoint Thoracic dermatomes were the most commonly affected site of
mean pain scores; specifically, the means of the last seven daily PHN (48%), and the mean duration of PHN was 33.8 months.
diary pain ratings recorded by patients while taking study medi- Overall, the two treatment groups were similar with respect to
cation. This analysis was conducted with the last observations— demographic and clinical variables (table 1).
seven daily pain ratings— carried forward for those patients who The flow of patient participation is depicted in figure 1. Seventy-
did not complete the trial. Supplemental analyses of the primary six percent of all patients randomized to treatment completed the
efficacy measure examined pain scores for each day during the study (88% of patients in the placebo group and 65% of patients in
first treatment week, weekly mean pain scores, and the propor- the pregabalin group). Concurrent pain medications were used by
tions of patients whose endpoint mean pain scores were reduced 118 patients (68%). The most frequently used pain medications were
ⱖ30% and ⱖ50% compared with their baseline week mean pain acetylsalicylic acid (32%) and acetaminophen (14%). At study termi-
scores. nation, 62 pregabalin-treated patients (70%) and 63 placebo patients
Secondary efficacy analyses were conducted of the SF-MPQ (75%) entered the open-label phase of the study.
sensory, affective, total, VAS, and PPI scores at endpoint and at A total of 29 patients had possibly important variations from
weeks 1, 3, 5, and 8; mean sleep interference scores at endpoint the protocol. One patient was randomized to treatment with a
and at each week separately; and the MOS Sleep Scale sleep baseline mean pain score that was less than four and two patients
problem index, SF-36 domain scores, POMS scale scores, and had VAS scores ⬍40 at randomization. Two patients had CLcr
PGIC and CGIC ratings at the end of the study. For outcome values ⬍60 mL/min but were randomized to the CLcr ⬎60 mL/
measures collected in daily diaries (pain and sleep interference), minute stratum, and two patients had normal CLcr values but
the endpoint mean score was the mean of the last seven diary were randomized to the CLcr ⬍60 mL/min stratum. Of these four
entries while on study medication, including the entry on the day patients, three were randomized to placebo and only one was
after the last dose. If fewer than seven scores were recorded by randomized to pregabalin treatment; this patient did not report
endpoint, the available scores were used to determine a mean. any adverse events. All other protocol variations involved the use
All testing was two-sided and performed using SAS procedures of prohibited medications or failure to maintain a stable regimen
without adjustment for testing multiple measures.26 The mean of allowed medications; none was expected to affect the results of
daily pain scores, SF-MPQ scale scores, mean daily sleep interfer- the study.
ence scores, MOS Sleep Scale sleep problem index, eight SF-36 Primary efficacy measure. There was a decrease in mean pain
domain scores, and seven POMS scale scores were analyzed using scores at endpoint for patients treated with pregabalin compared
an analysis of covariance (ANCOVA) main effects model, with to placebo (p ⫽ 0.0001; table 2). This improvement in endpoint
treatment, center, and creatinine clearance strata (a dichotomous mean pain scores in pregabalin-treated patients compared to
indicator variable) in the model, and the corresponding baseline placebo-treated patients remained significant when the patients
measure as the covariate.27 In each case, adjusted (least squares) in each of the two creatinine clearance strata were analyzed
means were obtained from the model and 95% CI of the difference separately.
in least-squares means between pregabalin and placebo were con- Significantly greater pain relief was found with pregabalin
structed. The proportions of responders with ⱖ30% and ⱖ50% compared to placebo on the second day of the trial (figure 2),
reduction from baseline to endpoint mean pain score were ana- which was the first full day of treatment during the titration
lyzed using the Cochran-Mantel-Haenszel (CMH) test with table phase because not all patients had their full dosage on the day
scores,28 adjusting for center and creatinine clearance strata. The treatment was initiated. The superior pain relief with pregabalin
PGIC and CGIC were analyzed using the CMH test with modified treatment relative to placebo continued to be significant through-
ridit scores28 adjusting for center and creatinine clearance strata. out all 8 weeks of double-blind treatment (figure 3A).
Pharmacokinetic measures were derived from two venous Sixty-three percent of the pregabalin-treated patients vs 25%
blood samples collected from each patient, at 2 weeks following of placebo-treated patients reported reductions in pain of 30% or
attainment of assigned dose and at the end of the study. Time of more (p ⫽ 0.001), a clinically important degree of pain relief.32
blood sample collection relative to time of last dose was specified Fifty percent of patients treated with pregabalin and 20% of pa-
in the protocol. All plasma samples from all treatment groups tients treated with placebo had a ⱖ50% decrease in their pain,
were analyzed for pregabalin concentrations using a sensitive, and this difference between the two groups was also significant.
1276 NEUROLOGY 60 April (2 of 2) 2003
Table 1 Patient demographics and baseline disease characteristics

Characteristic Placebo, n ⫽ 84 Pregabalin, n ⫽ 89 All patients, n ⫽ 173

Sex, n (%)
Men 44 (52.4) 37 (41.6) 81 (46.8)
Women 40 (47.6) 52 (58.4) 92 (53.2)
Race, n (%)
White 82 (97.6) 82 (92.1) 164 (94.8)
Hispanic 1 (1.2) 6 (6.7) 7 (4.0)
Asian or Pacific Islander 1 (1.2) 1 (1.1) 2 (1.2)
Age, y, mean (SD) 70.5 (11.3) 72.4 (10.5) 71.5 (10.9)
Age, y, n (%)
18–64 17 (20.2) 15 (16.9) 32 (18.5)
ⱖ65 67 (79.8) 74 (83.1) 141 (81.5)
Weight, kg, mean (SD) 79.8 (16.7) 74.8 (13.4)* 77.2 (15.3)
Estimated creatinine clearance, mL/min, mean (SD) 80.3 (27.3) 72.9 (27.5) 76.5 (27.6)
Creatinine clearance strata, n (%)
Low (⬎30, ⱕ60 mL/min) 25 (29.8) 30 (33.7) 55 (31.8)
Normal (⬎60 mL/min) 59 (70.2) 59 (66.3) 118 (68.2)
Duration of PHN, mo, mean (SD) 34.4 (36.7) 33.3 (35.4) 33.8 (35.9)
Predominantly affected dermatome, n (%)
Trigeminal 22 (26.2) 18 (20.2) 40 (23.1)
Cervical 6 (7.1) 9 (10.1) 15 (8.7)
Thoracic 41 (48.8) 42 (47.2) 83 (48.0)
Lumbar 11 (13.1) 16 (18.0) 27 (15.6)
Sacral 4 (4.8) 4 (4.5) 8 (4.6)
Baseline mean pain score (SD) 6.4 (1.5) 6.3 (1.4) 6.4 (1.5)

* n ⫽ 88 for the pregabalin-treated group.

PHN ⫽ postherpetic neuralgia.

When the patients who were taking any of the four types of tween the treatment groups for these two measures were also
medications for which there is evidence of efficacy in the treatment of observed at each time point during the study (weeks 1, 3, 5, and 8;
neuropathic pain—tricyclic antidepressants, opioid analgesics (in- figure 4, B and C).
cluding tramadol), anticonvulsants, and topical analgesics13—were Mean sleep interference scores were improved in the pregaba-
excluded (n ⫽ 46), the benefit of treatment with pregabalin vs lin vs the placebo group at study endpoint (p ⫽ 0.0001; see table
placebo on endpoint mean pain scores remained (endpoint mean 2). In addition, as observed with the pain measures, the mean
scores 3.17 vs 5.14; p ⫽ 0.0001). sleep interference score in pregabalin-treated patients demon-
Because side effects may compromise the integrity of the strated significant improvement beginning with the first week of
double-blind in clinical trials, the primary analysis of endpoint treatment, and this remained significant compared to placebo
mean pain scores was repeated after removing all patients who throughout the study (figure 3B). There was also a significant
had one or more of the five side effects reported by more than 10% improvement on the MOS Sleep Scale sleep problem index at
of pregabalin-treated patients at any time during the study (dizzi- study endpoint (see table 2).
ness, somnolence, peripheral edema, amblyopia, and dry mouth; More favorable mean scores were found in the pregabalin
table 3). In these patients, there was still greater improvement in
group compared with the placebo group for several of the SF-36
the endpoint mean pain scores in the patients treated with pre-
quality of life domains at the end of the trial; the differences were
gabalin (3.60 vs 5.29 in placebo patients, p ⫽ 0.0001). When all
significant for the SF-36 bodily pain and general health perception
patients who reported any of the 11 side effects listed in table 3 at
any time during the study were removed, the greater improve- scales (see table 2). Pregabalin-treated patients improved more
ment in endpoint mean pain scores of the pregabalin-treated pa- than placebo-treated patients on the POMS depression-dejection
tients remained significant (3.62 vs 5.37 in placebo patients). scale (pregabalin endpoint mean score ⫽ 6.70 vs 8.47; p ⫽ 0.051);
Secondary efficacy measures. Pregabalin-treated patients the differences between the two treatment groups were not signif-
were improved relative to the placebo-treated group at study end- icant for the other POMS scales.
point with respect to the SF-MPQ sensory, affective, and total Significant differences between pregabalin and placebo were
pain scores (p ⬍ 0.005; see table 2). The SF-MPQ total score seen in both the PGIC and the CGIC. Global assessments of
demonstrated significant pain reduction for pregabalin vs placebo change showed that at the end of the trial more patients receiving
at all time points during the study (figure 4A); significant im- pregabalin reported global improvements of very much improved,
provements in the sensory and affective scores were also observed much improved, and minimally improved (84% vs 26%; figure 5),
by the end of week 1 for pregabalin, and these effects were main- and fewer pregabalin-treated patients reported global worsening
tained throughout the study. In addition, the pregabalin group of minimally worse, much worse, and very much worse (4% vs
had significantly lower SF-MPQ pain scores on the VAS and PPI 14%; see figure 5). Clinician assessments of global change closely
at study endpoint (see table 2), and significant differences be- paralleled patients’ self-assessments.
April (2 of 2) 2003 NEUROLOGY 60 1277
a maximum intensity of mild or moderate (81% in the pregabalin
group, and 92% in the placebo group).
More of the adverse events experienced by pregabalin-treated
patients were considered to be related to study medication (73% vs
37% in the placebo group). Of the most commonly reported ad-
verse events related to pregabalin, dizziness, somnolence, periph-
eral edema, dry mouth, and various CNS-related adverse events
occurred more frequently in the pregabalin-treated patients than
in the placebo group (see table 3). More pregabalin-treated pa-
tients discontinued from the study because of an adverse event
(32%, n ⫽ 28) than did placebo-treated patients (5%, n ⫽ 4), but
no pregabalin-treated patients discontinued due to lack of efficacy,
whereas six placebo-treated patients did (7%; see figure 1). The
adverse event that most commonly led to discontinuation of pre-
gabalin treatment was somnolence (11.2%, n ⫽ 10).
The median time to onset of any adverse event was 6 days for
the pregabalin-treated patients and 8 days for the placebo-treated
patients. The median time to onset for somnolence or dizziness
was 2 to 3 days for both treatment groups. In pregabalin-treated
patients who completed the study, the median durations of somno-
lence (53 days) and dizziness (33 days) were longer than in
placebo-treated patients (17 days for somnolence, 3 days for
dizziness).
Peripheral edema was reported for 19 patients: 17 who re-
ceived pregabalin and 2 who received placebo. All cases were mild
to moderate in severity, and in no case was edema related to
worsening of an underlying cardiovascular condition. Median time
to onset of edema in the pregabalin-treated patients was 24 days,
and the median duration of edema was 31 days. Two patients
treated with pregabalin discontinued treatment due to edema.
There were no clinically significant findings involving the hema-
tology, blood chemistry, or urinalysis evaluations in the patients
exposed to pregabalin. Similarly, there were no clinically signifi-
cant findings in the electrocardiogram, visual examinations, or
physical and neurologic examinations.

Discussion. The results of this study demon-


strated that pregabalin is efficacious in relieving
pain and improving sleep in patients with PHN. A
Figure 1. Patient disposition, indicated numerically at consistent pattern of results among the different out-
each level of study progress. Dashed line reflects the fact come measures demonstrated that there is a rapid
that patients who were withdrawn from the trial were gen- and durable beneficial effect of pregabalin treat-
erally not allowed to enter the open-label extension. ment. Beginning with the first full day of treatment
and continuing to the end of the trial 8 weeks later,
Pharmacokinetics. Pregabalin dosage adjustments for creati- pain was significantly improved in the patients
nine clearance used in this study produced comparable plasma treated with pregabalin compared to those treated
pregabalin concentrations in the two renal function strata of the with placebo. In addition, sleep interference scores
pregabalin treatment group. Likewise, the predicted morning as well as the five major components of the SF-
Cminss and Cavg values were comparable in the two renal function
strata of the pregabalin treatment group (table 4). Estimates of total MPQ—total, sensory, affective, VAS, and PPI
body clearance were somewhat lower and volume of distribution scores—were all significantly improved relative to
somewhat higher in this study population of older individuals placebo by the end of the first week of treatment and
than previously reported,20 as would be expected due to age-
related diminished renal function.
they remained so until the end of the trial.
Several subjects had low plasma pregabalin concentration at Many of the patients enrolled in the study were
study endpoint but the low values can be explained by the time elderly, the mean duration of their PHN was almost
since the last pregabalin dose, which may reflect either poor com- 3 years, and although 68% were taking concomitant
pliance or subjects having exhausted their drug supply before the
termination visit. At visit four, only two subjects appeared non- pain medications, they still met the enrollment crite-
compliant based on low plasma pregabalin concentrations and the ria, which required that pain was moderate or se-
corresponding post-dose time; both were compliant based on their vere in intensity.33 These features are characteristic of
study endpoint results. Thus, the two subjects who can be consid-
ered noncompliant at the first visit in which there was pharmaco-
patients with PHN, as many as half of whom are par-
kinetic sampling were deemed compliant at the second visit in tially or totally refractory to all existing treatments.34
which there was sampling. Only one subject in the placebo group Because the individuals we studied appear represen-
had a significant plasma pregabalin concentration. This occurred tative of the general population of patients with PHN,
at study endpoint and it is assumed that the patient received his
open-label extension dose before the blood sample because the our results suggest that pregabalin will be effective in
visit four plasma concentration was zero. Therefore, no subjects in treating PHN in clinical practice and that it provides
the placebo group appeared to receive pregabalin. an important therapeutic option for patients with this
Safety. A majority of patients in both treatment groups— 87% chronic neuropathic pain syndrome.
in the pregabalin group and 63% in the placebo group—reported
an adverse event during the double-blind treatment phase. Re- It was recently concluded that a 30% decrease in
gardless of treatment, most patients with adverse events reported pain, which corresponds to PGIC ratings of much im-
1278 NEUROLOGY 60 April (2 of 2) 2003
Table 2 Summary of efficacy analyses at study endpoint

Pregabalin Placebo Endpoint comparison

Least Least
squares squares
Measure No. mean SE No. mean SE Difference 95% CI p Value

Endpoint mean pain score* 88 3.60 0.24 84 5.29 0.24 ⫺1.69 (⫺2.33, ⫺1.05) 0.0001
Short-form McGill Pain Questionnaire
Sensory score 89 8.15 0.73 84 11.90 0.74 ⫺3.75 (⫺5.71, ⫺1.79) 0.0002
Affective score 89 1.75 0.27 84 2.82 0.28 ⫺1.07 (⫺1.81, ⫺0.33) 0.0047
Total score 89 9.85 0.95 84 14.72 0.96 ⫺4.87 (⫺7.41, ⫺2.34) 0.0002
Visual analogue scale 89 38.68 2.90 84 56.30 2.93 ⫺17.62 (⫺25.37, ⫺9.86) 0.0001
Present pain intensity 89 1.58 0.12 84 1.98 0.12 ⫺0.40 (⫺0.71, ⫺0.09) 0.0127
Endpoint mean sleep interference score*† 88 1.93 0.23 84 3.51 0.23 ⫺1.58 (⫺2.19, ⫺0.97) 0.0001
MOS Sleep Scale sleep problem index† 85 26.63 1.77 82 36.43 1.75 ⫺9.80 (⫺14.49, ⫺5.11) 0.0001
SF-36 Health Survey‡
Physical functioning 85 62.25 1.96 83 61.41 1.90 0.84 (⫺4.25, 5.93) 0.7449
Physical role limitations 86 47.04 4.12 83 44.43 4.09 2.60 (⫺8.27, 13.48) 0.6369
Social functioning 86 78.15 2.44 83 74.68 2.44 3.47 (⫺3.02, 9.96) 0.2920
Bodily pain 86 55.14 2.13 83 46.14 2.13 9.00 (3.33, 14.66) 0.0021
Mental health 86 77.53 1.54 83 73.73 1.54 3.81 (⫺0.28, 7.89) 0.0676
Emotional role limitations 86 73.55 3.97 83 64.62 3.97 8.93 (⫺1.60, 19.46) 0.0960
Vitality 86 49.99 1.89 83 48.94 1.88 1.05 (⫺3.96, 6.05) 0.6798
General health perception 85 67.61 1.58 81 63.40 1.58 4.21 (0.02, 8.40) 0.0488

* Endpoint mean pain and mean sleep interference measures were derived from patients’ last 7 days of diary entries while taking
study drug.
† Higher values reflect greater sleep impairment.
‡ Higher values reflect better quality of life.

MOS ⫽ Medical Outcomes Study; SF ⫽ Short Form.

proved or very much improved, should be considered portant improvement of their average daily pain. A
clinically important in evaluating the results of chronic 50% decrease in pain ratings between baseline and
pain trials.32 Using this criterion, almost two-thirds of study endpoint—a level of pain relief that corresponds
the pregabalin-treated patients reported clinically im- to the highest degree of improvement on the PGIC, a
rating of very much improved32— has also been consid-
ered evidence of clinically important improvement. In
the current study, 50% of the patients in the
pregabalin-treated group but only 20% of those in the
placebo-treated group had a 50% decrease in mean
pain score.
There were no serious adverse events related to pre-
gabalin treatment; no clinically significant electrocardio-
gram, ophthalmologic, physical, or neurologic
examination findings; and the results of laboratory evalu-
ations related to pregabalin treatment were unremark-
able. The tolerability of pregabalin treatment was
acceptable, given the protocol’s brisk titration in a mostly
elderly sample of patients to a maximum dosage of 600
mg daily of pregabalin that patients were not allowed to
Figure 2. Daily diary pain scores for the first 7 days of
adjust during the trial. Indeed, one-third of the
treatment as rated on an 11-point numerical pain rating pregabalin-treated patients who withdrew from the study
scale from 0 (no pain) to 10 (worst possible pain). Solid because of adverse events entered the open-label exten-
curve ⫽ placebo-treated patients; dashed curve ⫽ sion, in which improved tolerability would be expected
pregabalin-treated patients; PBO ⫽ placebo; PGB ⫽ because of the lower dosages permitted. Of those
pregabalin. *p ⬍ 0.01. pregabalin-treated patients with adverse events, more
April (2 of 2) 2003 NEUROLOGY 60 1279
Table 3 Most frequently occurring adverse events*

Preferred term Placebo, n ⫽ 84 Pregabalin, n ⫽ 89

Dizziness 10 (11.9) 25 (28.1)


Somnolence 6 (7.1) 22 (24.7)
Peripheral edema 2 (2.4) 17 (19.1)
Amblyopia 1 (1.2) 10 (11.2)
Dry mouth 2 (2.4) 10 (11.2)
Abnormal gait 1 (1.2) 7 (7.9)
Headache 7 (8.3) 7 (7.9)
Ataxia 0 6 (6.7)
Confusion 0 6 (6.7)
Diarrhea 4 (4.8) 6 (6.7)
Speech disorder 0 5 (5.6)

Values are n (%).

* Adverse events reported by at least 5% of patients in the pre-


gabalin treatment group. Events sorted by decreasing fre-
quency in the pregabalin group.

levels of pregabalin in both renal function strata were


achieved. These pharmacokinetic analyses as well as
the results of the strata subgroup analyses of end-
point mean pain scores provide support for combining
both strata into one pregabalin treatment arm.
One of the exclusion criteria was failure to re-
spond to previous gabapentin treatment of PHN at
dosages ⱖ1,200 mg/day. Although it could be argued
that this methodologic feature of the trial increased
Figure 3. (A) Weekly mean pain score as rated on an 11-
the likelihood of finding a beneficial effect of pregaba-
point numerical pain rating scale from 0 (no pain) to 10 lin, this assumes that therapeutic response to gabap-
(worst possible pain). (B) Weekly mean score of sleep inter- entin predicts therapeutic response to pregabalin.
ference score during the past 24 hours as rated on an 11- Unfortunately, history of prior gabapentin treatment
point scale from 0 (did not interfere) to 10 (unable to sleep was not recorded, and so it is not possible to deter-
due to pain). Solid curves ⫽ placebo-treated patients; mine how many patients had a beneficial response to
dashed curves ⫽ pregabalin-treated patients; PBO ⫽ pla- gabapentin before enrollment in the trial and
cebo; PGB ⫽ pregabalin. *p ⬍ 0.01. whether these patients were more likely to respond
to treatment with pregabalin.
It has been proposed that there are distinct sub-
than 80% reported that the maximum intensity of these types of PHN, and that these subtypes differ in
events was mild to moderate. Dizziness and somnolence patterns of symptoms and signs, underlying patho-
were the most common side effects of pregabalin treat- physiologic mechanisms, and treatment response.35,36
ment, but could be tolerated by patients even though they No attempt was made in the current study to collect
may have persisted throughout the major portion of the the extensive data that would be required to accu-
treatment period. Approximately 11% of patients receiv- rately categorize patients into these putative PHN
ing pregabalin discontinued treatment due to somno- subtypes, which include epidermal nerve fiber den-
lence, which was not unexpected given the dosage tested. sity, response to pharmacologic challenge, and quan-
Although more pregabalin-treated patients discontinued titative sensory testing. An exploratory attempt was
the study because of an adverse event compared to pa- made to assess allodynia and hyperalgesia at base-
tients administered placebo, fewer pregabalin-treated pa- line and study endpoint and to determine whether
tients discontinued the study because of unrelieved pain. resolution of these sensory abnormalities was more
Several methodologic aspects of this trial require common in the patients treated with pregabalin com-
comment. The pregabalin treatment arm was strati- pared to placebo. Resolution of allodynia at study
fied based on renal function because the population endpoint occurred in 48.8% of pregabalin-treated pa-
of PHN patients is likely to include significant num- tients and 30.6% of placebo-treated patients who had
bers of elderly individuals with diminished renal allodynia at baseline; resolution of hyperalgesia at
function. The steady-state levels of pregabalin result- study endpoint occurred in 33.9% of pregabalin-
ing from the stratification suggest that therapeutic treated patients and 29.1% of placebo-treated pa-
1280 NEUROLOGY 60 April (2 of 2) 2003
Figure 5. Patient Global Impression of Change (PGIC),
rated by patients on a seven-point scale of overall change
experienced since beginning study medication (1 ⫽ very
much improved; 2 ⫽ much improved; 3 ⫽ minimally im-
proved; 4 ⫽ no change; 5 ⫽ minimally worse; 6 ⫽ much
worse; 7 ⫽ very much worse). *p ⫽ 0.001 for the overall
pregabalin vs placebo comparison using the Cochran-
Mantel-Haenszel test.

tients who had hyperalgesia at baseline. Neither of


these group differences was significant. Interpreta-
tion of these results is problematic, however, because
there were no specific guidelines provided to investi-
gators for determining the presence or absence of
allodynia and hyperalgesia and each investigator in-
dependently decided how to assess these sensory
abnormalities.
Chronic pain syndromes such as PHN are charac-
terized by a constellation of symptoms that extends
well beyond the experience of pain and that may
include profound disturbances of sleep, mood, and
quality of life.1,2,5,37,38 The consistent improvements
observed in this study in the pain and sleep diary
measures, which began early and were maintained
throughout treatment, suggest that pregabalin re-
duces not only the severity of daytime pain in PHN
but also the sleep disturbance that accompanies
chronic pain. Pregabalin-treated patients also re-
ported significantly greater improvements than
placebo-treated patients on the MOS Sleep Scale
sleep problem index as well as on the SF-36 bodily
Figure 4. (A) Total pain descriptor score from the Short- pain and general health perception scales.
Form McGill Pain Questionnaire (SF-MPQ). The score is Despite these significant improvements in pain,
the sum of the intensity ratings for pain intensity for 15 sleep, and overall perception of health, significant
sensory and affective pain descriptors on a scale of 0 benefits of treatment on other domains of mood and
(none) to 3 (severe). (B) Visual analog scale score from the quality of life were not observed. In previous trials of
SF-MPQ. Patients placed a slash on a 100-mm line from 0 neuropathic pain, the benefits of treatment across
(no pain) to 100 (worst possible pain). (C) SF-MPQ present
specific domains of quality of life have also been
pain intensity score, reported on a scale of 0 (no pain), 1
found less consistently than the benefits of treat-
(mild pain), 2 (discomfort), 3 (distressing), 4 (horrible),
ment on pain intensity.8,11,12,25,39,40 Possible explana-
and 5 (excruciating). Solid curves ⫽ placebo-treated pa-
tients; dashed curves ⫽ pregabalin-treated patients;
tions for this are that most trials have used general
PBO ⫽ placebo; PGB ⫽ pregabalin. *p ⬍ 0.01. rather than disease-specific measures of quality of
life and that these trials may not have been of suffi-
cient duration to demonstrate widespread beneficial
effects of pain relief on the various components of
quality of life.
April (2 of 2) 2003 NEUROLOGY 60 1281
Table 4 Pharmacokinetic values for the two renal function strata of the pregabalin treatment group

Pregabalin 300 mg/day, Pregabalin 600 mg/day,


Measure CLcr ⬎30 and ⱕ60 mL/min CLcr ⬎60 mL/min

Observed plasma pregabalin concentration, ␮g/mL, range 2.44–4.8 0.244–18.6


Time postdose for observed plasma pregabalin 1.00–6.67 0.75–17.8
concentration, h, range
Predicted Cavg concentrations, ␮g/mL (range) 6.64 (4.05–14.4) 8.36 (3.45–15.7)
Predicted morning CminSS concentrations, ␮g/mL (range) 4.69 (2.97–13.1) 5.27 (1.31–11.4)
Pregabalin clearance, mL/min, range 42.7–52.2
Pregabalin volume of distribution, L, range 29.8–34.2

CLcr ⫽ creatinine clearance; Cavg ⫽ average steady-state concentration; CminSS ⫽ predicted steady-state morning trough concentra-
tion.

Pregabalin is a structural analogue of the neuro- tin, tricyclic antidepressants, and opioid
transmitter gamma-aminobutyric acid (GABA), as is anagesics.11,56-60 Likewise, although few systematic
gabapentin, but neither compound interacts with ra- comparisons of differences in side effects among the
dioligand binding at either GABA-A or GABA-B re- medications used in the treatment of PHN have been
ceptors. Pregabalin, with a Ki close to 40 nM, avidly conducted,57 the number needed to harm (NNH) for
displaces [3H]gabapentin from the ␣2-␦ subunit of pregabalin based on all side effects was 4.3, which is
voltage-gated calcium channels, which are localized comparable to what has been reported in previous
heterogeneously in a number of regions of rat studies of gabapentin, tricyclic antidepressants, and
brain.41-43 With both gabapentin and pregabalin, this opioid analgesics.10,57,59 However, it is important to note
binding activity correlates with decreased calcium that except for the studies of gabapentin, previous ran-
channel function44,45 and decreased release of several domized controlled trials in PHN have been relatively
neurotransmitters, including glutamate, noradrena- small studies that have typically used a two-period
line, and substance P.46-48 This decreased neurotrans- crossover research design. Comparison of NNT and
mitter release is associated with reduced NNH among these different treatments is therefore
hyperexcitability in several neuronal models49 and problematic, especially given the use of intent-to-treat
may provide the mechanism of action of the analge- analyses in the gabapentin trials and in the current
sic effects of these two medications. study but not in the crossover trials of tricyclic antide-
In humans, the rate of gabapentin absorption is pressants, opioid analgesics, and lidocaine patch 5%.
relatively slow, with peak plasma concentrations oc- The results of the current study have demon-
curring around 3 hours postdose, and the extent of strated that pregabalin is safe and efficacious in the
absorption is saturable.50,51 These observations are treatment of PHN. The use of oral medications in
consistent with gabapentin being a substrate for Sys- PHN and other neuropathic pain syndromes has long
tem L transporter; this transporter is most likely the been guided by the following rule of thumb: start low
predominant pathway for the absorption of gabapen- and go slow. The results therefore also suggest that
tin.52 For pregabalin, the rate of absorption is rapid, the advantage of pregabalin in comparison to exist-
with peak plasma concentrations occurring within 1 ing treatments is that it provides clinically impor-
hour postdose, and the extent of absorption is pro- tant, rapid, and durable pain relief without the
portional to dose.53,54 Pregabalin is also a substrate necessity of a slow and lengthy titration to an effec-
for the System L transporter. Studies using in situ tive dosage.
intestine perfusion or brush-border membrane vesi-
cles suggest that multiple amino acid transport sys- Appendix
tems may be involved in the small intestinal Participating investigators: Charles Bernick, MD, Las Vegas, NV; E. Rich-
absorption of pregabalin.55 It is most likely the mul- ard Blonsky, MD, Chicago, IL; Paul K. Brownstone, MD, Boulder, CO;
Michael Z. Chesser, MD, Little Rock, AR; Steven Cohen, MD, St. Peters-
tiple amino acid transport systems that provide pre- burg, FL; Ghassan Kanazi, MD, Robert Dworkin, PhD, Rochester, NY;
gabalin with dose-proportional absorption and linear Bradley S. Galer, MD, New York, NY; Daniel T. Garber, MD, Asheville, NC;
pharmacokinetics. Gary Gerard, MD, Toledo, OH; Joseph S. Gimbel, MD, Phoenix, AZ; James
Storey, MD, Albany, NY; Sassan Hassassian, MD, Baltimore, MD; Everett
Gabapentin has demonstrated efficacy in the treat- Heinze, MD, Austin, TX; Ronica Kluge, MD, Fort Myers, FL; Montie Rom-
ment of PHN,8,9 as have tricyclic antidepressants,6,7 melman, MD, Paducah, KY; W. Alvin McElveen, MD, Bradenton, FL; Anne
L. Oaklander, MD, PhD, Boston, MA; Michael C. Rowbotham, MD, San
opioid analgesics,10,11 and lidocaine patch 5%.12 To com- Francisco, CA; Jay J. Rubin, MD, Ocala, FL; Richard Singer, MD, Pem-
pare the efficacy of different medications used in treat- broke Pines, FL; Brett R. Stacey, MD, David Sibell, MD, Portland, OR;
Michael M. Tuchman, MD, Palm Beach Gardens, FL; Jeanette Wendt, MD,
ing PHN, number-needed-to-treat (NNT) analyses Tucson, AZ; Naynesh R. Patel, MD, Kettering, OH; Richard Pellegrino, MD,
have been conducted.56-60 The NNT for pregabalin- PhD, Hot Springs, AR; Ruth Fredericks, MD, Jackson, MS.
associated decreases in endpoint mean pain scores
ⱖ30% was 2.7 and for decreases in endpoint mean pain References
scores ⱖ50% it was 3.4. These NNT are comparable to 1. Cluff RS, Rowbotham MC. Pain caused by herpes zoster infection. Neu-
what has been reported in previous studies of gabapen- rol Clin 1998;16:813– 832.

1282 NEUROLOGY 60 April (2 of 2) 2003


2. Dworkin RH, Portenoy RK. Pain and its persistence in herpes zoster. 32. Farrar JT, Young JP, LaMoreaux L, Werth JL, Poole M. Clinical impor-
Pain 1996;67:241–251. tance of changes in chronic pain intensity on an 11-point numerical
3. Arani RB, Soong S-J, Weiss HL, et al. Phase specific analysis of herpes pain rating scale. Pain 2001;94:149 –158.
zoster associated pain data: a new statistical approach. Stat Med 2001; 33. Serlin RC, Mendoza TR, Nakamura Y, Edwards KR, Cleeland CS.
20:2429 –2439. When is cancer pain mild, moderate, or severe? Grading pain severity
4. Desmond R, Weiss HL, Arani RB, et al. Clinical applications for change- by its interference with function. Pain 1995;61:277–284.
point analysis of herpes zoster pain. J Pain Symptom Manage 2002;23: 34. Watson CPN. Postherpetic neuralgia: the importance of preventing this
510 –516. intractable end-stage disorder. J Infect Dis 1998;178(suppl 1):S91–S94.
5. Dworkin RH, Schmader KE. The epidemiology and natural history of 35. Fields HL, Rowbotham M, Baron R. Postherpetic neuralgia: irritable
herpes zoster and postherpetic neuralgia. In: Watson CPN, Gershon nociceptors and deafferentation. Neurobiol Dis 1998;5:209 –227.
AA, eds. Herpes zoster and postherpetic neuralgia, 2nd ed. New York: 36. Rowbotham MC, Petersen KL, Fields HL. Is postherpetic neuralgia
Elsevier Press, 2001;39 – 64. more than one disorder? Pain Forum 1998;7:231–237.
6. Max MB. Treatment of post-herpetic neuralgia: antidepressants. Ann 37. Haythornthwaite JA, Benrud-Larson LM. Psychological aspects of neu-
Neurol 1994;35(suppl):S50 –S53. ropathic pain. Clin J Pain 2000;16:S101–S105.
7. Watson CPN, Vernich L, Chipman M, Reed K. Nortriptyline versus 38. Smith MT, Perlis ML, Smith MS, Giles DE, Carmody TP. Sleep quality
amitriptyline in postherpetic neuralgia: a randomized trial. Neurology and presleep arousal in chronic pain. J Behav Med 2000;23:1–13.
1998;51:1166 –1171. 39. Eisenberg E, Luria Y, Braker C, Daoud D, Ishay A. Lamotrigine re-
8. Rowbotham M, Harden N, Stacey B, Bernstein P, Magnus-Miller L. duces painful diabetic neuropathy: a randomized, controlled study.
Gabapentin for the treatment of postherpetic neuralgia: a randomized Neurology 2001;57:505–509.
controlled trial. JAMA 1998;280:1837–1842.
40. Finnerup NB, Sindrup SH, Bach FW, Johannesen IL, Jensen TS. Lam-
9. Rice ASC, Maton S, Postherpetic Neuralgia Study Group. Gabapentin
otrigine in spinal cord injury pain: a randomized controlled trial. Pain
in postherpetic neuralgia: a randomised, double blind, placebo con-
2002;96:375–383.
trolled study. Pain 2001;94:215–224.
41. Taylor CP, Vartanian MG, Yuen PW, Bigge C, Suman-Chauhan N, Hill
10. Watson CPN, Babul N. Efficacy of oxycodone in neuropathic pain: a ran-
DR. Potent and stereospecific anticonvulsant activity of 3-isobutyl
domized trial in postherpetic neuralgia. Neurology 1998;50:1837–1841.
11. Raja SN, Haythornthwaite JA, Pappagallo M, et al. Opioids versus GABA relates to in vitro binding at a novel site labeled by tritiated
antidepressants in postherpetic neuralgia: a randomized, placebo- gabapentin. Epilepsy Res 1993;14:11–15.
controlled trial. Neurology 2002;59:1015–1021. 42. Gee NS, Brown JP, Dissanayake VU, Offord J, Thurlow R, Woodruff GN. The
12. Galer BS, Rowbotham MC, Perander J, Friedman E. Topical lidocaine novel anticonvulsant drug, gabapentin (Neurontin), binds to the alpha2delta
patch relieves postherpetic neuralgia more effectively than a vehicle topi- subunit of a calcium channel. J Biol Chem 1996;271:5768–5776.
cal patch: results of an enriched enrollment study. Pain 1999;80:533–538. 43. Hill DR, Suman-Chauhan N, Woodruff GN. Localization of (3-
13. Kanazi GE, Johnson RW, Dworkin RH. Treatment of postherpetic neu- H)gabapentin to a novel site in rat brain: autoradiographic studies. Eur
ralgia: an update. Drugs 2000;59:1113–1126. J Pharmacol Mol Pharmacol 1993;244:303–309.
14. Field MJ, Oles RJ, Lewis AS, McCleary S, Hughes J, Singh L. Gabap- 44. Fink K, Dooley DJ, Meder WP, et al. Inhibition of neuronal Ca2⫹ influx
entin (Neurontin) and S-(⫹)-3-isobutyl GABA represent a novel class of by gabapentin and pregabalin in the human neocortex. Neuropharma-
selective antihyperalgesic agents. Br J Pharmacol 1997;121:1513–1522. cology 2002;42:229 –236.
15. Field MJ, Holloman EF, McCleary S, Hughes J, Singh L. Evaluation of 45. Martin DJ, McClelland D, Herd MB, et al. Gabapentin-mediated inhi-
gabapentin and S-(⫹)-3-isobutylGABA in a rat model of postoperative bition of voltage-activated Ca-2⫹ channel currents in cultured sensory
pain. J Pharmacol Exp Ther 1997;282:1242–1246. neurones is dependent on culture conditions and channel subunit ex-
16. Bockbrader HN, Burger PJ, Kugler AR, Knapp LE, Garofalo EA, La- pression. Neuropharmacology 2002;42:353–366.
londe RL. Population pharmacokinetics (PK) analyses of commonly pre- 46. Dooley DJ, Mieske CA, Borosky SA. Inhibition of K⫹-evoked glutamate
scribed antiepileptic drugs (AEDs) coadministered with pregabalin release from rat neocortical and hippocampal slices by gabapentin.
(PGB) in adult patients with refractory partial seizures. Epilepsia 2001; Neurosci Lett 2000;280:107–110.
42(suppl 7):84. 47. Maneuf YP, Hughes J, McKnight AT. Gabapentin inhibits the sub-
17. Bockbrader HN, Strand JC, Wesche DL, et al. Drug interaction studies stance P-facilitated K⫹-evoked release of [3H]-glutamate from rat cau-
of pregabalin (CI-1008, PGB) in patients with epilepsy maintained on dal trigeminal nucleus slices. Pain 2001;93:191–196.
either valproate (VPA), lamotrigine (LTG), phenytoin (PHT), or car- 48. Dooley DJ, Donovan CM, Pugsley TA. Stimulus-dependent modulation
bamazepine (CBZ). Epilepsia 2001;42(suppl 7):86. of [3H]norepinephrine release from rat neocortical slices by gabapentin
18. Richter RW, Dworkin RH, Sharma U, et al. Pregabalin is effective in and pregabalin. J Pharmacol Exp Ther 2002;296:1086 –1098.
patients with painful diabetic neuropathy as evaluated in three clinical 49. Kaneko M, Mestre C, Sanchez EH, Hammond DL. Intrathecally admin-
trials. Presented at the 10th World Congress on Pain; August 17–22, istered gabapentin inhibits formalin-evoked nociception and the expres-
2002; San Diego, CA. sion of Fos-like immunoreactivity in the spinal cord of the rat.
19. Melzack R. The Short-Form McGill Pain Questionnaire. Pain 1987;30: J Pharmacol Exp Ther 2000;292:743–751.
191–197. 50. Bockbrader HN. Clinical pharmacokinetics of gabapentin. Drugs Today
20. Bockbrader HN, Burger P, Corrigan BW. Population pharmacokinetics 1995;31:613– 619.
of pregabalin in healthy volunteers, renally impaired patients, and 51. Bockbrader HN, Breslin EM, Underwood BA, Posvar EL, Sedman AJ.
patients with chronic pain. Presented at the meeting of the American Multiple-dose, dose-proportionality study of Neurontin® (gabapentin)
Pain Society; March 14 –17, 2002; Baltimore, MD. in healthy volunteers. Epilepsia 1996;37(suppl 5):159.
21. Hayes RD, Stewart AL. Sleep measures. In: Stewart AL, Ware JE Jr, 52. Stewart BH, Kugler AR, Thompson PR, Bockbrader HN. A saturable
eds. Measuring functioning and well-being. Durham, NC: Duke Univer- transport mechanism in the intestinal absorption of gabapentin is the
sity Press, 1992;235–259. underlying cause of the lack of proportionality between increasing dose
22. Ware JE Jr, Snow KK, Kosinski M, Gandek B. SF-36-item Short Form
and drug levels in plasma. Pharm Res 1993;10:276 –281.
Health Survey (SF-36). I. Conceptual framework and item selection.
53. Busch JA, Strand JA, Posvar EL, Bockbrader HN, Radulovic LL. Pre-
Med Care 1992;30:473– 483.
gabalin (CI-1008) multiple-dose pharmacokinetics and safety/tolerance
23. McNair DM, Lorr M, Droppleman LF. Profile of Mood States: manual.
in healthy volunteers. Pharm Sci 1999;1(suppl 4):2033.
San Diego: Educational and Industrial Testing Service, 1981.
54. Alvey CW, Bockbrader HN, Busch JA, et al. Clinical pharmacokinetics
24. Guy W. ECDEU assessment manual for psychopharmacology, revised.
Rockville, MD: US Department of Health, Education, and Welfare, 1976. of pregabalin in healthy volunteers. Presented at the 10th World Con-
25. Backonja M, Beydoun A, Edwards KR, et al. Gabapentin for the symp- gress on Pain; August 17–22, 2002; San Diego, CA.
tomatic treatment of painful neuropathy in patients with diabetes mel- 55. Piyapolrungroj N, Li C, Bockbrader H, Liu G, Fleisher D. Mucosal
litus: a randomized controlled trial. JAMA 1998;280:1831–1836. uptake of gabapentin (Neurontin) vs pregabalin in the small intestine.
26. SAS Institute Inc. SAS/STAT user’s guide, version 6, fourth edition, Pharm Res 2001;18:1126 –1130.
volumes 1 and 2. Cary, NC: SAS Institute, 1989. 56. Sindrup SH, Jensen TS. Efficacy of pharmacological treatments of neu-
27. Steel R, Torrie J. Principles and procedures of statistics. New York: ropathic pain: an update and effect related to mechanism of drug ac-
McGraw-Hill Book Company, 1960;308 –310. tion. Pain 1999;83:389 – 400.
28. Berry D, ed. Statistical methodology in the pharmaceutical sciences. 57. Collins SL, Moore RA, McQuay HJ, Wiffen P. Antidepressants and anti-
New York: Marcel Dekker, 1990;464:403– 408. convulsants for diabetic neuropathy and postherpetic neuralgia: a quanti-
29. Windsor BL, Radulovic LL. Measurement of a new anticonvulsant, tative systematic review. J Pain Symptom Manage 2000;20:449 – 458.
(S)-3-(aminomethyl1)-5-methylhexanoic acid, in plasma and milk by 58. Jensen TS. Anticonvulsants in neuropathic pain: rationale and clinical
high-performance liquid chromatography. J Chromatogr (Biomed Appl) evidence. Eur J Pain 2002;6(suppl A):61– 68.
1995;674:143–148. 59. McQuay HJ. Neuropathic pain: evidence matters. Eur J Pain 2002;
30. Beal SL, Sheiner LB, eds. NONMEM user’s guides, part I–VII. San 6(suppl A):11–18.
Francisco: University of California San Francisco NONMEM Project 60. Rice ASC, Maton S. Response to comment on Rice ASC, Maton S, the
Group, 1988 –1992. Postherpetic Neuralgia Study Group (UK), Gabapentin in postherpetic
31. Pharsight. WinNonlin version 3.0 user’s guide. Mountain View, CA: neuralgia: a randomized, double-blind, placebo-controlled study, Pain
Pharsight Corporation, 1998 –1999. 2001;94:215–224 (letter to the editor). Pain 2002;96:411– 412.

April (2 of 2) 2003 NEUROLOGY 60 1283


Pregabalin for the treatment of postherpetic neuralgia : A randomized,
placebo-controlled trial
R.H. Dworkin, A.E. Corbin, J.P. Young, Jr., et al.
Neurology 2003;60;1274
DOI 10.1212/01.WNL.0000055433.55136.55
This information is current as of December 16, 2012

Updated Information & including high resolution figures, can be found at:
Services http://www.neurology.org/content/60/8/1274.full.html
References This article cites 44 articles, 7 of which can be accessed free at:
http://www.neurology.org/content/60/8/1274.full.html#ref-list-1

Citations This article has been cited by 35 HighWire-hosted articles:


http://www.neurology.org/content/60/8/1274.full.html#related-u
rls
Subspecialty Collections This article, along with others on similar topics, appears in the
following collection(s):
All Clinical trials
http://www.neurology.org/cgi/collection/all_clinical_trials
All Pain
http://www.neurology.org/cgi/collection/all_pain
Antiepileptic drugs
http://www.neurology.org/cgi/collection/antiepileptic_drugs
Clinical trials Randomized controlled (CONSORT
agreement)
http://www.neurology.org/cgi/collection/clinical_trials_randomi
zed_controlled_consort_agreement
Neuropathic pain
http://www.neurology.org/cgi/collection/neuropathic_pain
Permissions & Licensing Information about reproducing this article in parts (figures,
tables) or in its entirety can be found online at:
http://www.neurology.org/misc/about.xhtml#permissions
Reprints Information about ordering reprints can be found online:
http://www.neurology.org/misc/addir.xhtml#reprintsus

You might also like