throbber
Downloaded from
`
`http://painmedicine.oxfordjournals.org/
`
` by guest on November 13, 2016
`
`PA I N M E D I C I N E
`Volume 5 • Number S1 • 2004
`
`Pharmacologic Management Part 1: Better-Studied Neuropathic
`Pain Diseases
`
`Misha-Miroslav Backonja, MD,* Jordi Serra, MD†
`*University of Wisconsin Hospital and Clinics, Madison, Wisconsin; †Neuropathic Pain Unit, Hospital General de
`Catalunya, Barcelona, Spain
`
`A B S T R A C T
`
`Neuropathic pain impacts millions of people in the United States and around the world. Patients
`experience one of many symptoms, such as pain, paresthesia, dysesthesia, hyperalgesia, and allo-
`dynia, for many years because of unavailable or inadequate treatment. One of the major challenges
`in treating patients with neuropathic pain syndromes is a lack of consensus concerning the appro-
`priate first-line treatment options for conditions associated with neuropathic pain, including post-
`herpetic neuralgia, diabetic peripheral neuropathy, and trigeminal neuralgia.
`This review summarizes the published results of randomized trials involving treatment for neu-
`ropathic pain conditions. Anticonvulsants, such as gabapentin, carbamazepine, and lamotrigine, and
`tricyclic antidepressants, including amitriptyline and desipramine, have demonstrated efficacy in
`relieving pain associated with postherpetic neuralgia, diabetic peripheral neuropathy, and trigemi-
`nal neuralgia, in several studies. However, the lack of head-to-head comparison studies of these
`agents limits the conclusions that can be reached. Clinicians who must make decisions regarding
`the care of individual patients may find some guidance from the number of randomized trials with
`a positive outcome for each agent. Using quality-of-life study outcomes, treatment strategies must
`encompass the impact of therapeutic agents on the comorbid conditions of sleep disturbance and
`mood and anxiety disorders associated with neuropathic pain.
`Looking to the future, emerging therapies, such as pregabalin and newer N-methyl-D-aspartate–
`receptor blockers, may provide physicians and patients with new treatment options for more effec-
`tive relief of pain.
`
`Key Words. Neuropathic Pain; Diabetic Peripheral Neuropathy (DPN); Postherpetic Neuralgia
`(PHN); Trigeminal Neuralgia (TGN); Anticonvulsants; Tricyclic Antidepressants.
`
`Introduction
`
`It is estimated that over 4 million people in the
`
`United States suffer from neuropathic pain [1],
`which is defined as “pain initiated or caused by
`a primary lesion or dysfunction in the nervous
`system” by the International Association for the
`Study of Pain [2]. A more specific definition calls
`for pain that is the result of injury to the nervous
`system, peripheral, central, or both, and manifests
`with positive and negative sensory phenomena [3].
`Lesions that originate in the peripheral or central
`
`Reprint requests to: Dr. Jordi Serra, Neuropathic Pain Unit,
`Hospital General de Catalunya, c. Gomera s/n, 08190 Sant
`Cugat del Vallès, Barcelona, Spain. Tel: +34-93-565-6000;
`Fax: +34-93-589-2618; E-mail: jserrac@meditex.es.
`© American Academy of Pain Medicine 1526-2375/04/$15.00/S28 S28–S47
`
`nervous system may manifest as different neuro-
`pathic pain syndromes, depending on the anatomic
`location and type of impairment. Noncancer neu-
`ropathic pain syndromes are listed in Table 1.
`Patients with neuropathic pain experience a
`combination of positive and negative sensory,
`motor, and autonomic signs and symptoms. Posi-
`tive sensory symptoms include pain, paresthesia
`(abnormal sensation, either evoked or sponta-
`neous), dysesthesia
`(evoked or
`spontaneous
`unpleasant, abnormal sensation), hyperalgesia
`(increased response to a normally painful stimu-
`lus), and allodynia (painful response to a non-
`noxious stimulus). Negative sensory symptoms
`involve a loss of sensitivity to stimulation in
`general and painful stimuli in particular (hypoes-
`
`ARGENTUM Exhibit 1145
` Argentum Pharmaceuticals LLC v. Research Corporation Technologies, Inc.
`IPR2016-00204
`
`Page 00001
`
`

`
`Downloaded from
`
`http://painmedicine.oxfordjournals.org/
`
` by guest on November 13, 2016
`
`Pharmacologic Management Part 1
`
`Table 1 Noncancer neuropathic pain syndromes
`
`Peripheral
`
`Central
`
`Complex regional pain syndrome (type I and II)
`Posttraumatic nerve injury
`Radiculopathy
`HIV sensory neuropathy
`Diabetic peripheral neuropathy
`Phantom limb pain
`Postherpetic neuralgia
`Trigeminal neuralgia
`Central poststroke pain
`Multiple sclerosis pain
`Spinal cord injury pain
`
`Adapted with permission from Dworkin [90].
`
`thesia and hypoalgesia, respectively) [4]. Often,
`these symptoms of neuropathic pain are chronic
`and endure for many years with either no treat-
`ment or inadequate treatment [5]. A survey con-
`ducted by the American Pain Society in 1998
`found that most people with chronic pain had been
`experiencing pain for over 5 years, that approxi-
`mately one third of chronic pain sufferers rated
`their pain as “the worst pain one can possibly
`imagine,” and that many chronic pain sufferers
`had to visit more than one doctor in an effort to
`gain relief from their pain [6].
`Despite the large number of people who are
`affected by neuropathic pain and the degree of suf-
`fering they endure, there does not appear to be
`consensus regarding the best way to treat the more
`commonly encountered neuropathic pain condi-
`tions [7]. No one therapeutic drug class or agent
`has been proven to be effective for all patients with
`neuropathic pain from a given etiology. Identifi-
`cation of an effective pharmacologic regimen for
`a specific patient is further complicated by the fact
`that a particular pain symptom may be produced
`by different mechanisms and that one underlying
`mechanism may manifest as several different
`symptoms [8,9]. Theoretically, the ability to iden-
`tify the mechanism(s) underlying a patient’s pain
`would enable the clinician to target pharmacologic
`treatment based on a drug’s mechanism of action
`[10]. Currently, patients with neuropathic pain
`are often treated with agents such as nonsteroidal
`anti-inflammatory drugs, which do not have
`proven efficacy in relieving neuropathic pain, or
`are treated with inappropriately low doses of
`agents that have demonstrated efficacy.
`In addition, as Nicholson and Verma discuss
`elsewhere in this issue, an understanding of the
`impact of comorbid conditions on pain and the
`effect of pain treatment on comorbidities is a
`key component in the successful management of
`patients with neuropathic pain [11].
`
`S29
`
`In this first part of our review of the pharma-
`cologic management of neuropathic pain, we
`discuss three of the better-studied neuropathic
`pain conditions—postherpetic neuralgia (PHN),
`diabetic peripheral neuropathy
`(DPN), and
`trigeminal neuralgia (TGN)—by examining pub-
`lished reports of clinical trials to draw conclusions
`from the available data.
`
`Methods
`This review is not intended to be a complete, sys-
`tematic analysis of all available data concerning the
`treatment of these three neuropathic pain condi-
`tions. Rather, it provides a summary of published
`data from well-designed, randomized trials. We
`discuss several selected studies within the context
`of each specific neuropathic pain syndrome, high-
`lighting those that have a quantitatively measured
`effect on the treatment of specific neuropathic
`pain symptoms or quality-of-life parameters. We
`have excluded open-label studies, case studies,
`unpublished data, and study results reported only
`in abstracts or poster presentations.
`
`Postherpetic Neuralgia
`Disease Overview
`The varicella zoster virus that causes chicken pox
`can remain latent in sensory ganglia for many years
`following the original infection [12]. Each year in
`the United States, reactivation of this virus mani-
`fests as herpes zoster (i.e., shingles) in an estimated
`800,000 people [13]. During the acute phase of
`herpes zoster, a painful rash usually forms along a
`single dermatome related to the affected dorsal
`root or cranial nerve ganglion [12]. The rash and
`severe pain associated with herpes zoster usually
`lasts less than 4 weeks [14]. However, a common
`sequela of herpes zoster is PHN, a condition in
`which pain along the involved nerve territory per-
`sists for a prolonged period after the acute rash
`resolves. Evidence suggests that the pathogenesis
`of PHN involves both peripheral and central mech-
`anisms that change over time, such as irritable pe-
`ripheral nociceptors and central sensitization [9,15].
`People 50 years of age and older are most likely
`to develop PHN following herpes zoster, and this
`painful condition can severely impact all aspects
`of life, including mood, sleep, physical activity,
`appetite, social activity, and the performance of
`necessary functions of daily living [13]. Therefore,
`the identification of an appropriate treatment to
`optimize outcome is essential.
`
`Page 00002
`
`

`
`Downloaded from
`
`http://painmedicine.oxfordjournals.org/
`
` by guest on November 13, 2016
`
`S30
`
`Treatment Options
`Published data from randomized trials assessing
`the effect of various pharmacotherapeutic agents
`on pain in patients with PHN are summarized in
`Table 2 and discussed briefly below. The most
`commonly studied therapeutic classes in PHN are
`anticonvulsants and antidepressants; however,
`many other systemic and topical agents, including
`vincristine and magnesium sulfate, have also been
`investigated.
`Anticonvulsants
`The efficacy of anticonvulsants in relieving the
`pain associated with PHN has been demonstrated
`in several randomized, placebo-controlled trials.
`For example, Rice et al. [16] and Rowbotham
`et al. [17] demonstrated that gabapentin, at doses
`of 1,800mg/day, 2,400mg/day, and 3,600mg/day,
`was significantly better than placebo at reducing
`pain in patients with PHN (P < 0.01 for 1,800-mg
`and 2,400-mg doses [16]; P < 0.001 for 3,600-mg
`dose) [17]. In both of those studies, improvements
`in quality-of-life parameters were significantly
`greater in gabapentin-treated patients compared
`with patients who received placebo, including
`vitality (P < 0.05), mental health (P < 0.05) [16],
`sleep interference (P < 0.001) [17], mood, depres-
`sion, anger, fatigue (P < 0.001 for each), and con-
`fusion (P = 0.01) [17].
`The mechanisms by which gabapentin allevi-
`ates neuropathic pain have not been fully eluci-
`dated. Gabapentin binds to the a2d subunit of
`neurotransmitter-gated calcium ion channels [18];
`however, ongoing research suggests other mecha-
`nisms may be involved [19].
`Tricyclic Antidepressants
`Tricyclic antidepressants (TCAs) are prescribed
`frequently for the treatment of PHN. In 1982,
`Watson et al. demonstrated the efficacy of amitrip-
`tyline, versus placebo, in significantly (P £ 0.0001)
`reducing pain in patients with PHN, specifically
`paroxysmal, lancinating pain [20]. Subsequent
`studies found that amitriptyline was more effective
`in relieving PHN-related pain than lorazepam [21],
`fluphenazine [22], and maprotiline [23]. However,
`nortriptyline was as effective as amitriptyline in
`ameliorating neuropathic pain in patients with
`PHN [24], and other TCAs, such as desipramine,
`also have demonstrated efficacy [25]. Indeed, Raja
`et al. found no significant difference between pain
`relief with nortriptyline or desipramine and that
`seen with opioids in patients with PHN [26].
`Inhibiting the reuptake of norepinephrine and
`serotonin is how TCAs exert their effect. In gen-
`
`Backonja and Serra
`
`eral, the most common adverse events observed
`during treatment with these agents include seda-
`tion, anticholinergic effects (i.e., dry mouth, con-
`stipation), and hypotension. However, amitriptyline,
`clomipramine, and imipramine, which inhibit
`both norepinephrine and serotonin reuptake, have
`worse side-effect profiles than agents such as de-
`sipramine and nortriptyline that act on serotonin
`only [27].
`Opioids
`Controversy exists concerning the use of opioids to
`treat neuropathic pain. Although some pain spe-
`cialists believe that opioids are either ineffective or
`effective only at doses that cause intolerable side
`effects, others feel that it is possible to achieve pain
`relief while maintaining an acceptable side-effect
`profile [28]. Significant decreases in pain scores
`have been noted in patients with PHN during
`treatment with opioids [26,29]. Oxycodone in
`extended-release form was significantly more
`effective than placebo at relieving allodynia (P =
`0.0004) and paroxysmal pain (P = 0.0001) in
`patients with PHN [29]. However, oxycodone had
`no effect on mood and depression, and adverse
`events included constipation, nausea, and sedation.
`Extended-release morphine was associated with
`less cognitive side effects than nortriptyline, and
`it was preferred by patients over nortriptyline [26].
`Tramadol, a centrally acting analgesic, although
`chemically different from the opioids, has been
`shown to be effective in relieving the pain associ-
`ated with PHN. Tramadol was also shown to be
`more effective than placebo in decreasing visual
`analog scale (VAS) pain scores (P < 0.05), and
`patients in the tramadol group in that study
`required less rescue medication (P < 0.05) than
`those given placebo [30].
`Topical Agents
`The 5% lidocaine patch, a local anesthetic, has
`been shown to reduce the pain and allodynia asso-
`ciated with PHN in several randomized studies
`[31–33], possibly by reducing ectopic activity in
`the involved sensory nerves, and by providing a
`physical barrier to mechanical stimulation from
`contact with clothing, etc. [31,32]. In one study,
`involving 96 patients with PHN, the lidocaine
`patch was significantly more effective in relieving
`neuropathic pain than a vehicle patch (P = 0.043),
`and significant benefits were experienced by
`patients with nonallodynic pain (P = 0.022) and
`patients with “sharp,” “hot,” “dull,” and “deep”
`pain (P = 0.013) [33]. In a similar study, the only
`side effect reported was a mild redness at the
`
`Page 00003
`
`

`
`Pharmacologic Management Part 1
`
`Table 2 Pharmacotherapy of PHN
`
`S31
`
`Study
`
`Patients
`
`Treatment
`
`Outcomes
`
`Adverse events/withdrawals
`
`Randomized, double-blind, placebo-controlled trials
`Tramadol £400 mg/d (N = 63)
`Boureau et al.,
`Pts with PHN
`(N = 125)
`(£300 mg/d if 75 y or older)
`2003 [30]
`or placebo (N = 62) ¥6 wks
`
`Dowd et al.,
`1999 [91]
`
`Dworkin et al.,
`2003 [37]
`
`Pts with PHN Vincristine 0.01% (N = 11) or
`(N = 20)
`placebo (saline, N = 9)
`administered by
`iontophoresis over 1 h daily
`¥20 days
`
`Pts with PHN Pregabalin, 600 mg/d (N = 59)
`(N = 173)
`if creatinine clearance
`>60 mL/min or 300 mg/d
`(N = 30) if creatinine
`clearance 30–60 mL/min, or
`placebo (N = 84) ¥8 wks
`
`Mean pain intensity was
`significantly lower in the
`tramadol group than in the
`placebo group (P < 0.05) and
`required less rescue medication
`(P < 0.05)
`Pain scores were significantly
`lower in both groups on Day 20
`vs baseline. Pain relief was
`moderate or greater in 40% of
`vincristine-treated patients and
`55% of placebo-treated pts
`Pregabalin vs placebo
`demonstrated significantly
`greater decreases in pain (end
`point mean pain scores 3.60 vs
`5.29, P = 0.0001) and sleep
`interference (P = 0.0001)
`
`Downloaded from
`
`http://painmedicine.oxfordjournals.org/
`
` by guest on November 13, 2016
`
`No difference in rates of AEs,
`tramadol (29.7%) vs placebo
`(31.8%) or total AEs,
`tramadol (31) vs placebo
`(28)
`
`No neurological deficits or
`changes in blood profiles
`were detected in either
`group
`
`Withdrawals due to AEs:
`pregabalin 32% vs placebo
`5%; AEs, pregabalin vs
`placebo: dizziness, 28% vs
`12%; somnolence, 25% vs
`7%; peripheral edema, 19%
`vs 2%; amblyopia, 11% vs
`1%; dry mouth, 11% vs 2%;
`abnormal gait, 8% vs 1%;
`headache, 8% vs 8%; ataxia,
`7% vs 0%; confusion, 7% vs
`0%; diarrhea, 7% vs 5%;
`speech disorder, 6% vs 0%
`NA
`
`G2 (amitriptyline and
`fluphenazine) had the
`highest incidence of
`sleepiness and G1
`(amitriptyline) had the highest
`incidence of dry mouth
`AEs: dizziness and
`somnolence, particularly
`during titration phase
`
`Withdrawals: 13.3% with
`gabapentin; 9.5% with
`placebo; AEs: somnolence,
`dizziness, ataxia, peripheral
`edema, and infections were
`more frequent with
`gabapentin than placebo
`Withdrawals: gabapentin,
`32 pts; placebo, 41 pts; AEs,
`gabapentin vs placebo:
`dizziness, 24% vs 8%;
`somnolence, 14% vs 5%;
`infection, 9% vs 13%;
`headache, 9% vs 14%;
`nausea, 9% vs 9%; flu
`syndrome, 7% vs 5%;
`abdominal pain, 7% vs 4%;
`accidental injury, 6% vs
`5%; diarrhea, 5% vs 4%
`AEs: burning and stinging at
`application site in 60% of
`pts using capsaicin and
`33% using placebo
`
`Galer et al.,
`2002 [33]
`
`Pts with PHN
`(N = 96)
`
`5% lidocaine patch vs vehicle
`patch ¥3 wks
`
`Pts with PHN Pts randomly assigned to 1 of
`(N = 49)
`4 groups: G1, amitriptyline;
`G2, amitriptyline and
`fluphenazine; G3,
`fluphenazine; G4, active
`placebo, ¥8 wks
`Pts with PHN Gabapentin, 1,800 mg/d
`(N = 334)
`(N = 115), gabapentin,
`2,400 mg/d (N = 108), or
`placebo (N = 111) ¥7 wks
`(with dose titration during
`first 2 wks)
`
`Pts with PHN Gabapentin maximum dose:
`(N = 229)
`3,600 mg/d (range: 1,200–
`3,600 mg/d, N = 113) or
`placebo (N = 116) ¥8 wks;
`dose titration during first
`4 wks
`
`Graff-Radford
`et al., 2000
`[22]
`
`Rice et al.,
`2001 [16]
`
`Rowbotham
`et al., 1998
`[17]
`
`Serpell et al.,
`2002 [39]
`
`Lidocaine patch improved pain
`qualities as measured by NPS
`to a greater extent than vehicle
`patch (P = 0.043)
`Statistically significant decrease
`in pain (measured by VAS)
`compared with baseline
`occurred in G1 and G2
`(P < 0.001 and P = 0.04,
`respectively)
`Differences in pain scores vs
`baseline were -34.5% (1,800-mg
`dose), -34.4% (2,400-mg dose),
`and -15.7% (placebo). Both
`gabapentin doses were
`significantly better than placebo
`(P < 0.01 for each dose)
`Reduction in pain scores
`significantly greater with
`gabapentin vs placebo (from
`6.3 to 4.2 points vs 6.5 to 6.0
`points, respectively, P < 0.001)
`
`Pts with
`various
`symptoms
`(N = 307),
`PHN
`(43/307)
`
`Gabapentin, 900 mg/d (titrated Gabapentin demonstrated
`over 3 days), with escalation
`greater improvement in pain
`to 1,800 mg/d or 2,400 mg/d,
`score than placebo (21% vs
`for a total of 8 wks (N = 153),
`14%, P = 0.048)
`or placebo (N = 152)
`
`Watson et al.,
`1993 [35]
`
`Pts with PHN Capsaicin 0.075% cream vs
`(N = 143)
`placebo (vehicle) cream
`
`Capsaicin resulted in greater
`decrease in pain than placebo
`(measured by VAS) at 2 wks
`(19% vs 0.4%, P < 0.05) and
`6 wks (P = 0.032). Long-term
`follow up (£2 years, N = 77)
`showed clinical benefit in
`86% of patients
`
`Page 00004
`
`

`
`S32
`
`Table 2 Continued
`
`Backonja and Serra
`
`Study
`
`Patients
`
`Treatment
`
`Outcomes
`
`Adverse events/withdrawals
`
`Randomized, double-blind, placebo-controlled, crossover trials
`Baranowski
`Pts with PHN Pts received each of the
`(N = 24)
`et al., 1999
`following IV infusions over
`2 h ≥1 wk apart: placebo
`[92]
`(normal saline), lidocaine
`1 mg/kg, and lidocaine
`5 mg/kg
`
`Brill et al.,
`2002 [93]
`
`Pts with PHN Magnesium sulphate, 30 mg/kg,
`(N = 7)
`IV, or saline
`
`De Benedittis
`and
`Lorenzetti,
`1996 [94]
`
`Galer et al.,
`1999 [31]
`
`Pts with PHN Pts had 1 of 4 suspension/
`(N = 22)
`diethyl ether solutions
`AHN
`applied to affected areas in
`(N = 15)
`a randomized order on 4
`different days. Median
`doses of active suspensions:
`aspirin 1,000 mg;
`indomethacin 75 mg;
`diclofenac 100 mg; lactose
`was used for placebo
`Topical 5% lidocaine patch vs
`placebo (vehicle) patch for
`2–14 days, depending on
`increase in pain; then
`patients crossed over to
`alternative treatment
`
`Pts with PHN
`(N = 33)
`
`Ongoing pain (measured by VAS)
`was significantly reduced after
`all infusions (P < 0.05); dynamic
`pressure-evoked pain was
`significantly reduced by both
`lidocaine infusions (P < 0.05,
`for each dosage level); area of
`allodynia declined with lidocaine
`1 and 5 mg/kg (P < 0.05 and
`P < 0.001, respectively)
`Mean pain score decreased from
`6.7 to 1.9 at 30 minutes
`posttreatment with magnesium
`sulphate. Pain scores were
`significantly lower with
`magnesium sulphate than with
`placebo at 20 and 30 minutes
`(P = 0.016)
`Only aspirin was significantly
`superior to placebo for reduction
`in pain (based on VAS score)
`from baseline (P < 0.05) and
`duration of pain (P < 0.01).
`Good-to-excellent results were
`reported in >81% of PHN pts
`with topical aspirin suspension
`
`Primary end point was “time to
`exit,” i.e., pts were allowed
`to discontinue treatment if pain
`relief diminished. Median time
`to exit was significantly better
`with lidocaine than placebo
`(14 d vs 3.8 d, P < 0.001).
`Lidocaine patch was preferred
`by 78.1% of pts vs 9.4% for
`placebo (P < 0.001)
`
`Kishore-
`Kumar et al.,
`1990 [25]
`
`Pts with PHN Desipramine (mean dose:
`(N = 26)
`167 mg/d) or placebo
`¥6 wks; then pts crossed
`over to alternative treatment
`
`Pain relief with desipramine was
`significantly greater from weeks
`3 to 6 than with placebo
`(P < 0.001)
`
`Downloaded from
`
`http://painmedicine.oxfordjournals.org/
`
` by guest on November 13, 2016
`
`No pts at the lower dose
`reached toxic plasma levels;
`however, several pts at the
`higher dose did reach toxic
`levels. Thus, the lower doses
`may be considered safe
`
`No side effects were
`reported during treatment
`with magnesium sulphate
`
`Mild cutaneous rash in 1 pt
`each with indomethacin and
`diclofenac
`
`Withdrawals: 1 pt suffered a
`stroke prior to receiving
`study medication; 1 pt
`withdrew during placebo
`period because of increased
`pain and insomnia; 1 pt
`stopped placebo because
`of red, irritated skin; AEs
`were mild or moderate;
`application site reaction
`redness/rash reported in
`9 pts with lidocaine patch
`and 11 pts with placebo
`patch
`Withdrawals: 8 pts because of
`AEs or intercurrent medical
`illnesses; AEs: desipramine:
`syncope, 1 pt; left bundle
`branch block, 1 pt; jitteriness
`and atypical chest pain, 1 pt;
`fever, 1 pt; and vertigo, 1 pt;
`placebo: vertigo and nausea,
`1 pt; skin rash, 1 pt;
`unsteadiness + mental
`fogginess, 1 pt
`AEs: dry mouth, sedation,
`dizziness; occurred with
`both active treatments
`
`Max et al.,
`1988 [21]
`
`Nelson et al.,
`1997 [95]
`
`Pts with PHN Amitriptyline (12.5–150 mg/d),
`(N = 58)
`lorazepam (0.5–6 mg/d), or
`placebo (lactose) ¥2 wks;
`followed by 1-wk washout;
`then crossed to alternative
`treatment
`Pts with DPN Oral dextromethorphan (mean
`(N = 14)
`dose in PHN: 439 mg/d) or
`placebo ¥6 wks followed by
`and with
`PHN
`1-wk washout; then crossed
`(N = 18)
`over to alternative treatment
`
`Moderate or greater pain relief
`was reported by 47% of pts
`with amitriptyline, 16% of pts
`with placebo, and 15% of
`pts with lorazepam
`
`Dextromethorphan did not reduce Withdrawals: 5 PHN pts due
`pain in pts with PHN to a
`to sedation, ataxia and
`greater extent than placebo
`confusion, and (unrelated)
`(P = 0.72)
`6th cranial nerve palsy
`
`Page 00005
`
`

`
`Downloaded from
`
`http://painmedicine.oxfordjournals.org/
`
` by guest on November 13, 2016
`
`S33
`
`Adverse events/withdrawals
`
`Withdrawals: opioids, 20 pts;
`TCA = 6 pts; placebo, 1 pt
`(P < 0.01); AEs:
`constipation, nausea,
`dizziness, drowsiness, loss
`of appetite, and dry mouth
`
`AEs: bruising and pain upon
`patch removal, 1 pt; mild
`skin reddening, 2 pts (1 pt
`each with lidocaine and
`vehicle patch)
`
`AEs included sedation (71%,
`63%, and 38%), dry mouth
`(30%, 21%, and 25%), and
`GI distress (17%, 0%, and
`0%) for dextromethorphan,
`memantine, and lorazepam,
`respectively
`AEs resulting in withdrawal:
`amitriptyline: dry mouth
`combined with constipation,
`sedation, dizziness, lethargy,
`mouth ulcers, and nausea
`(1 pt each); maprotiline: dry
`mouth and nausea, nausea
`and vomiting, restless legs
`(1 pt each)
`Withdrawal: 1 pt because
`of erythema multiforme;
`AEs: dry mouth, 16 pts;
`drowsiness, 4 pts;
`constipation, 2 pts; increased
`breast size, 1 pt; rash, 1 pt
`
`Pts with PHN Pts randomly assigned to
`(N = 35)
`amitriptyline or maprotiline.
`Median dose of both agents
`was 100 mg/d ¥ 5 wks;
`followed by 2-wk washout
`then crossed over to
`alternative treatment
`
`Pts with PHN Amitriptyline, 12.5–25 mg/d,
`(N = 24)
`increased to median dose
`of 75 mg, or placebo ¥3 wk;
`followed by a 1- to 2-wk
`washout, then pts crossed
`over to alternative
`treatment
`Pts with PHN Oxycodone, 10 mg (increased
`(N = 50)
`to a maximum of 30 mg), or
`placebo every 12 h ¥ 4 wks,
`followed by crossover to the
`alternative treatment
`
`Pts with PHN Amitriptyline or nortriptyline,
`(N = 33)
`10 mg (≥65 y of age), or
`20 mg (<65 y of age), with
`dosage increases ¥5 wks;
`followed by 2-wk washout,
`then crossed over to
`alternative treatment
`
`Amitriptyline was significantly
`better than maprotiline at
`relieving pain as measured by
`VAS score (P < 0.01)
`
`Changes in VAS scores
`demonstrated that amitriptyline
`was superior to placebo in
`relieving pain (P £ 0.0001)
`
`Reduction in pain was significantly Withdrawals: 6 pts with
`better with oxycodone than with
`oxycodone; 5 pts with
`placebo (P = 0.0001)
`placebo; AEs with
`oxycodone: constipation,
`5 pts; nausea, 4 pts;
`sedation, 3 pts
`Withdrawals: 2 pts left due to
`AEs (1 pt taking nortriptyline
`experienced increased pain
`fever, epigastric pain, bad
`dreams, and perspiration;
`1 pt taking amitriptyline
`experienced slurred speech
`and urinary retention)
`
`No difference between the two
`drugs in any pain parameter
`
`Abbreviations: AEs = adverse events; AHN = acute herpetic neuralgia; d = day; GI = gastrointestinal; h = hour; min = minute; NA = not available; NPS = Neuro-
`pathic Pain Scale; pts = patients; wk = week; y = year.
`
`Pharmacologic Management Part 1
`
`Table 2 Continued
`
`Study
`
`Patients
`
`Treatment
`
`Outcomes
`
`Greater mean decreases in pain
`ratings on a 0–10 scale were
`achieved with TCAs (1.4) and
`with opioids (1.9) than with
`placebo (0.2, P < 0.001)
`
`Pts with PHN Each pt was scheduled to
`(N = 76)
`undergo three treatment
`periods (8 wks each) in
`random order: opioid (mean
`dose: morphine 91 mg or
`methadone 15 mg); TCA
`(mean dose nortriptyline
`89 mg or desipramine
`63 mg); and placebo
`Pts with PHN Pts randomly assigned (as to
`(N = 35)
`session order) to four 12-h
`long treatment sessions:
`5% lidocaine patch (2
`sessions), vehicle patch,
`and observations only
`
`Pain intensity reductions
`significantly greater with
`lidocaine patch than with vehicle
`patch (P < 0.001 to P = 0.038 at
`individual time points) and with
`observation only (P = 0.0001 to
`P = 0.021 at individual time
`points); pain reductions
`significantly greater with
`vehicle patch than with
`observation only at 2-h
`(P = 0.016) and 6-h (P = 0.041)
`time points
`Pts with DPN Median doses for pts with PHN Mean reductions in pain intensity
`(N = 23)
`were: dextromethorphan
`in pts with PHN were 6% with
`and with
`400 mg/d, memantine
`dextromethorphan, 2% with
`PHN
`35 mg/d, or lorazepam
`memantine, and 0%
`(N = 21)
`(active placebo) 1.2 mg/d
`with lorazepam
`
`Raja et al.,
`2002 [26]
`
`Rowbotham
`et al., 1996
`[32]
`
`Sang et al.,
`2002 [38]
`
`Watson et al.,
`1992 [23]
`
`Watson et al.,
`1982 [20]
`
`Watson and
`Babul, 1998
`[29]
`
`Watson et al.,
`1998 [24]
`
`Page 00006
`
`

`
`Downloaded from
`
`http://painmedicine.oxfordjournals.org/
`
` by guest on November 13, 2016
`
`S34
`
`application site of the patch in patients who
`used either the lidocaine patch or the vehicle
`patch [31].
`Reduction in pain, as measured on a VAS, has
`also been observed with the topical agent capsaicin.
`Capsaicin binds to receptors (VR1/TRPV1) on
`subpopulations of sensory nociceptive C or Ad
`fibers. Initially, capsaicin causes pain by initiating
`nociceptor firing, resulting in increased sensitivity
`to painful thermal and mechanical stimuli. An
`analgesic effect follows the painful experience as
`prolonged exposure to capsaicin desensitizes noci-
`ceptive terminals through partial or complete
`degeneration of axon terminals or the neuron [34].
`However, the initial burning and stinging at the
`application site may be intolerable to some
`patients [35].
`
`Emerging Treatment Regimens
`Pregabalin, a new a2d ligand with analgesic, anxi-
`olytic, and anticonvulsant activities, is currently
`being investigated for use in the treatment of neu-
`ropathic pain [36]. Dworkin et al. reported the
`results of a randomized, double-blind study involv-
`ing 173 patients with PHN [37]. Patients treated
`with pregabalin (600mg/day or 300mg/day based
`on creatinine clearances of >60mL/minute or
`30–60mL/minute, respectively) had a significantly
`lower mean pain score than patients who received
`placebo (3.60 vs 5.29, P = 0.0001), and 50% of
`the pregabalin-treated patients reported a ≥50%
`reduction in mean pain scores, compared with only
`20% of patients who received placebo (P = 0.001).
`Compared with patients in the placebo group, pre-
`gabalin-treated patients also experienced signifi-
`cantly greater improvements in sleep (P = 0.0001).
`Side effects associated with pregabalin included
`dizziness, somnolence, peripheral edema, and dry
`mouth. In most cases, side effects were of mild to
`moderate intensity.
`Sang et al.
`investigated the use of dex-
`tromethorphan and memantine, low-affinity N-
`methyl-d-aspartate (NMDA) antagonists, in the
`treatment of neuropathic pain in patients with
`DPN and PHN [38]. In patients with PHN, the
`mean reduction in pain intensity from baseline was
`7% with dextromethorphan, 2% with memantine,
`and 0% with lorazepam (active placebo). Although
`these results were not statistically significant, 29%
`of patients with PHN had moderate or better pain
`relief with dextromethorphan, as did 12% with
`memantine. Adverse events with dextromethor-
`phan and memantine included sedation, dry
`mouth, and gastrointestinal distress.
`
`Backonja and Serra
`
`Non–Disease-Specific Trials: Anticonvulsant
`Serpell et al. [39] conducted a study designed to
`assess the impact of gabapentin on specific neuro-
`pathic pain symptoms in patients with various neu-
`ropathic pain conditions. Evidence suggests that
`neuropathic pain symptoms reflect an underlying
`pathophysiology and, therefore, may respond to
`the same pharmacologic agents regardless of eti-
`ology. Of the 307 patients enrolled in that study,
`43 had PHN. Although data for patients with
`PHN were not reported specifically, overall,
`higher percentages of gabapentin-treated patients,
`than patients in the placebo group, experienced
`improvements in allodynia (23% vs 15%), shoot-
`ing pain (32% vs 24%), burning pain (23% vs
`15%), and hyperalgesia (26% vs 17%), although
`no clear inference on specific mechanisms that
`responded better to gabapentin could be estab-
`lished. The most common adverse events observed
`with gabapentin treatment in those studies were
`somnolence, dizziness, ataxia, peripheral edema,
`and infections.
`
`Number Needed to Treat
`The importance of data generated from clinical
`studies is that the knowledge gained can be trans-
`lated to clinical practice. Thus, clinicians who are
`making decisions about the management of indi-
`vidual patients need to have a measurement of
`treatment effect. This enables clinicians to evalu-
`ate the results of various clinical trials that have
`different end points and use different statistical
`analytic methods. The number needed to treat
`(NNT), which indicates the number of patients
`that would have to be treated in order to obtain a
`positive therapeutic response, or to prevent one
`untoward event, is calculated as the reciprocal of
`the absolute risk reduction, that is, the difference
`between event rates in the treatment and placebo
`groups [40]; for example NNT = 1/(response
`rate to treatment)–(response rate to placebo).
`However, when making clinical decisions based on
`NNT values, certain caveats should be consid-
`ered—pooled NNT values are not adjusted for
`study quality or size; different studies use different
`outcome measures; there can be differences in
`clinical settings or patient characteristics; and
`there may be secular trends in incidence or
`morality [41].
`Collins et al. analyzed data from randomized
`trials to calculate NNT values for anticonvulsants
`and antidepressants used to treat neuropathic pain
`in patients with PHN [42]. They determined that
`the NNT to obtain a ≥50% pain relief in one
`
`Page 00007
`
`

`
`Downloaded from
`
`http://painmedicine.oxfordjournals.org/
`
` by guest on November 13, 2016
`
`Pharmacologic Management Part 1
`
`patient is 3.2 for anticonvulsants and 2.1 for anti-
`depressants. Sindrup and Jensen reported NNT
`values of 2.3 for TCAs, 2.5 for oxycodone, and 5.3
`for capsaicin in PHN [43].
`
`Number Needed to Harm
`The number needed to harm (NNH) indicates the
`number of patients that would need to be treated
`in order for one patient to have an adverse event.
`Calculation of the NNH uses the same basic equa-
`tion as for the NNT, that is, NNH = 1/(adverse
`event rate to treatment)–(adverse event rate of
`placebo). In a report by Collins et al. [42], NNH
`data were pooled for both PHN and DPN in a
`comparison of antidepressants (TCAs and selec-
`tive serotonin reuptake inhibitors [SSRIs]) with
`placebo. TCAs were found to have an NNH value
`of 14 for major adverse events while SSRIs were
`shown to be no different than placebo. Although
`there were no NNH anticonvulsant data available
`for major adverse events, for m

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket