Open access

Predictive Factors for Postherpetic Neuralgia and Recent Pharmacotherapies

Written By

Yuko Kanbayashi and Toyoshi Hosokawa

Submitted: 11 May 2012 Published: 27 March 2013

DOI: 10.5772/55278

Chapter metrics overview

1,946 Chapter Downloads

View Full Metrics

1. Introduction

Postherpetic neuralgia (PHN) is a form of refractory chronic neuralgia that, despite the importance of prevention, currently lacks any effective prophylaxis. The efficacy of live zoster vaccine in preventing PHN was recently reported [1]. However, this vaccine appeared to be of limited use in prophylaxis[2]. PHN has a variety of symptoms and significantly affects patient quality of life [3-12]. Various studies have statistically analyzed predictive factors for PHN [13-23], but neither obvious pathogenesis nor established treatment has been clarified or established. We designed and conducted a study on the premise that statistical identification of significant predictors for PHN would contribute to the establishment of an evidence-based medicine approach to the optimal treatment of PHN. As a result, we reported our paper “Predictive Factors for Postherpetic Neuralgia Using Ordered Logistic Regression Analysis” [24].

In this review, we discuss predictors for PHN based on our results, and further review recent pharmacotherapeutic results.

Advertisement

2. Predictors for PHN

Previous studies have shown that older age, female sex, presence of a prodrome, greater rash severity, and greater acute pain severity are predictors of increased PHN [14-18, 25]. Some other potential predictors (ophthalmic localization, presence of anxiety and depression, presence of allodynia, and serological/virological factors) have also been studied [14, 18]. We conducted a retrospective study to identify significant predictors of PHN that would contribute to the establishment of evidence-based medicine approaches to the optimal treatment of PHN [24].

The participants were 73 patients with herpes zoster who had been treated at the pain clinic of our hospital between January 2008 and June 2010. Variables present at the initial visit were extracted from the clinical records for regression analysis of factors related to the occurrence of PHN. The following scores for response were used: 0 = no PHN after 3 months; 1 = PHN present after 3 months, but absent after 6 months; and 2 = PHN present after 6 months. Multivariate ordered logistic regression analysis was performed to identify predictive factors for PHN. Table 1 shows the clinical characteristics of patients and various factors that could be related to the occurrence of PHN. Multivariate ordered logistic regression analysis identified advanced age and deep pain at the initial visit to our outpatient pain clinic as factors predicting the occurrence of PHN. diabetes mellitus (DM) and pain reduced by bathing also showed high odds ratios, but were not significant predictive factors (Table 2). In conclusion, advanced age and deep pain at first visit were identified as predictive factors for PHN. DM and pain reduced by bathing should also be considered as potential predictors of PHN [24]. To improve patient safety and the likelihood of achieving satisfactory outcomes, prospective studies are needed to establish optimal treatments including pharmacotherapy for PHN. Next section we further review recent pharmacotherapeutic results for PHN.

(0/1) or (0/1/2), median (range)
Demographics
Significant zoster-associated pain 35/13/25
Age 8/46/19, 69 (27-90)
Sex 37/36
Complication
Hypertension 44/29
Angina 69/4
Diabetes mellitus 61/12
Malignant tumor 50/23
Autoimmune disease 68/5
Sleep disorder 36/37
Location
Trigeminal nerves 58/15
Cervical nerves 58/15
Thoracic nerves 42/31
Lumbar nerves 61/12
Sacral nerves 70/3
Period of onset, type and extent of pain
Period before herpes zoster onset (days) 30 (1-3000)
Log (period before herpes zoster onset) 1.477 (0-3.477)
Prodromal rash 39/34
Prodromal pain 34/39
Allodynia 35/38
VAS (mm) 66 (0-100)
Pain reduced when bathing 41/32
Pain (superficial) 35/38
Pain (deep) 38/35
Pain (continuous) 43/30
Pain (breakthrough) 31/42

Table 1.

Clinical characteristics of 73 patients and various factors that could be related to the occurrence of PHN [24]

Binary scales were: female = 0, male = 1 for sex; and absent = 0, present = 1 for others.


The ordered scale was: absent after 3 months = 0, present = 1, and present after 6 months = 2 for significant zoster-associated pain; and <50 years = 0, 51-74 years = 1, ≥75 years = 2 for age.


VAS, visual analog scale


Variable EV SE χ2 value P OR CI of OR
Lower 95% Upper 95%
Age 1.008 0.461 4.78 0.0288* 2.740 1.110 6.761
Prodromal pain 0.442 0.532 0.69 0.4059 1.556 0.549 4.413
DM 1.123 0.693 2.63 0.1049 3.075 0.791 11.952
Allodynia 0.201 0.630 0.10 0.7489 0.818 0.238 2.808
Pain reduced by bathing 1.221 0.745 2.68 0.1014 3.389 0.787 14.601
Deep pain 1.446 0.682 4.49 0.0341* 4.244 1.114 16.163
Breakthrough pain 0.687 0.598 1.32 0.2506 1.988 0.616 6.418
Sleep disorder 0.146 0.514 0.08 0.7757 1.158 0.423 3.169

Table 2.

Results of multivariate ordered logistic regression analysis for variables extracted by forward selection with addition of prodromal pain and allodynia [24].

EV, estimated value; SE, standard error; OR, odds ratio; CI, confidence interval; DM, diabetes mellitus


*P<0.05


Advertisement

3. Recent pharmacotherapy for PHN

To date, no clear predictors of treatment response have been identified in patients with neuropathic pain, including PHN. Various types of drugs have shown consistent efficacy in randomized clinical trials and meta-analysis [9]. The modes of action and information on dosing, precautions, and adverse events (AEs) for the different drug classes are summarized in Table 3 [9]. Numbers needed to treat (NNTs), numbers needed to harm (NNH) [26, 27] and AEs for the different drugs are shown in Table 4 [28].

Among systemic therapies, tricyclic antidepressants (TCAs), calcium channel α2-δ ligands (gabapentin and pregabalin), and extended-release formulations of morphine and oxycodone show good efficacy in patients with PHN. Current guidelines suggest, perhaps to maximize the benefit-risk ratio, that TCAs should be preferred over opioids, and among TCAs, second-generation TCAs such as nortriptyline, desipramine, and imipramine are preferred over the first-generation agent, amitriptyline [29]. Opioids, specifically oxycodone, morphine, methadone, and tramadol, are also effective against PHN. However, addiction and regulatory issues, coupled with the benign adverse event profile of anticonvulsants, make opioids a secondary choice in PHN [28].

The American Academy of Neurology (AAN; 2004), European Federation of Neurological Societies (NeuPSIG; 2007), and European Federation of Neurological Societies (EFNS; 2010) guidelines all recommend TCAs and pregabalin as first-line oral therapies for patients with PHN [11, 30, 31]. The NeuPSIG and EFNS guidelines also recommend gabapentin as first-line therapy for PHN. The NeuPSIG and EFNS guidelines state that secondary amine TCAs should generally be considered instead of the tertiary amine amitriptyline, due to the superior safety profiles [11, 29, 30].

Japan Society of Pain Clinicians (JSPC; 2011) guidelines recommend TCAs (particularly the secondary amines), calcium channel α2-δ ligands, and extract of cutaneous tissue of rabbits inoculated with vaccinia virus (Neurotropin®) as first-line oral therapies [32].

Mode of action Major Adverse- events

Precautions

Other benefits
Starting dose/maximum dose

Titration

Duration of adequate trial
Nortriptyline
Desipramine
Inhibition of reuptake of
serotonin and/or
noradrenaline, blockage of
sodium channels,
anticholinergic
Sedation, ant-cholinergic
effects (e.g., dry
mouth or urinary
retention, weight gain)

Cardiac disease ,
glaucoma, seizure
disorder, use of
tramadol

Improvement of
depression and
sleep disturbance
25 mg at bedtime/150 mg daily

Increase by 25 mg every 3-7 day as tolerated

6-8 weeks (at least 2 weeks
Maximum tolerated dose)
Gabapentin Decreases release of
glutamate, noradrenaline,
and substance P, with ligands
on α2-δ subunit of voltage-gated calcium channel
Sedation, dizziness, peripheral edema

Renal Insufficiency

No clinically significant drug interactions
100-300 mg once to
three times
daily/1200 mg three
times daily; reduce if
renal function impaired

Increase by
100-300 mg three
times daily every
1-7 days as
Tolerated

4 weeks
Pregabalin Decreases release of
glutamate, noradrenaline,
and substance P, with ligands on α2-δ subunit of voltage-gated calcium
channel
Sedation, dizziness,
Peripheral edema

Renal Insufficiency

No clinically significant drug interactions,
improvement of sleep disturbance and anxiety
50 mg three times daily
or 75 mg twice
daily/200 mg three
times or 300 mg twice
daily, reduce if
renal function impaired

Increase to 300 mg
daily after
3-7 days, then by
150 mg daily every
3-7 days, as
Tolerated

4 weeks
5% lidocaine
patch
Blockage of sodium channels Local erythema,
Rash

None

No systemic adverse events
1-3 patches/3 patches

None

2 weeks
Morphine,
oxycodone,
methadone,
levorphanol
µ-receptor agonism
(oxycodone also causes
к-receptor antagonism)
Nausea/
vomiting,
constipation,
dizziness

History of substance
abuse, suicide risk,
driving impairment

Rapid onset of
analgesic effect
10-15 mg morphine
every 4 h or as needed
(equianalgesic doses
should be used for other opioids)/no
maximum doses

After 1-2 weeks
convert to long-acting
opioids/
transdermal
application, use
short-acting drug
as needed and as
tolerated

4-6 weeks

Table 3.

Recommended first-line treatments for patients with PHN [9]

Recommendations are all grading level A = good scientific evidence suggesting that the benefits of the treatment substantially outweigh the potential risks.


Drug NNT (95%CI) NNH (95%CI) Specific/Common Adverse Effects
Amitriptyline 1.6 (1.2-2.4)
4.2 (2.13-81.6)
Minor harm:
8 (2.5-22)
Major harm:
24 (8-36)
Sedation, dry mouth, tachycardia,
constipation, urinary retention, weight gain,
prolonged QT interval
Desipramine 1.9 (1.3-3.7) Minor harm: 4.8 (2.5-36.7)
Major harm: 13*
Nortriptyline 3.7 (2.4-8) ND
Gabapentin 4.4 (3.3-6.1) Minor harm: 4.1 (3.2-5.7)
Major harm: 12.3 (7.7-30.2)
Somnolence, dry mouth, weight gain, peripheral edema, ataxia
Pregabalin 4.9 (3.7-7.6) Minor harm: 4.3 (2.8-9.2)
Major harm: ND
Dizziness, somnolence, peripheral edema
Oxycodone 2.5 (1.7-4.4) Minor harm: 3.6 (2.2-10.2)
Major harm: 6.3 (4.2-12.8)
Immune suppression, loss of libido, endocrine dysfunction
Morphine-controlled
release or methadone
2.8 (2-4.6) ND
Tramadol 4.8 (3.5-6.0) Minor harm: 7.2*
Major harm: 10.8*
Somnolence, constipation, nausea and vomiting,
seizures, serotonin syndrome when combined
with SSRIs and MAO inhibitors

Table 4.

NNT, NNH, and adverse effects of selected drugs for PHN [28]

Values for NNTs and NNHs were adapted from Hempenstall et al. [27] and Wu and Raja [26]. NNTs for amitriptyline were based on studies by Watson et al. [49] and Max et al. [50].


*The 95% confidence interval (CI) could not be determined by Hempenstall et al. [27] or by Wu and Raja [26].


ND, could not be determined by Hempenstall et al [27]; SSRIs, selective serotonin reuptake inhibitors; MAO, monoamine oxidase


3.1. Calcium hannel α2-δ Ligands

Gabapentin and pregabalin suppress the release of excitatory neurotransmitters on binding to the α2-δ subunit of potential-dependent calcium channels in the central nervous system (CNS). Both drugs are structurally similar and share similar mechanisms of action, but pregabalin shows a linear pharmacokinetic profile and strong affinity for the α2-δ subunit. Both drugs have been widely studied in peripheral pain syndromes, although pregabalin has been the focus of most studies in central neuropathic pain syndromes. Only a few drug interactions have been reported for both drugs and the agents are well tolerated, but must be used at lower doses in patients with decreased renal function [9, 32]. The EFNS, AAN, and NeuPSIG guidelines recommend the calcium channel α2-δ ligands gabapentin and/or pregabalin for the treatment of PHN [11, 29-31]. In a meta-analysis, NNTs before 1 patient achieves 50% pain reduction for gabapentin (3 trials, 559 patient episodes) and pregabalin (3 trials, 411 patient episodes) for a 50% reduction in pain were 4.4 and 4.9, respectively [27, 29] (Table 4). Subsequent Cochrane reviews for both drugs calculated NNTs of 7.5 for gabapentin [29, 33] and 3.9 for pregabalin 600 mg [29, 34]. Gabapentin and pregabalin are effective and are the most commonly prescribed drugs in PHN because of their efficacy and benign AEs profile.

3.1.1. Pregabalin

Pregabalin is a 3-isobutyl derivative of gamma-amino butyric acid (GABA) with anti-convulsant, anti-epileptic, anxiolytic, and analgesic activities. Although the exact mechanisms of action are unclear, pregabalin selectively binds to α2-δ subunits of presynaptic voltage-dependent calcium channels located in the CNS. Binding of pregabalin to α2-δ subunits of presynaptic voltage-dependent calcium channels prevents calcium influx and the subsequent calcium-dependent release of various neurotransmitters, including glutamate, noradrenaline, serotonin (5-Hydroxytryptamine: 5-HT), dopamine, and substance P, from the presynaptic nerve terminals of hyperexcited neurons; synaptic transmission is inhibited and neuronal excitability is diminished. Pregabalin does not bind directly to GABA-A or GABA-B receptors and does not alter GABA uptake or degradation [35].

Pregabalin has also been associated with dose-related risks of somnolence (5-14%), dizziness (7-28%), and peripheral edema (6-16%) [29, 32, 34]. At a higher dose (600 mg/day), pregabalin has been associated with weight gain (9%), asthenia (9%), dry mouth (6%), and vertigo (5%) [28, 29, 36]. Discontinuations due to these AEs were ≤1% in patients treated with pregabalin at 150 mg/day and 4-15% in patients receiving a 600-mg/day dose. Serious AEs were reported in 2-5% of patients treated with pregabalin at 150, 300, and 600 mg/day, compared with 3% in those assigned to receive a placebo [29, 36]. Symptoms including insomnia, nausea, headache, and diarrhea were reported by some patients following abrupt withdrawal of pregabalin. Pregabalin should therefore be tapered gradually over a minimum of 1 week rather than discontinued abruptly. Weight gain noted in clinical trials of pregabalin was not limited to patients with peripheral edema. Weight gain was related to dose and duration of exposure to pregabalin, but did not appear to be associated with sex, age, or baseline body-mass index. A higher incidence of weight gain and peripheral edema were noted in patients taking both pregabalin and a thiazolidinedione, compared with patients taking either agent alone. As a result, care should be taken when co-administering pregabalin and one of these agents [37].

JSPC guidelines recommend beginning pregabalin treatment at 75 mg/day once before bed, 150 mg/day as two doses after breakfast and supper, or 150 mg/day as three doses after each meal. Even when renal function is normal, consider a very low dose only just before bed, such as 25 mg/day once before bed, for elderly patients, patients of low body weight, and others prone to AEs [32].

3.1.2. Gabapentin

Gabapentin is a synthetic analogue of the neurotransmitter GABA with anticonvulsant activity. Although the exact mechanisms of action are unknown, gabapentin appears to inhibit excitatory neuron activity. This agent also exhibits analgesic properties [35].

In clinical trials, the most frequent AEs observed with gabapentin were somnolence (16%), dizziness (21%), and peripheral edema (8%) [33]. Twelve percent of patients discontinued gabapentin owing to AEs, compared with 8% who discontinued placebo. Serious AEs with gabapentin and placebo occurred in 4% and 3% of patients, respectively [29].

3.2. Opioids

Opioid analgesics are agonists at presynaptic and postsynaptic opioid receptors. Opioids offer comparable analgesic efficacy to TCAs. Concerns about long-term AEs, such as immunological changes, physical dependency, and misuse or abuse, can limit the use of strong opioids in patients with neuropathic non-cancer-related pain [9].

The place of opioids and tramadol in guidelines for the management of PHN has evolved over time. In the 2004 AAN guidelines, extended-release formulations of oxycodone and morphine were recommended as first-line agents. Guidelines issued by the NeuPSIG (2007) and EFNS (2010) recommend strong opioids and tramadol as second-line therapy, not because of new data on efficacy, but perhaps reflecting concerns about the risks of AEs and abuse [29].

Extended-release formulations of oxycodone [38] and morphine, as well as methadone [39], have shown efficacy in patients with PHN. The NNT for a 50% reduction in pain were 2.5 for oxycodone and 2.8 for morphine or methadone, compared with an NNT of 3.7 for the TCA nortriptyline (Table 4). In a placebo-controlled, active comparator-controlled crossover trial, the majority of patients (53%) preferred the opioids morphine or methadone over TCAs. Opioids tended to provide greater pain relief, although the difference was not significant [40]. A meta-analysis of the 2 trials evaluating oxycodone, morphine and methadone calculated an NNT for opioids of 2.7 [27]. Tramadol has proven less effective than strong opioids. In a randomized controlled trial, tramadol was not significantly better than placebo on a 5-point verbal scale or on measures of quality of life [40]. An analysis of this trial calculated an NNT of 4.8 [27]. Oxycodone, morphine, and methadone were associated with typical opioid-related AEs, including nausea, diarrhea, and constipation [38, 39]. Tramadol was better tolerated, but less effective [40].

In the JSPC guideline, although multiple clinical studies have demonstrated the analgesic effects of opioid analgesics (narcotics for medical use) in patients with neuropathic pain, opioids are generally recommended as second- or third-line treatment for several reasons. First, opioid analgesics have a high incidence of associated AEs that may persist over the entire course of treatment. Second, the lack of any systematic investigation into the long-term safety of opioid analgesics means that the opioid analgesics may not be fundamentally safer than other drug options. Third, opioid analgesics may cause hyperalgesia. This effect, if present, would adversely modify the risk/benefit profile of long-term treatment in neuropathic pain patients. Finally, opioid abuse and addiction are substantial problems [32].

3.2.1. Morphine

The sulfate salt of morphine is an opiate alkaloid isolated from the plant Papaver somniferum and produced synthetically. Morphine binds to and activates specific opiate receptors (δ, μ and κ), each of which are involved in controlling different brain functions. In the CNS and gastrointestinal system, this agent exerts widespread effects including analgesia, anxiolysis, euphoria, sedation, respiratory depression, and smooth muscle contraction in the gastrointestinal system.

A sustained-release tablet formulation contains the sulfate salt of the opiate alkaloid morphine to provide analgesic activity. Morphine binds to and activates μ-opioid receptors in the CNS, thereby mimicking the effects of endogenous opioids. Binding of morphine to opioid receptors stimulates exchange of guanosine 5'-triphosphate for guanosine 5'-diphosphate, inhibits adenylate cyclase, and decreases intracellular cyclic adenosine monophosphate. This inhibits the release of various nociceptive neurotransmitters, such as substance P, GABA, dopamine, acetylcholine, noradrenaline, vasopressin, and somatostatin. In addition, morphine closes N-type voltage-gated calcium channels and opens calcium-dependent inwardly rectifying potassium channels, causing hyperpolarization of neuronal membranes and reductions in neuronal excitability, with subsequent analgesia and sedation [35].

3.2.2. Oxycodone hydrochloride

The hydrochloride salt of oxycodone is a methylether of oxymorphone and a semisynthetic opioid agonist with analgesic and antitussive properties. Oxycodone binds to μ-receptors in the CNS, thereby mimicking the effects of endogenous opiates as well as morphine. In addition to analgesia and a depressive effect on the cough center in the medulla, this agent may cause euphoria, anxiolysis, miosis, sedation, physical dependence, constipation, and respiratory depression, depending on dosage and variations in individual metabolism [35].

3.2.3. Tramadol

A synthetic codeine analogue, tramadol has central analgesic properties with effects similar to opioids, such as morphine and codeine, acting on specific opioid receptors. Used as a narcotic analgesic for severe pain, this agent can be addictive. Tramadol is used to treat moderate-to-severe pain in adults, and binds to opioid receptors in the CNS[41]. Tramadol also weakly inhibits noradrenaline and 5-HT reuptake and can therefore interact with serotonergic drugs (selective noradrenaline reuptake inhibitors [SNRIs] and selective serotonin reuptake inhibitors [SSRIs]), causing serotonin syndrome, although this risk seems to be low in clinical practice [9].

3.3. TCAs

Clinical data support the efficacy of TCAs for PHN. In a 2005 meta-analysis of 4 clinical trials (248 patient episodes), the NNT for 1 patient to achieve a 50% reduction in pain with the TCAs amitriptyline, nortriptyline, and desipramine was 2.6 [27]. A 2007 Cochrane review of 4 clinical trials (244 patients) calculated an NNT of 2.7 [41]. Although effective, TCAs have been associated with significant systemic AEs, most notably anticholinergic AEs [43, 44], and have been associated with cardiotoxicity [45-47]. Electrocardiography (ECG) before the start of treatment is mandatory and careful dose titration is needed [9]. Individual trials of amitriptyline and desipramine in patients with PHN suggest that the second-generation TCA desipramine produces fewer anticholinergic side effects than amitriptyline [48, 49].

3.3.1. Amitriptyline hydrochloride

The hydrochloride salt of the tricyclic dibenzocycloheptadiene amitriptyline with antidepressant and antinociceptive activities. Amitriptyline hydrochloride is the hydrochloride salt of the tricyclic dibenzocycloheptadiene amitriptyline, and shows antidepressant and antinociceptive activities. Amitriptyline inhibits the re-uptake of noradrenaline and serotonin by the presynaptic neuronal membrane in the CNS, thereby increasing synaptic concentrations of noradrenaline and serotonin. Due to constant stimulation of these receptors, amitriptyline may produce a downregulation of adrenergic and serotonin receptors, which may contribute to the antidepressant activity. In the CNS, the antinociceptive activity of this agent may involve high-affinity binding to and inhibition of N-methyl-D-aspartate (NMDA) receptors and/or enhancement of the action of serotonin at the spinal terminals of an opioid-mediated intrinsic analgesia system [35].

Amitriptyline has an NNT of 2.2 for PHN, but the AEs, lack of recommendations for use in the elderly, and small sample sizes of studies have relegated this agent behind antiepileptics for certain PHN cases [30, 42, 50, 51]. Nortriptyline, maprotiline, and desipramine have also proven effective, but less so than amitriptyline.

3.3.2. Nortriptyline

A TCA used for short-term treatment of various forms of depression, nortriptyline blocks the noradrenaline presynaptic receptors, thereby blocking reuptake of this neurotransmitter and raising concentrations in the synaptic cleft in the CNS. Nortriptyline also binds to α-adrenergic, histaminergic and cholinergic receptors. Long-term treatment with nortriptyline produces downregulation of adrenergic receptors due to increased stimulation of these receptors [41].

3.4. Selective serotonin and noradrenaline reuptake inhibitors

Patients who are unable to tolerate TCAs may do better with selective serotonin and noradrenaline reuptake inhibitors, such as duloxetine or venlafaxine. Although less effective than TCAs, the selective 5-HT and noradrenaline reuptake inhibitors (SSNRIs) offer efficacy for both pain and depression with fewer AEs. SSRIs effectively relieve depression symptoms but they do not specifically relieve neuropathic pain. Nevertheless, some patients with chronic pain due to PHN will experience clinical depression, and the use of SSRIs can be useful for the management of depressive symptoms [5].

SNRIs are safer and easier to use than TCAs and represent a better option for patients with heart disease. The mechanism of SNRI analgesia is likely associated with activation of the descending inhibitory system of pain.

3.4.1. Paroxetine hydrochloride

The hydrochloride salt form of paroxetine is a phenylpiperidine derivative and a SSRI with antidepressant and anxiolytic properties. Paroxetine binds to the pre-synaptic serotonin transporter complex, resulting in negative allosteric modulation of the complex and thereby blocking reuptake of serotonin by the pre-synaptic transporter. Inhibition of serotonin recycling enhances serotonergic function through serotonin accumulation in the synaptic cleft, resulting in long-term desensitization and downregulation of 5-HT receptors and leading to symptomatic relief of depressive illness [35].

3.4.2. Duloxetine hydrochloride

The hydrochloride salt of duloxetine is a fluoxetine derivative belonging to the class of SSNRIs and exhibiting antidepressant activity. Duloxetine selectively prevents reuptake of 5-HT and noradrenaline via transporter complexes on the pre-synaptic membrane, thereby increasing the levels of these neurotransmitters within the synaptic cleft. As a result, this agent potentiates serotonergic and noradrenergic activities in the CNS, alleviating depression and neuropathic sensations such as pain and tingling. Furthermore, duloxetine does not show significant affinity for dopaminergic, adrenergic, cholinergic, histaminergic, opioid, glutamatergic, or GABAergic receptors [35].

Duloxetine is recommended for painful diabetic neuropathies [11, 29, 30], but not for PHN; little clinical trial data has been accumulated supporting use in PHN. SNRIs are also associated with a number of systemic AEs, including nausea, vomiting, somnolence, anorexia, constipation, dizziness, fatigue, insomnia, increased sweating, and dry mouth [52-54].

3.5. Other oral therapies

Several other oral agents have been investigated in the treatment of PHN. Specifically, the NMDA antagonists dextromethorphan and memantine [55, 56] and the benzodiazepine lorazepam [56] have not shown good efficacy in clinical trials.

In treatment guidelines, therapies described as lacking sufficient evidence of efficacy include anticonvulsants that target sodium channels (carbamazepine and oxcarbazepine), other anticonvulsants (lamotrigine, topiramate, and valproic acid), other antidepressants (bupropion, citalopram, paroxetine), and the oral lidocaine analog, mexiletine [11, 30, 31]. For other neuropathic pain syndromes, mexiletine has only shown efficacy at doses that produce systemic AEs, including proarrhythmic effects [43]. This suggests that the benefits of sodium channel antagonism for PHN may be better achieved via local lidocaine administration.

JSPC guidelines also recommend extract of cutaneous tissue from rabbits inoculated with vaccinia virus (Neurotropin®) as a first-line oral therapy[32].

3.5.1. Carbamazepine

Carbamazepine is a tricyclic compound chemically related to TCAs with anticonvulsant and analgesic properties. This agent exerts anticonvulsant activity by reducing polysynaptic responses and blocking post-tetanic potentiation. The analgesic activity is not understood, but carbamazepine is commonly used to treat pain associated with trigeminal neuralgia [35].

3.5.2. Valproic acid

Valproic acid is a synthetic derivative of propylpentanoic acid with antiepileptic properties and potential antineoplastic and antiangiogenesis activities. In epilepsy, valproic acid appears to act by increasing concentrations of GABA in the brain. The antitumor and antiangiogenesis activities of this agent may be related to the inhibition of histone deacetylases and nitric oxide synthase, resulting in the inhibition of nitric oxide synthesis [35].

3.5.3. Extract of cutaneous tissue from rabbits inoculated with vaccinia virus

Extract of cutaneous tissue from rabbits inoculated with vaccinia virus (Neurotropin®) has been shown to exert analgesic effects in a Japanese clinical study in PHN as one type of neuropathic pain. Producing no serious AEs, the drug product was very well tolerated, in addition to conferring analgesic effects. The drug product has been clinically used in patients 20 years old and older and is very safe [32, 57, 58].

3.5.4. Antiarrhythmics (mexiletine)

Mexiletine hydrochloride is a class 1B antiarrhythmic, and works by blocking sodium channels. A local anesthetic and antiarrhythmic (Class IB) agent structurally related to lidocaine, mexiletine exerts antiarrhythmic effects by inhibiting the inward sodium current in cardiac cells, thus reducing the rate of increase in the cardiac action potential (phase 0) and decreasing automaticity in Purkinje fibers. This slows nerve impulses in the heart and stabilizes the heartbeat. The anesthetic activity is due to the ability of mexiletine to block sodium influx in peripheral nerves, thereby reducing the rate and intensity of pain impulses reaching the CNS [41].

3.5.5. Lidocaine patch 5%

Lidocaine relieves pain through non-specific blockage of sodium channels on ectopic peripheral afferent fibers without causing numbness of the treated skin. Topical application without a relevant systemic absorption offers a good benefit-to-risk ratio only with local AEs, such as erythema or rash [9].

The transdermal patch contains a 5% aqueous base solution of the synthetic amide-type anesthetic lidocaine with analgesic activity. Upon topical application and transdermal delivery, the active ingredient lidocaine binds to and blocks voltage-gated sodium channels in the neuronal cell membrane; lidocaine-mediated stabilization of neuronal membranes inhibits the initiation and conduction of nerve impulses and produces reversible local anesthesia [34]. The lidocaine 5% patch is recommended as first-line therapy in the AAN [31], NeuPSIG [30], and EFNS [11] guidelines [29].

3.6. Combination therapy

In clinical practice, a combination of two or more drugs is often needed to achieve satisfactory pain relief, although few trials have been conducted to support this clinical observation. However, combination therapy with gabapentin or pregabalin and extended-release morphine in patients with PHN achieved higher pain relief with lower doses compared to administration of one drug alone. These results have also been confirmed for the combination of nortriptyline and gabapentin as well as for pregabalin and topical lidocaine in patients with PHN. Taken together, these results substantiate the usefulness of combination therapies in patients with PHN [9].

Advertisement

4. Conclusion

Neither obvious pathogenesis nor an established treatment have been clarified or established for PHN. Our study indicated that advanced age and deep pain at the first visit were shown to be predictive factors for PHN. DM and pain reduced by bathing should also be considered as potential predictors of PHN. We further reviewed recent pharmacotherapeutic results for PHN. To improve patient safety and the likelihood of achieving satisfactory outcomes, prospective studies are needed to establish optimal treatments including pharmacotherapy for PHN.

References

  1. 1. Gilden D. Efficacy of live zoster vaccine in preventing zoster and postherpetic neuralgia J Intern Med 2011; 269: 496-506.
  2. 2. Li Q, Chen N, Yang J, Zhou M, Zhou D, Zhang Q, He L. Antiviral treatment for preventing postherpetic neuralgia. Cochrane Database Syst Rev 2009; 2. CD006866.
  3. 3. Schmader KE. Epidemiology and impact on quality of life of postherpetic neuralgia and painful diabetic neuropathy. Clin J Pain 2002; 18: 350-354.
  4. 4. Dworkin RH, Portenoy RK. Pain and its persistence in herpes zoster. Pain 1996; 67: 241-251.
  5. 5. Sampathkumar P, Drage LA, Martin DP. Herpes zoster (shingles) and postherpetic neuralgia. Mayo Clin Proc. 2009; 84: 274-280.
  6. 6. Johnson RW. Herpes zoster and postherpetic neuralgia. Expert Rev Vaccines 2010; 9: 21-26.
  7. 7. McElhaney JE. Herpes zoster: a common disease that can have a devastating impact on patients' quality of life. Expert Rev Vaccines 2010; 9: 27-30.
  8. 8. Johnson RW, McElhaney J. Postherpetic neuralgia in the elderly. J Int J Clin Pract 2009; 63: 1386-1391.
  9. 9. Baron R, Binder A, Wasner G. Neuropathic pain: diagnosis, pathophysiological mechanisms, and treatment. Lancet Neurol 2010; 9: 807-819.
  10. 10. Whitley RJ, Volpi A, McKendrick M, Wijck A, Oaklander AL. Management of herpes zoster and post-herpetic neuralgia now and in the future. J Clin Virol 2010; 48:S20-S28.
  11. 11. Attal N, Cruccu G, Baron R, Haanpää M, Hansson P, Jensen TS, Nurmikko T; European Federation of Neurological Societies. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. Eur J Neurol 2010; 17: 1113-e88.
  12. 12. Doth AH, Hansson PT, Jensen MP, Taylor RS. The burden of neuropathic pain: a systematic review and meta-analysis of health utilities. Pain 2010; 149: 338-344.
  13. 13. Jung BF, Johnson RW, Griffin DR, Dworkin RH. Risk factors for postherpetic neuralgia in patients with herpes zoster. Neurology 2004; 62: 1545-1551.
  14. 14. Dworkin RH, Nagasako EM, Johnson RW, Griffin DR. Acute pain in herpes zoster: the famciclovir database project. Pain 2001; 94: 113-119.
  15. 15. Kurokawa I, Kumano K, Murakawa K. Hyogo Prefectural PHN Study Group. Clinical correlates of prolonged pain in Japanese patients with acute herpes zoster. J Int Med Res 2002; 30: 56-65.
  16. 16. Scott FT, Leedham-Green ME, Barrett-Muir WY, Hawrami K, Gallagher WJ, Johnson R, Breuer J. A study of shingles and the development of postherpetic neuralgia in East London. J Med Virol 2003; 70: S24-S30.
  17. 17. Nithyanandam S, Dabir S, Stephen J, Joseph M. Eruption severity and characteristics in herpes zoster ophthalmicus: correlation with visual outcome, ocular complications, and postherpetic neuralgia. Int J Dermatol 2009; 48: 484-487.
  18. 18. Opstelten W, Zuithoff NP, van Essen GA, van Loon AM, van Wijck AJ, Kalkman CJ, Verheij TJ, Moons KG. Predicting postherpetic neuralgia in elderly primary care patients with herpes zoster: prospective prognostic study. Pain 2007; 132: S52-S59.
  19. 19. Parruti G, Tontodonati M, Rebuzzi C, Polilli E, Sozio F, Consorte A, Agostinone A, Di Masi F, Congedo G, D'Antonio D, Granchelli C, D'Amario C, Carunchio C, Pippa L, Manzoli L, Volpi A; VZV Pain Study Group. Predictors of pain intensity and persistence in a prospective Italian cohort of patients with herpes zoster: relevance of smoking, trauma and antiviral therapy. BMC Med 2010; 8: 58.
  20. 20. Volpi A, Gatti A, Pica F, Bellino S, Marsella LT, Sabato AF. Clinical and psychosocial correlates of post-herpetic neuralgia. J Med Virol 2008; 80: 1646-1652.
  21. 21. McKendrick MW, Ogan P, Care CC. A 9 year follow up of post herpetic neuralgia and predisposing factors in elderly patients following herpes zoster. J Infect 2009; 59: 416-420.
  22. 22. Drolet M, Brisson M, Schmader K, Levin M, Johnson R, Oxman M, Patrick D, Camden S, Mansi JA. Predictors of postherpetic neuralgia among patients with herpes zoster: a prospective study. J Pain 2010; 11: 1211-1221.
  23. 23. Mick G, Gallais JL, Simon F, Pinchinat S, Bloch K, Beillat M, Serradell L, Derrough T. Burden of herpes zoster and postherpetic neuralgia: Incidence, proportion, and associated costs in the French population aged 50 or over. Rev Epidemiol Sante Publique 2010; 58: 393-401.
  24. 24. Kanbayashi Y, Onishi K, Fukazawa K, Okamoto K, Ueno H, Takagi T, Hosokawa T. Predictive Factors for Postherpetic Neuralgia Using Ordered Logistic Regression Analysis. Clin J Pain 2011, in press.
  25. 25. Johnson RW. Zoster-associated pain: what is known, who is at risk and how can it be managed? Herpes 2007; 14: 30-34.
  26. 26. Wu CL, Raja SN. An update on the treatment of postherpetic neuralgia. J Pain 2008; 9: S19-30.
  27. 27. Hempenstall K, Nurmikko TJ, Johnson RW, A’Hern RP, Rice AS. Analgesic therapy in postherpetic neuralgia: a quantitative systematic review. PLoS Med 2005; 2: e164.
  28. 28. Benzon HT, Chekka K, Darnule A, Chung B, Wille O, Malik K. Evidence-based case report: the prevention and management of postherpetic neuralgia with emphasis on interventional procedures. Reg Anesth Pain Med 2009; 34: 514-21.
  29. 29. Argoff CE. Review of current guidelines on the care of postherpetic neuralgia. Postgrad Med 2011; 123: 134-142.
  30. 30. Dworkin RH, O'Connor AB, Backonja M, Farrar JT, Finnerup NB, Jensen TS, Kalso EA, Loeser JD, Miaskowski C, Nurmikko TJ, Portenoy RK, Rice AS, Stacey BR, Treede RD, Turk DC, Wallace MS. Pharmacologic management of neuropathic pain: evidence-based recommendations. Pain 2007; 132: 237-251.
  31. 31. Dubinsky RM, Kabbani H, El-Chami Z, Boutwell C, Ali H; Quality Standards Subcommittee of the American Academy of Neurology. Practice parameter: treatment of postherpetic neuralgia: an evidence-based report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2004; 63: 959-965.
  32. 32. The Committee for the Guidelines for the Pharmacologic Management of Neuropathic Pain of JSPC. Guidelines for the Pharmacologic Management of Neuropathic Pain. Tokyo: Shinko Trading; 2011.
  33. 33. Moore RA, Wiffen PJ, Derry S, McQuay HJ. Gabapentin for chronic neuropathic pain and fibromyalgia in adults. Cochrane Database Syst Rev 2011; 3: CD007938.
  34. 34. Moore RA, Straube S, Wiffen PJ, Derry S, McQuay HJ. Pregabalin for acute and chronic pain in adults. Cochrane Database Syst Rev 2009; 3: CD007076.
  35. 35. National Cancer Institute. National Cancer Institute of the National Institutes of Health (NIH). Cancer topics, In: Cancer Drug Information. 2012 http://www.cancer.gov/drugdictionary (accessed 12 August).
  36. 36. Freeman R, Durso-Decruz E, Emir B. Efficacy, safety, and tolerability of pregabalin treatment for painful diabetic peripheral neuropathy: findings from seven randomized, controlled trials across a range of doses. Diabetes Care 2008; 31: 1448-1454.
  37. 37. Cappuzzo KA. Treatment of postherpetic neuralgia: focus on pregabalin. Clin Interv Aging 2009; 4: 17-23.
  38. 38. Watson CP, Babul N. Efficacy of oxycodone in neuropathic pain: a randomized trial in postherpetic neuralgia. Neurology 1998; 50: 1837-1841.
  39. 39. Raja SN, Haythornthwaite JA, Pappagallo M, Clark MR, Travison TG, Sabeen S, Royall RM, Max MB. Opioids versus antidepressants in postherpetic neuralgia: a randomized, placebo-controlled trial. Neurology 2002; 59: 1015-1021.
  40. 40. Boureau F, Legallicier P, Kabir-Ahmadi M. Tramadol in post-herpetic neuralgia: a randomized, double-blind, placebo-controlled trial. Pain 2003; 104: 323–331.
  41. 41. National Cancer Institute. National Cancer Institute of the National Institutes of Health (NIH). NCI Thesaurus. 2012 http://ncit.nci.nih.gov/ncitbrowser/pages/home.jsf (accessed 12 August).
  42. 42. Saarto T, Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Database Syst Rev 2007; 4: CD005454.
  43. 43. Finnerup NB, Otto M, McQuay HJ, Jensen TS, Sindrup SH. Algorithm for neuropathic pain treatment: an evidence based proposal. Pain 2005; 118: 289-305.
  44. 44. Sindrup SH, Otto M, Finnerup NB, Jensen TS. Antidepressants in the treatment of neuropathic pain. Basic Clin Pharmacol Toxicol 2005; 96: 399-409.
  45. 45. Cohen HW, Gibson G, Alderman MH. Excess risk of myocardial infarction in patients treated with antidepressant medications: association with use of tricyclic agents. Am J Med 2000; 108: 2-8.
  46. 46. Ray WA, Meredith S, Thapa PB, Hall K, Murray KT. Cyclic antidepressants and the risk of sudden cardiac death. Clin Pharmacol Ther 2004; 75: 234-241.
  47. 47. Tata LJ, West J, Smith C, Farrington P, Card T, Smeeth L, Hubbard R. General population based study of the impact of tricyclic and selective serotonin reuptake inhibitor antidepressants on the risk of acute myocardial infarction. Heart 2005; 91: 465-471.
  48. 48. Max MB, Lynch SA, Muir J, Shoaf SE, Smoller B, Dubner R. Effects of desipramine, amitriptyline, and fluoxetine on pain in diabetic neuropathy. N Engl J Med 1992; 326: 1250-1256.
  49. 49. Watson CP, Vernich L, Chipman M, Reed K. Nortriptyline versus amitriptyline in postherpetic neuralgia: a randomized trial. Neurology 1998; 51: 1166-1171.
  50. 50. Watson CP, Evans RJ, Reed K, Merskey H, Goldsmith I, Warsh J. Amitriptyline versus placebo in postherpetic neuralgia. Neurology 1982; 32: 671-673.
  51. 51. Max MB, Schafer SC, Culnane M, Smoller B, Dubner R, Gracely RH. Amitriptyline, but not lorazepam, relieves postherpetic neuralgia. Neurology 1988; 38: 1427-1432.
  52. 52. Lunn MP, Hughes RA, Wiffen PJ. Duloxetine for treating painful neuropathy or chronic pain. Cochrane Database Syst Rev 2009; 4: CD007115.
  53. 53. Raskin J, Smith TR, Wong K, Pritchett YL, D'Souza DN, Iyengar S, Wernicke JF. Duloxetine versus routine care in the long-term management of diabetic peripheral neuropathic pain. J Palliat Med 2006; 9: 29-40.
  54. 54. Wernicke JF, Pritchett YL, D'Souza DN, Waninger A, Tran P, Iyengar S, Raskin J. A randomized controlled trial of duloxetine in diabetic peripheral neuropathic pain. Neurology 2006; 67: 1411-1420.
  55. 55. Sang CN, Booher S, Gilron I, Parada S, Max MB. Dextromethorphan and memantine in painful diabetic neuropathy and postherpetic neuralgia: efficacy and dose-response trials. Anesthesiology 2002; 96: 1053-1061.
  56. 56. Wallace MS, Rowbotham MC, Katz NP, Dworkin RH, Dotson RM, Galer BS, Rauck RL, Backonja MM, Quessy SN, Meisner PD. A randomized, double-blind, placebo-controlled trial of a glycine antagonist in neuropathic pain. Neurology 2002; 59: 1694-1700.
  57. 57. Kawashiri T, Egashira N, Watanabe H, Ikegami Y, Hirakawa S, Mihara Y, Yano T, Ikesue H, Oishi R. Prevention of oxaliplatin-induced mechanical allodynia and neurodegeneration by neurotropin in the rat model. Eur J Pain. 2011; 15:344-50.
  58. 58. Kudo T, Kushikata T, Kudo M, Kudo T, Hirota K. Antinociceptive effects of neurotropin in a rat model of central neuropathic pain: DSP-4 induced noradrenergic lesion. Neurosci Lett. 2011; 503:20-2.

Written By

Yuko Kanbayashi and Toyoshi Hosokawa

Submitted: 11 May 2012 Published: 27 March 2013