Implications and mechanism of action of gabapentin in ...€¦ · REVIEW Implications and mechanism...

15
REVIEW Implications and mechanism of action of gabapentin in neuropathic pain Ankesh Kukkar Anjana Bali Nirmal Singh Amteshwar Singh Jaggi Received: 20 September 2012 / Accepted: 14 December 2012 / Published online: 24 February 2013 Ó The Pharmaceutical Society of Korea 2013 Abstract Gabapentin is an anti-epileptic agent but now it is also recommended as first line agent in neuropathic pain, particularly in diabetic neuropathy and post herpetic neu- ralgia. a2d-1, an auxillary subunit of voltage gated calcium channels, has been documented as its main target and its specific binding to this subunit is described to produce different actions responsible for pain attenuation. The binding to a2d-1 subunits inhibits nerve injury-induced trafficking of a1 pore forming units of calcium channels (particularly N-type) from cytoplasm to plasma membrane (membrane trafficking) of pre-synaptic terminals of dorsal root ganglion (DRG) neurons and dorsal horn neurons. Furthermore, the axoplasmic transport of a2d-1 subunits from DRG to dorsal horns neurons in the form of antero- grade trafficking is also inhibited in response to gabapentin administration. Gabapentin has also been shown to induce modulate other targets including transient receptor poten- tial channels, NMDA receptors, protein kinase C and inflammatory cytokines. It may also act on supra-spinal region to stimulate noradrenaline mediated descending inhibition, which contributes to its anti-hypersensitivity action in neuropathic pain. Keywords Gabapentin Á Neuropathic pain Á Diabetic neuropathy Á Post herpetic neuralgia Á Dorsal root ganglion Á Descending inhibition Introduction Pain arising as direct consequence of a lesion on a nerve/ disease affecting the somato-sensory system, either at the peripheral or central nervous system, is described as neu- ropathic pain (Geber et al. 2009). Following peripheral nerve injury, a cascade of events in the primary afferents leads to peripheral sensitization which is characterized by spontaneous nociceptor activity, decreased threshold (allodynia) and increased response to supra-threshold stimuli (hyperalgesia). A series of molecular changes in the spinal cord and the different brain centres is associated with central sensitization which is responsible for the pain to non-injured extra-territory regions (extraterritorial pain) and contra-lateral parts (mirror-image pain) (Jaggi and Singh 2011). There are various conditions associated with neuro- pathic pain like diabetic neuropathy, post herpetic neural- gia (PHN), cancer, trigeminal neuralgia etc. Diabetic neuropathic pain is one of the long term complications of diabetes mellitus and, both metabolic and ischemic mech- anisms have a role in diabetic neuropathies (Said 2007). The patients with Herpes zoster infection usually experi- ence pain, however, some patients experience pain beyond the typical 4-week duration. About 10 % patients develop the distressing complication of PHN with complex patho- physiology and involve both the peripheral as well as central processes (Argoff 2011). Trigeminal neuralgia is defined as sudden, usually unilateral, severe, brief, stabbing recurrent episodes of pain within one or more branches of the trigeminal nerve and it results from abnormalities in trigeminal afferent neurons in the trigeminal root or gan- glion (Zakrzewska and McMillan 2011). Neuropathic cancer pain, commonly encountered in clinical practice, may also be cancer-related due to tumor invasion in the A. Kukkar Á A. Bali Á N. Singh Á A. S. Jaggi (&) Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, Punjab, India e-mail: [email protected] 123 Arch. Pharm. Res. (2013) 36:237–251 DOI 10.1007/s12272-013-0057-y

Transcript of Implications and mechanism of action of gabapentin in ...€¦ · REVIEW Implications and mechanism...

REVIEW

Implications and mechanism of action of gabapentinin neuropathic pain

Ankesh Kukkar • Anjana Bali • Nirmal Singh •

Amteshwar Singh Jaggi

Received: 20 September 2012 / Accepted: 14 December 2012 / Published online: 24 February 2013

� The Pharmaceutical Society of Korea 2013

Abstract Gabapentin is an anti-epileptic agent but now it

is also recommended as first line agent in neuropathic pain,

particularly in diabetic neuropathy and post herpetic neu-

ralgia. a2d-1, an auxillary subunit of voltage gated calcium

channels, has been documented as its main target and its

specific binding to this subunit is described to produce

different actions responsible for pain attenuation. The

binding to a2d-1 subunits inhibits nerve injury-induced

trafficking of a1 pore forming units of calcium channels

(particularly N-type) from cytoplasm to plasma membrane

(membrane trafficking) of pre-synaptic terminals of dorsal

root ganglion (DRG) neurons and dorsal horn neurons.

Furthermore, the axoplasmic transport of a2d-1 subunits

from DRG to dorsal horns neurons in the form of antero-

grade trafficking is also inhibited in response to gabapentin

administration. Gabapentin has also been shown to induce

modulate other targets including transient receptor poten-

tial channels, NMDA receptors, protein kinase C and

inflammatory cytokines. It may also act on supra-spinal

region to stimulate noradrenaline mediated descending

inhibition, which contributes to its anti-hypersensitivity

action in neuropathic pain.

Keywords Gabapentin � Neuropathic pain �Diabetic neuropathy � Post herpetic neuralgia �Dorsal root ganglion � Descending inhibition

Introduction

Pain arising as direct consequence of a lesion on a nerve/

disease affecting the somato-sensory system, either at the

peripheral or central nervous system, is described as neu-

ropathic pain (Geber et al. 2009). Following peripheral

nerve injury, a cascade of events in the primary afferents

leads to peripheral sensitization which is characterized by

spontaneous nociceptor activity, decreased threshold

(allodynia) and increased response to supra-threshold

stimuli (hyperalgesia). A series of molecular changes in the

spinal cord and the different brain centres is associated

with central sensitization which is responsible for the pain

to non-injured extra-territory regions (extraterritorial pain)

and contra-lateral parts (mirror-image pain) (Jaggi and

Singh 2011).

There are various conditions associated with neuro-

pathic pain like diabetic neuropathy, post herpetic neural-

gia (PHN), cancer, trigeminal neuralgia etc. Diabetic

neuropathic pain is one of the long term complications of

diabetes mellitus and, both metabolic and ischemic mech-

anisms have a role in diabetic neuropathies (Said 2007).

The patients with Herpes zoster infection usually experi-

ence pain, however, some patients experience pain beyond

the typical 4-week duration. About 10 % patients develop

the distressing complication of PHN with complex patho-

physiology and involve both the peripheral as well as

central processes (Argoff 2011). Trigeminal neuralgia is

defined as sudden, usually unilateral, severe, brief, stabbing

recurrent episodes of pain within one or more branches of

the trigeminal nerve and it results from abnormalities in

trigeminal afferent neurons in the trigeminal root or gan-

glion (Zakrzewska and McMillan 2011). Neuropathic

cancer pain, commonly encountered in clinical practice,

may also be cancer-related due to tumor invasion in the

A. Kukkar � A. Bali � N. Singh � A. S. Jaggi (&)

Department of Pharmaceutical Sciences and Drug Research,

Punjabi University, Patiala 147002, Punjab, India

e-mail: [email protected]

123

Arch. Pharm. Res. (2013) 36:237–251

DOI 10.1007/s12272-013-0057-y

nerves, surgical nerve damage during tumor removal,

radiation-induced nerve damage or chemotherapy-related

neuropathy (Vadalouca et al. 2012).

The recommended first-line treatments for neuropathic

pain include antidepressants (tricyclic antidepressants and

dual reuptake inhibitors of both serotonin and norepi-

nephrine), calcium channel a2d ligands (gabapentin and

pregabalin) and topical lidocaine. Opioid analgesics and

tramadol are generally recommended as second-line treat-

ments that may be considered for first-line use in selected

clinical circumstances. Other medications that are generally

used as third-line treatments (may be used as second-line

treatments in some circumstances) are other antiepileptic

and antidepressant medications, mexiletine, N-methyl-D-

aspartate receptor antagonists, and topical capsaicin (Dworkin

et al. 2007).

Gabapentin is a structural analogue of GABA (Fig. 1)

and has been approved for adjunctive treatment of patients

(12 years or older) with partial seizures (with/without

secondary generalization), mixed seizure disorders and

refractory partial seizures in children (Honarmand et al.

2011). However, recent studies have also documented its

efficacy in ameliorating different types of neuropathic pain

in preclinical as well as in clinical settings. Earlier, gaba-

pentin was considered as second line treatment with

tricyclic antidepressants (TCA) as drug of choice. How-

ever, in patients with a history of cardiovascular disorders,

glaucoma, and urine retention, gabapentinoid drugs have

emerged as first-line treatment for neuropathic pain. In

addition, gabapentin has a more favourable safety profile

with minimal concerns regarding drug interactions and

showing no interference with hepatic enzymes, therefore, it

has been employed as first line agent in various neuropathic

conditions like diabetic neuropathy and PHN (Vranken

2009). More patients with neuropathic pain reported an

improvement with pregabalin (a2d ligands) (33 %) than

duloxetine (21 %). Duloxetine (38 %) had a higher fre-

quency of side effects compared to pregabalin (30 %)

(Mittal et al. 2011). In patients with spinal cord injury,

gabapentin has been shown to be more effective for pain

relief than amitriptyline (Selph et al. 2011). The present

review discusses the effectiveness of gabapentin in differ-

ent types of neuropathic pain in preclinical as well in

clinical settings and also discusses the possible mechanism

of action at different levels including at dorsal root

ganglion (DRG) and dorsal horn neurons along with at

supra-spinal centres.

Gabapentin in preclinical studies

There have been number of preclinical studies document-

ing the beneficial effects of gabapentin in different models

of neuropathic pain. Back and co-workers demonstrated

that intraperitoneal (i.p.) injection of gabapentin at differ-

ent doses (30, 100, 300 mg/kg) significantly alleviates

mechanical, warm and cold allodynia in partial tail nerve

injury-induced neuropathic pain in a dose-dependent

manner (Back et al. 2004). The systemic (30–60 mg/kg i.p.)

as well as the spinal (10–20 lg) administration of gabapentin

in the adult rats has been shown to attenuate resiniferotoxin

(a potent TRPV1 agonist)-induced long-lasting changes in

mechanical and thermal sensitivities in a model of PHN

(Chen and Pan 2005). Walczak and co-workers demon-

strated the efficacy of gabapentin (50 mg/kg i.p.) in attenu-

ating cold as well as mechanical allodynia, but not

hyperalgesia, in a saphenous partial ligation (unilateral

partial injury to the saphenous nerve) rodent model of neu-

ropathic pain (Walczak et al. 2005). In chronic constriction

injury model of neuropathic pain, administration of gaba-

pentin (30, 100 and 300 mg/kg i.p.) every 12 h for 4 days has

been shown to reduce the development of hyperalgesia in a

rats (Coderre et al. 2007).

The systemic administration of gabapentin (i.p.) has

been shown to increase the paw withdrawal latency and

produce anti-allodynic effects in a mice model of partial

sciatic nerve ligation of neuropathic pain in a dose

dependent manner (Kusunose et al. 2010). A recent study

has shown the effectiveness of gabapentin (5 or 50 mg/kg, i.p.)

in attenuating neuropathic pain behavior in forelimb neuro-

pathic pain model (due to partial injury to medial and

ulner nerves) in a dose-dependent manner (Yi et al. 2011). An

intra-thecal administration of gabapentin at the dose

of 1.05 lmol/day for 14 days in a chronic constriction

injury model and at the dose of 20 lg/h for 7 days in spinal

nerve ligation model has also been shown to attenuate

OH

NH2

O

GABA

NH2

O

OH

Gabapentin

Fig. 1 Structures of GABA and gabapentin

238 A. Kukkar et al.

123

neuropathic pain in an effective manner in rats (Chu et al.

2011; Yeh et al. 2011)

Furthermore, the effectiveness of gabapentin in attenu-

ating pain behavior has also been described in chemother-

apeutic agents and viruses-induced peripheral neuropathic

pain. Xiao and co-workers demonstrated that systemic

multiple dosing of gabapentin (100 mg/kg i.p.) signifi-

cantly reduced paclitaxel- and vincristine-evoked mec-

hano-allodynia and mechano-hyperalgesia (Xiao et al.

2007). The systemic treatment with gabapentin has also

been shown to attenuate ddC (Zalcitabine), an anti-retro-

viral agent, and HIV-gp120, (delivered to the rat sciatic

nerve) (gp120 ? ddC)-induced neuropathic pain (Wallace

et al. 2007). Moreover, gabapentin (30 mg/kg i.p.) also has

also been shown to reverse Varicella Zoster virus-induced

increase in mechanical hypersensitivity (s.c. injection of

Varicella Zoster virus into the glabrous footpad of the hind

limb), a model which resembles Herpes Zoster virus-

induced clinical pain (Hasnie et al. 2007). Oral adminis-

tration of gabapentin (50 mg/kg twice a day for 5 days) is

reported to attenuate mechanical allodynia in a model of

diabetic neuropathy (Wodarski et al. 2009).

Apart from producing the direct beneficial effects in

neuropathic pain, the studies have also shown that it may

increase the effectiveness of co-administered drugs and

may result in synergistic effect. Gabapentin shows syner-

gistic effect with anti-depressant venlafaxine in treating

neuropathic pain. This combination was found to be

superior in comparison to gabapentin alone in the rat

spared nerve injury (SNI) model of neuropathic pain (Garry

et al. 2005). The studies have shown that co-administration

of gabapentin with benfotiamine or cyanocobalamin in a

fixed ratio markedly reduces spinal nerve ligation-induced

tactile allodynia, showing a synergistic interaction between

anticonvulsants and B vitamins. Oral administration of

gabapentin (15–300 mg/kg), benfotiamine (30–600 mg/kg)

or cyanocobalamin (0.3–6.0 mg/kg) has been shown to

significantly reduce neuropathic pain in rats (Mixcoatl-

Zecuatl et al. 2008). Co-administration of duloxetine with

gabapentin/donepezil (p.o.) is also shown to exhibit syn-

ergistic effect against spinal nerve ligation-induced neu-

ropathic pain. The combination of all three drugs was also

shown to produce synergistic action (Hayashida and

Eisenach 2008). The other studies have also demonstrated

that use of donepezil as an adjunctive to gabapentin

improves the therapeutic outcome in the management of

neuropathic pain in spared nerve injury model. Co-

administration of donepezil (0.5 mg/kg s.c.) and low doses

of gabapentin (10 and 30 mg/kg s.c.), both single dosing,

resulted in a three- to fourfold increase of the analgesic

effect, in comparison with gabapentin administered alone.

Addition of donepezil (1.5 mg/kg p.o.) from day 11 to day

20 to gabapentin (25 mg/kg p.o., once daily over 20 days)

treatment regimen was shown to improve analgesic effects

as compared to gabapentin monotherapy (Folkesson et al.

2010). In another study with same model, the intra-thecal

co-administration of gabapentin and clonidine at a ratio of

20:7 exerted a synergistic action on the mechanical anti-

allodynic effect (Yamama et al. 2010).

Gabapentin in clinical studies:

There have been number of clinical studies documenting

the beneficial effects of gabapentin in different types of

neuropathic pain like neuropathy due to cancer, HIV

infection, diabetic neuropathy, trigeminal neuralgia and

post-operative neuropathic pain (Table 1).

Cancer and chemotherapeutic agents-induced pain

Gabapentin monotherapy (300 mg/d to 1.8 g/d) has been

reported to beneficial and well tolerated in cancer as well

as chemotherapy-induced neuropathic pain (Ross et al.

2005). Administration of fixed low-dose of gabapentin

(800 mg/day) in anti-cancer drugs-induced neuropathic

pain is also reported to produce partial to complete

remission (Tsavaris et al. 2008). In randomized open

clinical trial, the combination of gabapentin with opioid

analgesics was shown to provide better relief in neuro-

pathic pain in cancer patients as compared to opioid

analgesics alone in terms of reduction in pain intensity for

burning and shooting pain at different days of the study.

Furthermore, the rate of side effects was also shown to be

comparatively less in combination therapy as compared to

opioid monotherapy (Keskinbora et al. 2007). Arai and

co-workers demonstrated that low dose gabapentin (200–

400 mg/12 h)-imipramine (10 mg/12 h) combination with

opioids was effective in managing neuropathic cancer pain

(in terms of total pain score and paroxysmal pain episodes)

without severe adverse effects (Arai et al. 2010). In con-

trast, Takahashi and co-workers demonstrated that combi-

nation of gabapentin and opioid analgesic was of minimal

clinical benefit in the study conducted on Japanese patients

with neuropathic cancer pain in an open-label and single-

center clinical trial (Takahashi and Shimoyama 2010).

Patarica-Huber demonstrated that intergroup difference

between three groups i.e., gabapentin, gabapentin-NSAID,

gabapentin-NSAID-morphine was not statistically signifi-

cant in breast cancer patients. Although during the 6-week

study the decrease of pain intensity was significant in all 3

groups, the correlation between the increase trend of side

effects and the frequency of additional medication was also

significant (Patarica-Huber et al. 2011).

Gabapentin in neuropathic pain 239

123

Trigeminal neuralgia

In treating trigeminal neuralgia, gabapentin has been

considered as second drug of choice (20 % patients) as

compared to carbamazepine, employed in 70 % of patients

as the first choice drug (Cheshire 2007; Hon and Fei 2008).

Pandey and co-workers demonstrated the successful man-

agement of idiopathic trigeminal neuralgia in patients

resistant to carbamazepine without untoward side-effects

(Pandey et al. 2008). Lemos and co-workers demonstrated

that the combination of gabapentin and ropivacain (applied

as analgesic block to trigeminal neuralgia trigger points) is

more effective and safe in trigeminal neuralgia patients

resistant to carbamazepine (Lemos et al. 2010).

Diabetic neuropathic pain

The most of the diabetic patients require pain control therapy

and the TCA drugs remain a first-line approach with gaba-

pentin as alternative to TCA. However due to predictable and

troublesome side effects associated with TCA, gabapentin is

the main therapy in case of elderly patients with diabetic

peripheral neuropathy (Haslam and Nurmikko 2008; Vranken

2009) Gabapentin is described as first line agent for the

treatment of diabetic neuropathic pain in the United Kingdom

and is generally better tolerated than TCA (Boulton 2003).

The various studies have indicated the usefulness of gaba-

pentin in treating diabetic neuropathy comparable to TCA.

A randomized, double-blind, placebo-controlled study demon-

strated that gabapentin monotherapy (dose titrated from 900

to 3,600 mg/d or maximum tolerated dosage) for 8 weeks

significantly lowered the pain and enhanced the quality of

life as compared to placebo (Backonja et al. 1998). Further-

more, in a randomized, double-blind, crossover, 6 week study,

no significant difference between gabapentin (900–1,800 mg/d)

and amitriptyline (25–75 mg/d) was reported (Morello et al.

1999). In a 12-week, open-label, prospective, randomized

trial gabapentin (1,200–2,400 mg/d) was shown to produce

greater improvement in pain and paresthesias associated

with diabetic neuropathy as compared to amitriptyline

(30 mg/d to 90 mg/d). Furthermore, gabapentin was shown

to be better tolerated than amitriptyline in diabetic neuro-

pathic pain patients (Dallocchio et al. 2000). The other

studies have also shown that gabapentin in doses of 1,800 to

3,600 mg/d is well tolerated, superior to placebo, and

equivalent to amitriptyline (Hemstreet and Lapointe 2001;

Backonja and Glanzman 2003; Paradowski and Bilinska

2003). In contrast to previous studies with indirect compar-

isons between TCA and gabapentin, Chou and co-workers

demonstrated the comparable effects of gabapentin and tri-

cyclic antidepressants for pain relief in patients with diabetic

neuropathy and PHN in a direct comparison study (Chou

et al. 2009). Besides peripheral neuropathic pain, diabetic

cardiac neuropathy also co-exists in diabetes patients and

studies have also demonstrated that therapeutic doses of

gabapentin not only alleviate neuropathic symptoms but also

improve cardiac autonomic function in diabetic patients with

peripheral neuropathy (Ermis et al. 2010).

The large placebo-controlled studies have also provided

the evidence of the efficacy of gabapentinoid group of

drugs (gabapentin and pregabalin) in diabetic pain. The

potential availability of less expensive generic formula-

tions of gabapentin, together with greater experience with

its use, favour gabapentin as the main antiepileptic drug for

alleviating diabetic neuropathy. Topiramate, lamotrigine,

sodium valproate and oxcarbazepine have been shown to

be effective in smaller studies but do not have the same

evidence base as the gabapentinoid group of drugs (Chong

and Hester 2007). Hanna and co-workers demonstrated that

co-administration of prolonged-release oxycodone and

existing gabapentin therapy (low dose) has a clinically

meaningful effect in painful diabetic neuropathy in a

Table 1 The summarized clinical implications of gabapentin in different forms of neuropathic pain

S. no. Types of pain Treatment schedule/general comments References

1. Cancer and chemotherapeutic

agents-induced pain

Gabapentin (800 mg/day) produce partial

to complete remission

Tsavaris et al. (2008),

Arai et al. (2010)

Low dose gabapentin (200–400 mg/day)

? imipramine (10 mg/12 h) ? opioids

alleviate pain with minimal side effects

2. Trigeminal neuralgia Gabapentin as second drug of choice; the

combination with ropivacain produces

significant effect in carbamazepine resistant cases

Lemos et al. (2010)

3. Diabetic neuropathic pain Main stay drug for elderly patients at a doses

of 1,800–3,600 mg/d gabapentin is

Vranken (2009), Paradowski

and Bilinska (2003)

Equivalent/better tolerated than TCA

with comparable/higher efficacy

4. Post-herpetic neuralgia Gabapentin is one of the first line treatment in PHN Jean et al. (2005),

Rice and Maton (2001)600 mg/day may be the safe and effective

starting dose with adequate relief at higher

doses (1,200–2,400 mg/day)

240 A. Kukkar et al.

123

randomized, double-blind, placebo-controlled study con-

ducted for 12 weeks and this combination provides better

pain relief as compared to gabapentin monotherapy (Hanna

et al. 2008).

Post-herpetic neuralgia (PHN)

Initially, TCA were the most commonly used agents for

treating PHN patients and were effective in a significant

proportion of patients. However, various adverse events

including anticholinergic and sedative effects limit treat-

ment. These side effects tend to be more acute in the

elderly, the population most likely to suffer from PHN and

over past few years gabapentin has received increasing

attention due to its safety profile (Beydoun 1999). In a

randomized, double-blind, parallel-group trial of 9 weeks

duration clinical study, Chandra and co-workers described

that gabapentin was equally efficacious and better tolerated

as compared to nortriptyline and may be considered a

suitable alternative for the treatment of PHN (Chandra

et al. 2006). On the contrary, O’connor and co-workers

demonstrated that desipramine (100 mg/day) is more

effective and less expensive than gabapentin (1,800 mg/day)

or pregabalin (450 mg/day) for the treatment of older

patients with PHN in whom it is not contraindicated

(O’Connor et al. 2007). At present, the first-line treatments

for PHN include TCA, gabapentin, pregabalin, topical

lidocaine patch and opioids, tramadol, capsaicin cream and

patch are recommended as either second- or third-line ther-

apies in different guidelines (Argoff 2011).

The multicenter, randomized, double-blind, placebo-

controlled, parallel design, 8-week trial evidenced that

gabapentin is effective in the treatment of pain and sleep

interference associated with PHN. The mood and quality

of life was also shown to be improved with gabapentin

therapy (Rowbotham et al. 1998). Another multi-centre

double blind, randomized, placebo controlled 7-week study

described the efficacy and safety of gabapentin (1,800 or

2,400 mg/day) in treating PHN patients (Rice and Maton

2001). Berger and co-workers reported that initiation of

gabapentin therapy in patients with PHN was associated

with a reduction in the use of opioid analgesics (Berger

et al. 2003). Furthermore, Gilron and co-workers demon-

strated that morpine-gabapentin combination is having

better analgesic effect as compared to monotherapy with

these drugs at maximal tolerated dose in a randomized,

double-blind, active placebo-controlled, four-period cross-

over trial, for 5 weeks (Gilron et al. 2005).

Although gabapentin has been shown to provide pain

relief in PHN patients at dosage of 1,200–2,400 mg/day,

Jean and co-workers demonstrated that 600 mg/day gaba-

pentin could be a safe and effective starting dose for PHN

patients (Jean et al. 2005). Due to circadian rhythm, the

concentrations of b-endorphin levels are significantly

higher in the morning as compared to the afternoon in both

human adults and neonates. It may be probably responsible

for the diurnal variation in neuropathic pain intensity,

however, the temporal profile appears to be unaffected by

treatment with gabapentin as it decreases the pain scores to

a similar degree at all study time points (Petraglia et al.

1983; Hindmarsh et al. 1989; Odrcich et al. 2006).

Side effects of gabapentin

The common adverse events experienced with the gaba-

pentin include dizziness (23.9 %), somnolence (27.4 %),

ataxia (7.1 %), peripheral edema (9.7 %) and confusion

(Backonja et al. 1998; Rowbotham et al. 1998). Jacob and

co-workers reported the development of asterixis (a nega-

tive myoclonus caused by sudden pauses of innervation for

more than 200 ms) in PHN patient treated with gabapentin

(Jacob 2000). Asterixis has been described to occur in

response to accumulation of endogenous benzodiazepine

receptor ligands that act on GABA-A receptors in the brain

(Butterworth 1996). The drugs such as phenobarbitone,

carbamazepine and valproate are known to produce aster-

ixis by enhancing GABA transmission (Bodensteiner et al.

1981). Accordingly, GABAergic mechanism in the form of

enhancement in GABA release (Taylor 1997) has been

described as the possible mechanism for the development

of asterixis in response to gabapentin therapy (Jacob 2000).

A case of cholestasis (jaundice, dark urine, pale stool,

fatigue, and epigastric tenderness) with serious hepato-

toxicity is also reported (Richardson et al. 2002). Parsons

and co-workers from 3 randomized, double-blind, placebo-

controlled, parallel-group studies of gabapentin in PHN

patients described that the safety concerns limit the titration

of gabapentin dosing (300 mg/d at the start to 1,800–

3,600 mg/d as maintenance dose) to achieve optimal effi-

cacy. The incidence of peripheral edema was reported to be

increased with an increase in the dose of gabapen-

tin C1,800 mg/d (7.5 % vs. 1.4 %) (Parsons et al. 2004).

Due to development of peripheral edema with the recom-

mended doses of gabapentin, The New York Heart Asso-

ciation has issued a warning about using caution while

prescribing these drugs to type III-IV heart failure patients

(Erdogan et al. 2011). Bookwalter and Gitlin reported a

case of a 75-year-old man with renal dysfunction who

developed neurologic toxicity due to gabapentin accumu-

lation (Bookwalter and Gitlin 2005). Gabapentin is not

metabolized in the liver and is mainly excreted through

kidney, accordingly, in patients with renal dysfunctions the

gabapentin levels tend to rise and produce severe side

effects. A patient was described to develop delusions of

parasitosis after been initiated with gabapentin treatment

Gabapentin in neuropathic pain 241

123

for neuropathic pain and complete disappearance of

symptoms was reported after discontinuation of the medi-

cation (Lopez et al. 2010). The patients tend to develop

dependence on gabapentin even after 3 weeks of admin-

istration and it is required to gradually taper the dose to

avoid withdrawal symptoms. See and co-workers have

reported the development of akthesia in response to gaba-

pentin withdrawal (at doses ranging from 400–8,000 mg/day)

(See et al. 2011). The randomized, double-blind, placebo-

controlled studies have provided the evidence that extended

release gabapentin shows improved bioavailability, tolera-

bility, convenience, along with minimized incidence of

adverse events in treating neuropathic pain (Sandercock et al.

2009; Backonja et al. 2011).

The studies have shown that gabapentin causes sexual

dysfunction including loss of libido, anejaculation, anor-

gasmia, and impotence at a minimum total daily dose of

900 mg. However, a recent case report has suggested the

dysfunction at a daily dose of only 300 mg (Kaufman and

Struck 2011). Gabapentin-associated anorgasmia is dose

dependent and has been shown to be more common in

older patients (Perloff et al. 2011). Calcium channels are

widely present in the central nervous system; therefore,

inhibition of these channels is likely to influence many

neural functions including sexual behavior. It has been

hypothesized that gabapentin mediated inhibition of cal-

cium currents may lead to alterations in the levels of

neurotransmitters release (particularly dopamine, which

promotes sexual desire, and serotonin, which inhibits sex-

uality) and attenuate post-synaptic excitability required to

maintain sexuality (Calabro 2011). The incidences of

severe myopathy and rhabdomyolysis have also been

reported with gabapentin therapy (Tuccori et al. 2007;

Bilgir et al. 2009). The exact mechanism of gabapentin-

induced myopathy is not known. It may be possible that

gabapentin by acting on voltage-gated calcium and sodium

channels in muscle cells may cause alterations in the

intracellular calcium/sodium balance to promote myopathy

(Alden and Garcia 2001; Liu et al. 2006; Tuccori et al.

2007). In a retrospective real world study, it was demon-

strated that switching from long-term treatment with alpha-

lipoic acid to gabapentin in painful diabetic neuropathy led

to higher rates of side effects, frequencies of outpatient

visits, and daily costs of treatment (Ruessmann 2009).

Amongst the gabapentinoid drugs, the various studies

have shown the superiority of pregabalin over the gaba-

pentin. Perez and co-workers demonstrated that pregabalin

administration is associated with greater reduction in mean

weekly intensity of pain as compared to gabapentin, with

no significant differences in cost (Perez et al. 2010). In a

direct comparison study, it was demonstrated that the

analgesic action of pregabalin in PHN was six times that of

gabapentin in terms of effectiveness in dosage conversion

(Ifuku et al. 2011). In an open, randomized, comparative

study suggested that the tramadol/acetaminophen combi-

nation treatment is as effective as gabapentin in the treat-

ment of painful diabetic neuropathy in patients with type 2

diabetes (Ko et al. 2010).

Mechanism of action

Gabapentin may produce pain attenuating effects by acting

on both the central nervous system (on the spinal and the

supra-spinal areas) and on the peripheral region (DRG

neurons) (Fig. 2). Accordingly, gabapentin has been found

useful in the spinal cord injury- induced pain as well as in

peripheral neuropathic pain (Attal et al. 2009). Gabapentin

acts peripherally to suppress the nociceptive afferent

stimuli from the injured DRG neurons (critical source of

triggering hyperalgesia and spontaneous pain) to the spinal

cord by reducing the sub-threshold membrane potential

oscillation (SMPO) (Yang et al. 2005). Gabapentin medi-

ated persistent inhibition of sodium current in chronically

compressed DRG neurons (A type) has been described to

produce inhibition of SMPO dependent repetitive firing and

bursting (Yang et al. 2009). Gabapentin was originally

designed as GABA mimetic with the intention that it would

be able to cross the blood–brain barrier and interact with

GABAergic systems to enhance GABA mediated inhibi-

tion. However, the studies have shown that it produces pain

attenuating effects by modulating other targets.

DRG and dorsal horn neurons

Voltage gated calcium channels

a2d-1 subunit a2d subunit of the voltage gated calcium

channels on the DRG neurons has been defined as the main

molecular target for gabapentin and amongst the different

types of a2d, a2d-1 has been the key binding target of

gabapentin (Jaggi and Singh 2011). a2d-1 is the extracel-

lular auxillary subunit of voltage gated calcium channels

particularly the N- and L-types, but not the T- types

(Davies et al. 2007). The strongest evidence that the a2d-1

subunit is the key target for gabapentinoid drugs has come

from the genetic studies. The knock-in replacement of the

wild-type a2d-1 subunit with a mutant (a2d-1 R217A)

incapable of binding pregabalin has been shown to result in

complete loss of the drug’s analgesic efficacy (Field et al.

2006). Although, there have also been some studies

reporting that gabapentin produces no effect on freshly

dissociated DRG neurons and inhibition of Ca2? channels

in this cell-type did not contribute to its mechanism of

action (Rock et al. 1993). Furthermore, several studies have

also reported that there is little/no acute inhibition of

242 A. Kukkar et al.

123

calcium currents either in neurons or in heterologous

expression systems (Li et al. 2006; Davies et al. 2007). The

disparity in results of heterologous expression system and

DRG neurons may be due to channel subunit heterogeneity

and the pathologic state of the tissue i.e., hyperalgesic or

normal. This point has been emphasized by a study

showing that gabapentin specifically inhibited Ca2? cur-

rents in mice over-expressing the a2d-1 subunits and did

not produce any effect in wild-type mice (Li et al. 2006).

The external application of gabapentin is not shown to

produce an acute effect on Ca2? channel current amplitude/

voltage dependent gating behavior. However, chronic

external exposure is shown to slow the rate of expression of

Ca2? channels indicating that gabapentin must be trans-

ported across the cell membrane to produce the inhibitory

effect (Mich and Horne 2008). It has also been shown that

inhibitory effects of gabapentin are eliminated by pre-

treatment with pertussis toxin suggesting involvement of G

protein in its inhibitory mechanism (Martin et al. 2002). In

contrast to an earlier study suggesting the time independent

effectiveness of gabapentin in a day (Odrcich et al. 2006),

Kusunose and co-workers reported the time-dependent

difference in the anti-allodynic effects of gabapentin and

attributed the difference in activity to the circadian oscil-

lation of calcium a2d-1 subunit expression in the DRG

(Kusunose et al. 2010).

Inhibition of membrane trafficking: Gabapentin binds to

the accessory a2d-1 subunits, not the major a1 pore-

forming unit, of the calcium channels and the major

function of a2d-1 subunits is to direct trafficking of pore

forming a1 subunits of Ca2? channels from the endoplas-

mic reticulum to the plasma membrane (membrane traf-

ficking) (Jarvis and Zamponi 2007). Earlier studies had

shown that the surface expression of Cav2.1 channels is

increased to 7-fold when a2d-1 is combined in Xenopus

laevis oocytes with a1 subunits (Gurnett et al. 1996).

Neuropathic pain, due to injuries to peripheral/central

nervous system, is associated with over-expression of the

a2d subunits of calcium channels (particularly N- types) in

the DRG neurons and the dorsal horn neurons of the spinal

cord. The binding of gabapentin to over-expressing a2d-1

proteins may be responsible for the down regulation of

Fig. 2 Proposed sites of action of gabapentin. (A) dorsal horn of

spinal cord; (B) Locus coeruleus (LC); (C) dorsal root ganglion

(DRG). Bold arrows indicate magnification, broken arrows show

inhibition and normal arrows show signal pathway. (A) Gabapentin

inhibits anterograde trafficking of a2d-1 subunits from DRG to spinal

cord dorsal horn and further inhibits receptor trafficking from cytosol

to cell membrane at pre synaptic site, which as a result inhibits release

of glutamate and substance P. It also inhibits the formation of Ca2?/

calmodulin-dependent protein kinase II (CaMKII). (B) Gabapentin

inhibits release of GABA pre synaptically which further increases

glutamate level which in turn causes release of NA in spinal cord

which stimulates descending inhibition. (C) Gabapentin blocks Na?

channel activity which further inhibits SMPO

Gabapentin in neuropathic pain 243

123

N- type calcium channels in the spinal cord and pain

attenuating effects in neuropathic pain (Luo et al. 2002;

Bauer et al. 2009; Taylor 2009; Boroujerdi et al. 2011).

Gabapentin acts mainly on a2d-1 isoform of a2d with

lower affinity for a2d-2 isoform and no effect on other two

isoforms of a2d. The arginine residue of a2d-1 at position

217 close to the VWA (Von willbrand domain A) domain

is critical for gabapentin binding and subsequent calcium

channel inhibition (Davies et al. 2007). VMA is present on

the extracellular sequence of all a2d subunits and contains

a perfect metal ion dependent adhesion site (MIDAS),

which is essential for the trafficking function of a2d. The

mutation within the VWA domains prevents the trafficking

of voltage gated calcium channels from cytoplasm to the

plasma membrane and decreases the Ca2? currents

(Whittaker and Hynes 2002; Cantı et al. 2005; Hoppa et al.

2012).

Inhibition of anterograde trafficking (axoplasmic

transport): Recently in spinal nerve ligation model, the protein

level of the a2d-1 subunit has been reported to be significantly

higher in the spinal dorsal horn and DRG on the injured side

(Morimoto et al. 2012). In contrast, the mRNA levels of the

a2d-1 subunit were shown to be selectively increased on the

injured side of L5 DRG, not in the dorsal horn, suggesting that

an increase in the a2d-1 subunit in the dorsal horn of the spinal

cord is secondary to increased a2d-1 levels in the DRG. Fur-

thermore, gabapentin was shown to suppress the elevated

protein levels of the a2d-1 subunits in the spinal dorsal horns

without affecting the mRNA levels of the a2d-1 subunit.

Therefore, it has been suggested that gabapentin inhibits the

anterograde (axonal) transport ofa2d-1 subunits from L5 DRG

to the primary afferent nerve terminals in the L5 dorsal horn

and normalizes the a2d-1 protein level in the spinal dorsal horn

by inhibiting the transport of the a2d-1 subunit (Morimoto

et al. 2012). Furthermore, the continuous administration of

gabapentin is reported to alleviate neuropathic pain for several

days after the termination of the administration indicating that

several days are required for the recovery of the transport of the

a2/d-1 subunit from the DRG to the primary afferent nerve

terminals (Morimoto et al. 2012). An earlier study has also

suggested that chronic administration of gabapentinoid drug

(pregabalin) attenuated nerve injury-induced increased a2d-1

in the pre-synaptic terminals of the dorsal horns and ascending

axon tracts, without affecting a2d-1 mRNA and protein in the

DRGs neurons. It was concluded that anti-allodynic effect of

pregabalin is associated with impaired anterograde trafficking

of a2d-1 from DRG neurons to the pre-synaptic terminals of

the dorsal horns resulting in reduced neurotransmitter release

and spinal sensitization (Bauer et al. 2009).

Inhibition of neurotransmitter release: Gabapentin

mediated decrease in the density of calcium channels in the

pre-synaptic terminals leads to decreased release of neu-

rotransmitters such as glutamate, CGRP and substance P

that are involved in neuropathic pain progression (Yaksh

2006; Quintero et al. 2011). The role of substance P in the

induction and the maintenance of neuropathic pain is well

defined (Cahill and Coderre 2002). It has been demon-

strated that gabapentin not only suppresses the release of

substance P but also decreases substance P induced-NFkB

activation which is an essential mediator of substance

P-induced cytokine synthesis. Therefore, gabapentin regu-

lates inflammation-related intracellular signaling in both

neuronal and glial cells that is effective in alleviating

symptoms of inflammatory and neuropathic pain (Park

et al. 2008). Gabapentin has also been shown to attenuate

paclitaxel and vinorelbine-induced substance P release

from cultured DRG neurons (Miyano et al. 2009).

Inhibition of inflammation: The a2d-1 dependent anal-

gesic actions of gabapentin in neuropathic pain have also

been correlated with inhibition of inflammatory cytokines

and inflammation. Wodarski and co-workers demonstrated

the effectiveness of gabapentin in attenuating allodynia in

STZ-induced diabetic neuropathic pain and correlated the

beneficial effect with decreased microglial activation (Iba-

1 as a marker) and reduced number of astrocytes (GFAP as

a marker) (Wodarski et al. 2009). Recently, it has been

reported that unilateral intra articular injection of CFA

[complete freund’s adjuvent] associated up-regulation of

a2d-1 subunit is associated with activation of microglia

and astrocytes and increased expression of CX3CL1 and

CX3CR1 in the spinal cord. CX3CL1 is defined as a

potential trigger to activate microglia and is co-localized

with a2d-1 subunits in the spinal dorsal horn. Adminis-

tration of gabapentin was shown to down-regulate the

spinal a2d-1 subunit expression and decrease CX3CL1

levels suggesting the critical role of CX3CL1 in gabapentin

mediated analgesic effects (Yang et al. 2012).

b subunits The trafficking of voltage gated Ca2? channel

from cytoplasm to plasma membrane and Ca2? channel

surface density also depends on b-subunit of calcium

channels. Using whole cell patch clamp method and fura-2

fluorescence imaging, Martin and co-workers demonstrated

that inhibitory actions of gabapentin on voltage gated

channels are dependent on a2d-1 as well as on b subunits

(Martin et al. 2002). Amongst the different b variants, b4a

splice variant is largely expressed at synapses, whereas b4b

is found in cell bodies of neurons and glial cells (Vendel

2007). Furthermore, only b4a form is expressed in the

spinal cord (Helton et al. 2002). Mich and Horne demon-

strated that gabapentin reduces the plasma membrane

trafficking of b4a-bound Cav2.1 complexes in the heterol-

ogous expression system (Mich and Horne 2008). It was

demonstrated that the inhibitory effects of gabapentin on

Ca2? channel expression could be reversed by increasing

concentrations of b4a subunit suggesting that the drug

244 A. Kukkar et al.

123

competes with b4a subunits in the process responsible for

Ca2? channel trafficking. The competition was reported to

be specific for b4a subunit and gabapentin had no effect in

the presence of b4b. The presence of b4a subunit in the

spinal cord also supports that the analgesic actions of

gabapentin are dependent of presence of b4a subunits

(Mich and Horne 2008).

NMDA receptors

Glutamate is an excitatory neurotransmitter in the nervous

system and it is released from pre-synaptic terminals in an

activity dependent manner between the primary afferents

and the spinal neurons involved in pain processing. The

studies have suggested its key role in pain development via

activation of NMDA (ionotropic) receptors (Jaggi and

Singh 2011). Apart from gabapentin mediated decreased

release of glutamate from pre-synaptic terminals of DRG,

gabapentin has been shown to directly inhibit NMDA

receptors in Xenopus oocytes (Hara and Sata 2007) that

may also be responsible for its anti-nociceptive activity.

Gabapentin has been shown to significantly inhibit NMDA

receptor-activated ion current and protect against NMDA-

induced excitotoxicity in rat cultured hippocampal CA1

neurons (Kim et al. 2009). A recent study has reported that

NMDA receptors blocker, MK801, potentiates the neuro-

pathic pain attenuating effects of intrathecal gabapentin in

CCI model (Yeh et al. 2011). A prospective, double blind

clinical trial has also demonstrated the efficacy of low dose

ketamine, an NMDA receptor antagonist, as adjuvant to

gabapentin in chronic pain (Amr 2010).

Protein kinase C (PKC)

The nerve injury-induced pain behaviour is associated with

over-expression of PKC-c in the spinal cord and its

involvement in nociceptive neuroplasticity in the spinal

cord has been well defined (Polgar et al. 1999; Furuta et al.

2009). Yeh and co-workers demonstrated that gabapentin

suppresses the PKC-c over-expression at the end of first

week and attributed the analgesic effects to inhibition of

PKC-c (Yeh et al. 2011). The contribution of Ca2?/cal-

modulin-dependent protein kinase II (CaMKII) to the

analgesic effect of gabapentin in a chronic constriction

injury model has also been defined and the analgesic

effects of gabapentin has been related to decreased

expression and phosphorylation of CaMKII in the spinal

cord (Ma et al. 2011).

Transient receptor potential (TRP) ion channels

The key role of temperature-sensitive transient recep-

tor potential ion channels (TRPs) including TRPA1 as

important pain sensors has been defined in number of

studies (Jaggi and Singh 2011) and TRPA1-deficient mice

generally show lack of sensitivities to different chemical

ligands and inflammatory mediators (Bautista et al. 2006;

Kwan et al. 2006). Bang and co-workers demonstrated that

gabapentin suppresses cinnamaldehyde-induced increase in

TRPA1 activity in Chinese hamster ovarian (CHO) heter-

ologous expression system and in cultured trigeminal

neurons without affecting other TRPs. It probably suggests

that TRPA1 on the dorsal horns or DRGs may also be a

critical target in pain alleviating effects of gabapentin

(Bang et al. 2009).

Supra-spinal actions of gabapentin

Gabapentin may also act supra-spinally to treat neuropathic

pain by stimulating descending inhibition to produce anti-

hypersensitivity in peripheral nerve injury. Peripheral

nerve injury induces differential changes in the plasticity of

GABAergic neurons in the locus coeruleus (LC) (increase

in GABA release) and spinal dorsal horn (decrease in

GABA release) and gabapentin is reported to selectively

reduce pre-synaptic GABA release in the LC, not in the

spinal dorsal horn (Yoshizumi et al. 2012b). The gaba-

pentin mediated reduction in GABAergic activity in the LC

is associated with an increased noradrenaline release that in

turn suppresses neurotransmission of pain in the spinal cord

via activation of a2 adrenoceptors (activation of descend-

ing pain inhibitory pathway to the spinal cord) (Hayashida

et al. 2008; Takasu et al. 2008; Yoshizumi et al. 2012b).

The earlier studies have shown that the anti-hypersensi-

tivity effect of both systemic and intra cerebro-ventricular

gabapentin are blocked by intrathecal injection of selective

a2-adrenoceptor antagonist, idazoxan suggesting the key

role of increased noradrenaline release in gabapentin

mediated analgesic effects (Hayashida et al. 2008).

The studies have demonstrated that gabapentin-induced

increased norepinephrine release in the spinal cord may

lead to G protein coupled inwardly rectifying potassium

channels (GIRK) activation which may eventually partici-

pate in its anti-hypersensitivity effects (Jones 1991; Tanabe

et al. 2005; Hayashida et al. 2007). Gabapentin-induced

noradrenaline may also activate GABAergic neurons in the

spinal dorsal horn via a1 adrenoceptors, therefore, gaba-

pentin-induced increase in spinal noradrenaline release

may also contribute to increased spinal GABA release

(Baba et al. 2000; Gassner et al. 2009). PKA-mediated

phosphorylation seems to be important for supra-spinal

actions of gabapentin in neuropathic conditions and it has

been described that gabapentin produces PKA-dependent

pre-synaptic enhancement of inhibitory GABAergic syn-

aptic transmission (Takasu et al. 2008) A recent study has

shown that activation of BDNF-trkB signaling is essential

Gabapentin in neuropathic pain 245

123

for gabapentin mediated activation of descending inhibi-

tory pathway involving a2 receptors (Hayashida and

Eisenach 2011).

There is involvement of glutamate dependent mecha-

nisms also in gabapentin stimulated descending inhibitory

pathway from activated LC neurons with an increased

spinal noradrenaline release in rats and humans (Hayashida

and Eisenach 2001; Hayashida et al. 2007). The anti-

hypersensitivity effects of systemically administered

gabapentin are shown to be blocked by intra-LC AMPA

receptor antagonist suggesting the key role of glutamate

signaling in gabapentin mediated effects in neuropathic

pain (Hayashida et al. 2001; Yoshizumi et al. 2012b).

Glutamate via activation of Ca2? permeable ionotropic

glutamate receptors increases intracellular Ca2? in astro-

cytes from internal stores through 1,4,5-inositol-trisphos-

phate signalling (Hansson et al. 2000; Verkhratsky and

Kirchhoff 2007). However in some astrocytes, glutamate

transporters (responsible for glutamate reuptake) may

also result in Ca2? influx (Kirischuk et al. 1997; Rojas

et al. 2007) and may paradoxically result in glutamate

release from astrocytes via Ca2? dependent mechanisms

(Malarkey and Parpura 2008).

Difference in mechanism for acute and chronic effects

of gabapentin

The slow developing and long lasting analgesic actions of

gabapentin are dependent upon inhibition nerve injury-

induced up-regulation of a2d-1 subunits in the dorsal horn

neurons by anterograde trafficking (axoplasmic transport)

and by inhibiting trafficking from cytoplasm to plasma

membrane (membrane trafficking) of pre-synaptic mem-

branes of DRG neurons and dorsal horn neurons. Since

these actions of gabapentin may require exposures of hours

to days, therefore, these mechanisms have been suggested

to contribute to chronic and sustained actions of gabapen-

tin. The sustained actions of gabapentin after the termina-

tion of its administration may be probably attributed to

time required for transport of the a2/d-1 subunit from the

DRG to the primary afferent nerve terminals (Morimoto

et al. 2012). However, gabapentin also produces acute and

transient analgesic effects (Morimoto et al. 2012) which are

dependent on constitutively expressed and functional

spinal system that does not require additional changes in

protein expression/channel mobilization and extended drug

exposure (Takasusuki and Yaksh 2011). These mechanisms

may include decreased release of nociceptive neurotrans-

mitters in pain processing pathway as Takasusuki and co-

workers have reported that acute analgesic effects of

gabapentin are dependent on decreased release of sub-

stance P from small primary afferents (Takasusuki and

Yaksh 2011) (Fig. 3).

Gabapentin versus other neuropathic drugs

TCA including imipramine and amitriptyline have gener-

ally been the first drugs of choice for management of

Fig. 3 Proposed mechanisms

for acute, transient and chronic,

sustained analgesic actions

of gabapentin

246 A. Kukkar et al.

123

neuropathic pain. However, gabapentinoid drugs have

emerged as first-line treatment for neuropathic pain due to

better efficacy and safety profile (Haslam and Nurmikko

2008; Vranken 2009). In patients with the spinal cord

injury, gabapentin has been shown to be more effective for

pain relief than amitriptyline (Selph et al. 2011). Among

gabapentinoid drugs, gabapentin and pregabalin have been

shown to be equally in reducing pain intensity, improving

sleep quality and depression in patients with painful

peripheral neuropathy (Biyik 2012). However, Saldana and

co-workers have demonstrated the efficacy of pregabalin in

gabapentin refractory patients (Saldana et al. 2012). Some

other studies have also the superiority of pregabalin in

alleviating cancer related neuropathic pain (Mishra et al.

2012), PHN (Ifuku et al. 2011) and fibromyalgia (Lloyd

et al. 2012).

Synergistic effects of gabapentin and antidepressants

Gabapentin shows synergistic effect with anti-depressant

venlafaxine in treating neuropathic pain and the combina-

tion was reported to be superior in comparison to gaba-

pentin alone in the SNI model of neuropathic pain (Garry

et al. 2005). The combined gabapentin and nortriptyline

therapy has also been reported to be more efficacious than

either drug alone for neuropathic pain management (Gilron

et al. 2009). Arai and co-workers demonstrated that low

dose gabapentin (200–400 mg/12 h)-imipramine (10 mg/

12 h) combination with opioids was effective in managing

neuropathic cancer pain without severe adverse effects

(Arai et al. 2010). The synergistic actions of gabapentin

and TCA may probably be attributed to potentiation of

noradrenaline mediated activation of descending pain

inhibitory pathway from the LC to the spinal cord. Gaba-

pentin by virtue of reduction in GABAergic activity in the

LC leads to an increased noradrenaline release, while

antidepressants increase noradrenaline levels by blocking

pre-synaptically located noradrenaline-serotonin reuptake

pump. An increase in noradrenaline release in the LC may

potentiate descending pain inhibitory pathway from the LC

to the spinal cord and attenuate neuropathic pain.

Conclusion

There preclinical and clinical evidences have shown the

importance of gabapentin in neuropathic pain management

and it has emerged as one of the first line agents for pain

management particularly in diabetic neuropathy and post-

herpetic neuralgia. The majority of its actions have been

ascribed to its binding to a2d-1 subunits leading to

decreased expression of voltage gated calcium channels on

dorsal horn neurons via inhibition of membrane and

anterograde trafficking. However, its analgesic actions

involve more targets whose critical participation in pain

alleviation needs more studies.

Acknowledgments The authors are grateful to Department of

Pharmaceutical Sciences & Drug Research, Punjabi University, Pat-

iala, India for providing technical facilities.

References

Alden, K.J., and J. Garcia. 2001. Differential effect of gabapentin on

neuronal and muscle calcium currents. Journal of Pharmacologyand Experimental Therapeutics 297: 727–735.

Amr, Y.M. 2010. Multi-day low dose ketamine infusion as adjuvant

to oral gabapentin in spinal cord injury related chronic pain: A

prospective, randomized, double blind trial. Pain Physician 13:

245–249.

Arai, Y.C., T. Matsubara, K. Shimo, K. Suetomi, M. Nishihara,

T. Ushida, K. Kobayashi, C. Suzuki, A. Kinoshita, M. Kondo,

S. Matsubara, R. Hayashi, Y. Tohyama, K. Nishida, and

M. Arakawa. 2010. Low-dose gabapentin as useful adjuvant to

opioids for neuropathic cancer pain when combined with low-

dose imipramine. Journal of Anesthesia 24: 407–410.

Argoff, C.E. 2011. Review of current guidelines on the care of post

herpetic neuralgia. Postgraduate Medicine 123: 134–142.

Attal, N., G. Mazaltarine, B. Perrouin-Verbe, T. Albert, and SOFMER

French Society for Physical Medicine and Rehabilitation. 2009.

Chronic neuropathic pain management in spinal cord injury

patients. What is the efficacy of pharmacological treatments

with a general mode of administration? (oral, transdermal, intra-

venous). Annals of Physical and Rehabilitation Medicine 52:

124–141.

Baba, H., K. Shimoji, and M. Yoshimura. 2000. Norepinephrine

facilitates inhibitory transmission in substantia gelatinosa of

adult rat spinal cord (part 1): Effects on axon terminals of

GABAergic and glycinergic neurons. Anesthesiology 92:

473–484.

Back, S.K., S.Y. Won, S.K. Hong, and H.S. Na. 2004. Gabapentin

relieves mechanical, warm and cold allodynia in a rat model of

peripheral neuropathy. Neuroscience Letters 368: 341–344.

Backonja, M., A. Beydoun, K.R. Edwards, S.L. Schwartz, V.

Fonseca, M. Hes, L. LaMoreaux, and E. Garofalo. 1998.

Gabapentin for the symptomatic treatment of painful neuropathy

in patients with diabetes mellitus: A randomized controlled trial.

JAMA 280: 1831–1836.

Backonja, M.M., D.M. Canafax, and K.C. Cundy. 2011. Efficacy of

gabapentin enacarbil vs placebo in patients with postherpetic

neuralgia and a pharmacokinetic comparison with oral gabapen-

tin. Pain Medicine 12: 1098–1108.

Backonja, M., and R.L. Glanzman. 2003. Gabapentin dosing for

neuropathic pain: Evidence from randomized, placebo-con-

trolled clinical trials. Clinical Therapeutics 25: 81–104.

Bang, S., S. Yoo, and S.W. Hwang. 2009. Gabapentin attenuates the

activation of transient receptor potential A1 by cinnamaldehyde.

Experimental Neurobiology 18: 1–7.

Bauer, C.S., M. Nieto-Rostro, W. Rahman, A. Tran-Van-Minh,

L. Ferron, L. Douglas, I. Kadurin, Y. Sri Ranjan, L. Fernandez-

Alacid, N.S. Millar, A.H. Dickenson, R. Lujan, and

A.C. Dolphin. 2009. The increased trafficking of the calcium

channel subunit alpha2delta-1 to presynaptic terminals in

neuropathic pain is inhibited by the alpha2delta ligand pregab-

alin. Journal of Neuroscience 29: 4076–4088.

Gabapentin in neuropathic pain 247

123

Bautista, D.M., S.E. Jordt, T. Nikai, P.R. Tsuruda, A.J. Read, J.

Poblete, E.N. Yamoah, A.I. Basbaum, and D. Julius. 2006.

TRPA1 mediates the inflammatory actions of environment tal

irritants and proalgesic agents. Cell 124: 1269–1282.

Berger, A., E. Dukes, B. McCarberg, M. Liss, and G. Oster. 2003.

Change in opioid use after the initiation of gabapentin therapy in

patients with postherpetic neuralgia. Clinical Therapeutics 25:

2809–2821.

Beydoun, A. 1999. Postherpetic neuralgia: Role of gabapentin and

other treatment modalities. Epilepsia 40(Suppl 6): S51–S56.

Bilgir, O., M. Calan, F. Bilgir, L. Kebapcilar, A. Yuksel, Y. Yildiz,

and I. Sari. 2009. Gabapentin-induced rhabdomyolysis in a

patient with diabetic neuropathy. Internal Medicine 48:

1085–1087.

Biyik, Z., Y. Solak, H. Atalay, A. Gaipov, F. Guney, and S. Turk.

2012. Gabapentin versus pregabalin in improving sleep quality

and depression in hemodialysis patients with peripheral neurop-

athy: A randomized prospective crossover trial. InternationalUrology and Nephrology, May 29. [Epub ahead of print].

Bodensteiner, J.B., H.H. Morris, and G.S. Golden. 1981. Asterixis

associated with sodium valproate. Neurology 31: 195–196.

Bookwalter, T., and M. Gitlin. 2005. Gabapentin-induced neurologic

toxicities. Pharmacotherapy 25: 1817–1819.

Boroujerdi, A., J. Zeng, K. Sharp, D. Kim, O. Steward, and Z.D. Luo.

2011. Calcium channel alpha-2-delta-1 protein upregulation in

dorsal spinal cord mediates spinal cord injury-induced neuro-

pathic pain states. Pain 152: 649–655.

Boulton, A.J. 2003. Treatment of symptomatic diabetic neuropathy.

Diabetes/Metabolism Research and Reviews 19(Suppl 1): S16–

S21.

Butterworth, R.F. 1996. Neuroactive aminoacids in hepatic enceph-

alopathy. Metabolic Brain Disease 11: 165–173.

Cahill, C.M., and T.J. Coderre. 2002. Attenuation of hyperalgesia in a

rat model of neuropathic pain after intrathecal pre- or post-

treatment with a neurokinin-1 antagonist. Pain 95: 277–285.

Calabro, R.S. 2011. Gabapentin and sexual dysfunction: An over-

looked and underreported problem? Epilepsy & Behavior 22:

818.

Cantı, C., M. Nieto-Rostro, I. Foucault, F. Heblich, J. Wratten, M.W.

Richards, J. Hendrich, L. Douglas, K.M. Page, A. Davies, and

A.C. Dolphin. 2005. The metal-ion-dependent adhesion site in

the Von Willebrand factor—A domain of alpha2delta subunits is

key to trafficking voltage-gated Ca2? channels. Proceedings ofthe National Academy of Sciences of the United States ofAmerica 102: 11230–11235.

Chandra, K., N. Shafiq, P. Pandhi, S. Gupta, and S. Malhotra. 2006.

Gabapentin versus nortriptyline in post-herpetic neuralgia

patients: A randomized, double-blind clinical trial—the GONIP

Trial. International Journal of Clinical Pharmacology andTherapeutics 44: 358–363.

Chen, S.R., and H.L. Pan. 2005. Effect of systemic and intrathecal

gabapentin on allodynia in a new rat model of postherpetic

neuralgia. Brain Research 1042: 108–113.

Cheshire, W.P. 2007. Trigeminal neuralgia: For one nerve a multitude

of treatments. Expert Review of Neurotherapeutics 7: 1565–1579.

Chong, M.S., and J. Hester. 2007. Diabetic painful neuropathy:

Current and future treatment options. Drugs 67: 569–585.

Chou, R., S. Carson, and B.K. Chan. 2009. Gabapentin versus

tricyclic antidepressants for diabetic neuropathy and post-

herpetic neuralgia: Discrepancies between direct and indirect

meta-analyses of randomized controlled trials. Journal ofGeneral Internal Medicine 24: 178–188.

Chu, L.C., M.L. Tsaur, C.S. Lin, Y.C. Hung, T.Y. Wang, C.C. Chen,

and J.K. Cheng. 2011. Chronic intrathecal infusion of gabapentin

prevents nerve ligation-induced pain in rats. British Journal ofAnaesthesia 106: 699–705.

Coderre, T.J., N. Kumar, C.D. Lefebvre, and J.S. Yu. 2007. A

comparison of the glutamate release inhibition and anti-allodynic

effects of gabapentin, lamotrigine, and riluzole in a model of

neuropathic pain. Journal of Neurochemistry 100: 1289–1299.

Dallocchio, C., C. Buffa, P. Mazzarello, and S. Chiroli. 2000.

Gabapentin vs. amitriptyline in painful diabetic neuropathy: An

open-label pilot study. Journal of Pain and Symptom Manage-ment 20: 280–285.

Davies, A., J. Hendrich, A.T. Van Minh, J. Wratten, L. Douglas, and

A.C. Dolphin. 2007. Functional biology of the alpha (2) delta

subunits of voltage gated calcium channels. Trends in Pharma-cological Sciences 28: 220–228.

Dworkin, R.H., A.B. O’Connor, M. Backonja, J.T. Farrar, N.B.

Finnerup, T.S. Jensen, E.A. Kalso, J.D. Loeser, C. Miaskowski,

T.J. Nurmikko, R.K. Portenoy, A.S. Rice, B.R. Stacey, R.D.

Treede, D.C. Turk, and M.S. Wallace. 2007. Pharmacologic

management of neuropathic pain: Evidence-based recommenda-

tions. Pain 132: 237–251.

Erdogan, G., D. Ceyhan, and S. Gulec. 2011. Possible heart failure

associated with pregabalin use: Case report. Agri 23: 80–83.

Ermis, N., H. Gullu, M. Caliskan, A. Unsal, M. Kulaksizoglu, and

H. Muderrisoglu. 2010. Gabapentin therapy improves heart rate

variability in diabetic patients with peripheral neuropathy.

Journal of Diabetes and Its Complications 24: 229–233.

Field, M.J., P.J. Cox, E. Stott, H. Melrose, J. Offord, T.Z. Su,

S. Bramwell, L. Corradini, S. England, J. Winks, R.A. Kinloch,

J. Hendrich, A.C. Dolphin, T. Webb, and D. Williams. 2006.

Identification of the a2-d-1 subunit of voltage dependent calcium

channels as a molecular target for pain mediating the analgesic

actions of pregabalin. Proceedings of the National Academy ofSciences of the United States of America 103: 17537–17542.

Folkesson, A., P.H. Honore, L.M. Andersen, P. Kristensen, and O.J.

Bjerrum. 2010. Low dose of donepezil improves gabapentin

analgesia in the rat spared nerve injury model of neuropathic

pain: Single and multiple dosing studies. Journal of NeuralTransmission 117: 1377–1385.

Furuta, S., T. Shimizu, M. Narita, K. Matsumoto, N. Kuzumaki,

S. Horie, T. Suzuki, and M. Narita. 2009. Subdiaphragmatic

vagotomy promotes nociceptive sensitivity of deep tissue in rats.

Neuroscience 164: 1252–1262.

Garry, E.M., A. Delaney, H.A. Anderson, E.C. Sirinathsinghji,

R.H. Clapp, W.J. Martin, P.R. Kinchington, D.L. Krah, C.

Abbadie, and S.M. Fleetwood-Walker. 2005. Varicella zoster

virus induces neuropathic changes in rat dorsal root ganglia and

behavioral reflex sensitisation that is attenuated by gabapentin or

sodium channel blocking drugs. Pain 118: 97–111.

Gassner, M., R. Ruscheweyh, and J. Sandkuhler. 2009. Direct

excitation of spinal GABAergic interneurons by noradrenaline.

Pain 145: 204–210.

Geber, C., U. Baumgartner, R. Schwab, H. Muller, P. Stoeter,

M. Dieterich, C. Sommer, F. Birklein, and R.D. Treede. 2009.

Revised definition of neuropathic pain and its grading system:

An open case series illustrating its use in clinical practice.

American Journal of Medicine 122(10 Suppl): S3–S12.

Gilron, I., J.M. Bailey, D. Tu, R.R. Holden, A.C. Jackson, and

R.L. Houlden. 2009. Nortriptyline and gabapentin, alone and in

combination for neuropathic pain: A double-blind, randomised

controlled crossover trial. Lancet 374: 1252–1261.

Gilron, I., J.M. Bailey, D. Tu, R.R. Holden, D.F. Weaver, and R.L.

Houlden. 2005. Morphine, gabapentin, or their combination for

neuropathic pain. New England Journal of Medicine 352: 1324–1334.

Gurnett, C.A., M. De Waard, and K.P. Campbell. 1996. Dual function

of the voltage dependent Ca2? channel a2-d subunit in current

stimulation and subunit interaction. Neuron 16: 431–440.

Hanna, M., C. O’Brien, and M.C. Wilson. 2008. Prolonged-release

oxycodone enhances the effects of existing gabapentin therapy in

248 A. Kukkar et al.

123

painful diabetic neuropathy patients. European Journal of Pain12: 804–813.

Hansson, E., H. Muyderman, J. Leonova, L. Allansson, J. Sinclair,

F. Blomstrand, T. Thorlin, M. Nilsson, and L. Ronnback. 2000.

Astroglia and glutamate in physiology and pathology: Aspects

on glutamate transport, glutamate-induced cell swelling and

gap-junction communication. Neurochemistry International 37:

317–329.

Hara, K., and T. Sata. 2007. Inhibitory effect of gabapentin on

N-methyl-D-aspartate receptors expressed in Xenopus oocytes.

Acta Anaesthesiologica Scandinavica 51: 122–128.

Haslam, C., and T. Nurmikko. 2008. Pharmacological treatment of

neuropathic pain in older persons. Clinical Interventions inAging 3: 111–120.

Hasnie, F.S., J. Breuer, S. Parker, V. Wallace, J. Blackbeard, I. Lever,

P.R. Kinchington, A.H. Dickenson, T. Pheby, and A.S. Rice.

2007. Further characterization of a rat model of varicella zoster

virus-associated pain: Relationship between mechanical hyper-

sensitivity and anxiety-related behaviour, and the influence of

analgesic drugs. Neuroscience 144: 1495–1508.

Hayashida, K., and J.C. Eisenach. 2008. Multiplicative interactions to

enhance gabapentin to treat neuropathic pain. European Journalof Pharmacology 598: 21–26.

Hayashida, K., and J.C. Eisenach. 2011. A tropomyosine receptor

kinase inhibitor blocks spinal neuroplasticity essential for the

anti-hypersensitivity effects of gabapentin and clonidine in rats

with peripheral nerve injury. Journal of Pain 12: 94–100.

Hayashida, K., H. Obata, K. Nakajima, and J.C. Eisenach. 2008.

Gabapentin acts within the locus coeruleus to alleviate neuro-

pathic pain. Anesthesiology 109: 1077–1084.

Hayashida, K., R. Parker, and J.C. Eisenach. 2007. Oral gabapentin

activates spinal cholinergic circuits to reduce hypersensitivity

after peripheral nerve injury and interacts synergistically with

oral donepezil. Anesthesiology 106: 1213–1219.

Helton, T.D., D.J. Kojetin, J. Cavanagh, and W.A. Horne. 2002.

Alternative splicing of a b4 subunit proline-rich motif regulates

voltage-dependent gating and toxin block of Cav2.1 Ca2?

channels. Journal of Neuroscience 22: 9331–9339.

Hemstreet, B., and M. Lapointe. 2001. Evidence for the use of

gabapentin in the treatment of diabetic peripheral neuropathy.

Clinical Therapeutics 23: 520–531.

Hindmarsh, K.W., L. Tan, K. Sankaran, and V.A. Laxdal. 1989.

Diurnal rhythms of cortisol, ACTH, and beta-endorphin levels in

neonates and adults. Western Journal of Medicine 151: 153–156.

Hon, N.C., and Y.J. Fei. 2008. Prescription pattern of oral healthcare

professionals in the use of anticonvulsants for trigeminal

neuralgia. Annals of the Royal Australasian College of DentalSurgeons 19: 162–164.

Honarmand, A., M. Safavi, and M. Zare. 2011. Gabapentin: An

update of its pharmacological properties and therapeutic use in

epilepsy. Journal of Medical Sciences Research 16: 1062–1069.

Hoppa, M.B., B. Lana, W. Margas, A.C. Dolphin, and T.A. Ryan.

2012. a2d expression sets presynaptic calcium channel abun-

dance and release probability. Nature 486: 122–125.

Ifuku, M., M. Iseki, I. Hidaka, Y. Morita, S. Komatus, and E. Inada.

2011. Replacement of gabapentin with pregabalin in postherpetic

neuralgia therapy. Pain Medicine 12: 1112–1116.

Jacob, P. C., R. P. Chand, and el-S. Omeima. 2000. Asterixis induced

by gabapentin. Clin. Neuropharmacol, 23, 53.

Jaggi, A.S., and N. Singh. 2011. Role of different brain areas in

peripheral nerve injury-induced neuropathic pain. BrainResearch 1381: 187–201.

Jarvis, S.E., and G.W. Zamponi. 2007. Trafficking and regulation of

neuronal voltage gated calcium channels. Current Opinion inCell Biology 19: 474–482.

Jean, W.H., C.C. Wu, M.S. Mok, and W.Z. Sun. 2005. Starting dose

of gabapentin for patients with post-herpetic neuralgia—a dose-

response study. Acta Anaesthesiologica Taiwanica 43: 73–77.

Jones, S.L. 1991. Descending noradrenergic influences on pain.

Progress in Brain Research 88: 381–394.

Kaufman, K.R., and P.J. Struck. 2011. Gabapentin-induced sexual

dysfunction. Epilepsy & Behavior 21: 324–326.

Keskinbora, K., A.F. Pekel, and I. Aydinli. 2007. Gabapentin and an

opioid combination versus opioid alone for the management of

neuropathic cancer pain: A randomized open trial. Journal ofPain and Symptom Management 34: 183–189.

Kim, Y.S., H.K. Chang, J.W. Lee, Y.H. Sung, S.E. Kim, M.S. Shin,

J.W. Yi, J.H. Park, H. Kim, and C.J. Kim. 2009. Protective effect

of gabapentin on N-methyl-D-aspartate-induced excitotoxicity in

rat hippocampal CA1 neurons. Journal of PharmacologicalScience 109: 144–147.

Kirischuk, S., H. Kettenmann, and A. Verkhratsky. 1997. Na?/Ca2?

exchanger modulates kainate-triggered Ca2? signaling in Berg-

mann glial cells in situ. FASEB Journal 11: 566–572.

Ko, S.H., H.S. Kwon, J.M. Yu, S.H. Baik, I.B. Park, J.H. Lee, K.S.

Ko, J.H. Noh, D.S. Kim, C.H. Kim, J.O. Mok, T.S. Park, H.S.

Son, and B.Y. Cha. 2010. Comparison of the efficacy and safety

of tramadol/acetaminophen combination therapy and gabapentin

in the treatment of painful diabetic neuropathy. DiabeticMedicine 27: 1033–1040.

Kusunose, N., S. Koyanagi, K. Hamamura, N. Matsunaga, M.

Yoshida, T. Uchida, M. Tsuda, K. Inoue, and S. Ohdo. 2010.

Molecular basis for the dosing time-dependency of anti-

allodynic effects of gabapentin in a mouse model of neuropathic

pain. Molecular Pain 6: 83.

Kwan, K.Y., A.J. Allchorne, M.A. Vollrath, A.P. Christensen, D.S.

Zhan, C.J. Woolf, and D.P. Corey. 2006. TRPA1 contributes

cold, mechanical, and chemical nociception but is n essential for

hair-cell transduction. Neuron 50: 277–289.

Lemos, L., R. Fontes, S. Flores, P. Oliveira, and A. Almeida. 2010.

Effectiveness of the association between carbamazepine and

peripheral analgesic block with ropivacaine for the treatment of

trigeminal neuralgia. Journal of Pain Research 3: 201–212.

Li, C.Y., X.L. Zhang, E.A. Matthews, K.W. Li, A. Kurwa, A.

Boroujerdi, J. Gross, M.S. Gold, A.H. Dickenson, G. Feng, and

Z.D. Luo. 2006. Calcium channel a2-d-1 subunit mediates spinal

hyperexcitability in pain modulation. Pain 125: 20–34.

Liu, Y., N. Qin, T. Reitz, Y. Wang, and C.M. Flores. 2006. Inhibition

of the rat brain sodium channel Nav1.2 after prolonged exposure

to gabapentin. Epilepsy Research 70: 263–268.

Lloyd, A., C.S. Boomershine, E.H. Choy, A. Chandran, and G. Zlateva.

2012. The cost-effectiveness of pregabalin in the treatment of

fibromyalgia: US perspective. Journal of Medicinal Economics15: 481–492.

Lopez, P.R., T. Rachael, S. Leicht, and R.D. Smalligan. 2010.

Gabapentin-induced delusions of parasitosis. Southern MedicalJournal 103: 711–712.

Luo, Z.D., N.A. Calcutt, E.S. Higuera, C.R. Valder, Y.H. Song, C.I.

Svensson, and R.R. Myers. 2002. Injury type-specific calcium

channel alpha 2 delta-1 subunit up-regulation in rat neuropathic

pain models correlates with antiallodynic effects of gabapentin.

Journal of Pharmacology and Experimental Therapeutics 303:

1199–1205.

Ma, L.L., W. Liu, Y.G. Huang, N. Yang, and P.P. Zuo. 2011.

Analgesic effect of gabapentin in a rat model for chronic

constrictive injury. Chinese Medical Journal (English Edition)124: 4304–4309.

Malarkey, E.B., and V. Parpura. 2008. Mechanisms of glutamate

release from astrocytes. Neurochemistry International 52:

142–154.

Gabapentin in neuropathic pain 249

123

Martin, D.J., D. McClelland, M.B. Herd, K.G. Sutton, M.D. Hall,

K. Lee, R.D. Pinnock, and R.H. Scott. 2002. Gabapentin-

mediated inhibition of voltage-activated Ca2? channel currents

in cultured sensory neurones is dependent on culture conditions

and channel subunit expression. Neuropharmacology 42: 353–366.

Mich, P.M., and W.A. Horne. 2008. Alternative splicing of the Ca

channel beta4 subunit confers specificity for gabapentin inhibi-

tion of Cav2.1 trafficking. Molecular Pharmacology 74:

904–912.

Mishra, S., S. Bhatnagar, G.N. Goyal, S.P. Rana, and S.P. Upadhya.

2012. A comparative efficacy of amitriptyline, gabapentin, and

pregabalin in neuropathic cancer pain: A prospective random-

ized double-blind placebo-controlled study. American Journal ofHospice & Palliative Care 29: 177–182.

Mittal, M., M. Pasnoor, R.B. Mummaneni, S. Khan, A. McVey, D.

Saperstein, L. Herbelin, L. Ridings, Y. Wang, M.M. Dimachkie,

and R.J. Barohn. 2011. Retrospective chart review of duloxetine

and pregabalin in the treatment of painful neuropathy. Journal ofNeuroscience 121: 521–527.

Mixcoatl-Zecuatl, T., G.N. Quinonez-Bastidas, N.L. Caram-Salas,

M. Ambriz-Tututi, C.I. Araiza-Saldana, H.I. Rocha-Gonzalez,

R. Medina-Santillan, G. Reyes-Garcia, and V. Granados-Soto.

2008. Synergistic antiallodynic interaction between gabapentin

or carbamazepine and either benfotiamine or cyanocobalamin in

neuropathic rats. Methods and Findings in Experimental andClinical Pharmacology 30: 431–441.

Miyano, K., H.B. Tang, Y. Nakamura, N. Morioka, A. Inoue, and Y.

Nakata. 2009. Paclitaxel and vinorelbine, evoked the release of

substance P from cultured rat dorsal root ganglion cells through

different PKC isoform-sensitive ion channels. Neuropharmacol-ogy 57: 25–32.

Morello, C.M., S.G. Leckband, C.P. Stoner, D.F. Moorhouse, and

G.A. Sahagian. 1999. Randomized double-blind study compar-

ing the efficacy of gabapentin with amitriptyline on diabetic

peripheral neuropathy pain. Archives of Internal Medicine 159:

1931–1937.

Morimoto, S., M. Ito, S. Oda, A. Sugiyama, M. Kuroda, and S.

Adachi-Akahane. 2012. Spinal mechanism underlying the anti-

allodynic effect of gabapentin studied in the mouse spinal nerve

ligation model. Journal of Pharmacological Science 118:

455–466.

O’Connor, A.B., K. Noyes, and R.G. Holloway. 2007. A cost-

effectiveness comparison of desipramine, gabapentin, and pre-

gabalin for treating postherpetic neuralgia. Journal of theAmerican Geriatrics Society 55: 1176–1184.

Odrcich, M., J.M. Bailey, C.M. Cahill, and I. Gilron. 2006.

Chronobiological characteristics of painful diabetic neuropathy

and postherpetic neuralgia: Diurnal pain variation and effects of

analgesic therapy. Pain 120: 207–212.

Pandey, C.K., N. Singh, and P.K. Singh. 2008. Gabapentin for

refractory idiopathic trigeminal neuralgia. Journal of the IndianMedical Association 106: 124–125.

Paradowski, B., and M. Bilinska. 2003. Gabapentin in the treatment

of neuropathic pain in patients with type 2 diabetes mellitus. PolMerkur Lekarski 15: 61–64.

Park, S., E.S. Ahn, D.W. Han, J.H. Lee, K.T. Min, H. Kim, and

Y.W. Hong. 2008. Pregabalin and gabapentin inhibit substance

P-induced NF-kappaB activation in neuroblastoma and glioma

cells. Journal of Cellular Biochemistry 105: 414–423.

Parsons, B., L. Tive, and S. Huang. 2004. Gabapentin: A pooled

analysis of adverse events from three clinical trials in patients

with postherpetic neuralgia. American Journal of GeriatricPharmacotherapy 2: 157–162.

Patarica-Huber, E., N. Boskov, and M. Pjevic. 2011. Multimodal

approach to therapy-related neuropathic pain in breast cancer.

Journal of BUON 16: 40–45.

Perez, C., A. Navarro, M.T. Saldana, X. Masramon, and J. Rejas.

2010. Pregabalin and gabapentin in matched patients with

peripheral neuropathic pain in routine medical practice in a

primary care setting: Findings from a cost-consequences analysis

in a nested case-control study. Clinical Therapeutics 32:

1357–1370.

Perloff, M.D., D.E. Thaler, and J.A. Otis. 2011. Anorgasmia with

gabapentin may be common in older patients. American Journalof Geriatric Pharmacotherapy 9: 199–203.

Petraglia, F., F. Facchinetti, D. Parrini, G. Micieli, S. De Luca, and

A.R. Genazzani. 1983. Simultaneous circadian variations of

plasma ACTH, beta-lipotropin, beta-endorphin and cortisol.

Hormone Research 17: 147–152.

Polgar, E., J.H. Fowler, M.M. McGill, and A.J. Todd. 1999. The types

of neuron which contain protein kinase C gamma in rat spinal

cord. Brain Research 833: 71–80.

Quintero, J.E., D.J. Dooley, F. Pomerleau, P. Huettl, and G.A.

Gerhardt. 2011. Amperometric measurement of glutamate

release modulation by gabapentin and pregabalin in rat neocor-

tical slices: Role of voltage-sensitive Ca2? a2d-1 subunit.

Journal of Pharmacology and Experimental Therapeutics 338:

240–245.

Rice, A.S., and S. Maton. 2001. Gabapentin in postherpetic neuralgia:

A randomised, double blind, placebo controlled study. Pain 94:

215–224.

Richardson, C.E., D.W. Williams, and J.G. Kingham. 2002. Gaba-

pentin induced cholestasis. BMJ 325: 635.

Rock, D.M., K.M. Kelly, and R.L. Macdonald. 1993. Gabapentin

actions on ligand- and voltage-gated responses in cultured rodent

neurons. Epilepsy Research 16: 89–98.

Rojas, H., C. Colina, M. Ramos, G. Benaim, E.H. Jaffe, C. Caputo,

and R. DiPolo. 2007. Na? entry via glutamate transporter

activates the reverse Na?/Ca2? exchange and triggers Ca(i)2?-

induced Ca2? release in rat cerebellar Type-1 astrocytes.

Neurochemistry 100: 1188–1202.

Ross, J.R., K. Goller, J. Hardy, J. Riley, K. Broadley, R. A’hern, and

J. Williams. 2005. Gabapentin is effective in the treatment of

cancer-related neuropathic pain: A prospective, open-label study.

Journal of Palliative Medicine 8: 1118–1126.

Rowbotham, M., N. Harden, B. Stacey, P. Bernstein, and L. Magnus-

Miller. 1998. Gabapentin for the treatment of postherpetic

neuralgia: A randomized controlled trial. JAMA 280: 1837–1842.

Ruessmann, H.J. 2009. Switching from pathogenetic treatment with

alpha-lipoic acid to gabapentin and other analgesics in painful

diabetic neuropathy: A real-world study in outpatients. Journalof Diabetes and Its Complications 23: 174–177.

Said, G. 2007. Diabetic neuropathy—a review. Nature ClinicalPractice Neurology 3: 331–340.

Saldana, M.T., C. Perez, A. Navarro, X. Masramon, and J. Rejas.

2012. Pain alleviation and patient-reported health outcomes

following switching to pregabalin in individuals with gabapen-

tin-refractory neuropathic pain in routine medical practice.

Clinical Drug Investigation 32: 401–412.

Sandercock, D., M. Cramer, J. Wu, Y.K. Chiang, V. Biton, and

M. Heritier. 2009. Gabapentin extended release for the treatment

of painful diabetic peripheral neuropathy: Efficacy and tolera-

bility in a double-blind, randomized, controlled clinical trial.

Diabetes Care 32: e20.

See, S., E. Hendriks, and L. Hsiung. 2011. Akathisia induced by

gabapentin withdrawal. Annals of Pharmacotherapy 45: e31.

Selph, S., S. Carson, R. Fu, S. Thakurta, A. Low, and M. McDonagh.

2011. Drug class review: neuropathic pain: Final update 1report [Internet]. Portland (OR): Oregon Health & Science

University.

Takahashi, H., and N. Shimoyama. 2010. A prospective open-label

trial of gabapentin as an adjuvant analgesic with opioids for

250 A. Kukkar et al.

123

Japanese patients with neuropathic cancer pain. InternationalJournal of Clinical Oncology 15: 46–51.

Takasu, K., H. Ono, and M. Tanabe. 2008. Gabapentin produces

PKA-dependent pre-synaptic inhibition of GABAergic synaptic

transmission in LC neurons following partial nerve injury in

mice. Journal of Neurochemistry 105: 933–942.

Takasusuki, T., and T.L. Yaksh. 2011. The effects of intrathecal and

systemic gabapentin on spinal substance P release. Anesthesiaand Analgesia 112: 971–976.

Tanabe, M., K. Takasu, N. Kasuya, S. Shimizu, M. Honda, and H.

Ono. 2005. Role of descending noradrenergic system and spinal

alpha2-adrenergic receptors in the effects of gabapentin on

thermal and mechanical nociception after partial nerve injury in

the mouse. British Journal of Pharmacology 144: 703–714.

Taylor, C.P. 1997. Mechanisms of action of gabapentin. Rev Neurol(Paris) 153(suppl 1): 539–545.

Taylor, C.P. 2009. Mechanisms of analgesia by gabapentin and

pregabalin–calcium channel alpha2-delta [Cavalpha2-delta]

ligands. Pain 142: 13–16.

Tsavaris, N., P. Kopterides, C. Kosmas, A. Efthymiou, H. Skopelitis,

A. Dimitrakopoulos, E. Pagouni, D. Pikazis, P.V. Zis, and C.

Koufos. 2008. Gabapentin monotherapy for the treatment of

chemotherapy-induced neuropathic pain: A pilot study. PainMedicine 9: 1209–1216.

Tuccori, M., G. Lombardo, F. Lapi, A. Vannacci, C. Blandizzi, and

M. Del Tacca. 2007. Gabapentin-induced severe myopathy.

Annals of Pharmacotherapy 41: 1301–1305.

Vadalouca, A., E. Raptis, E. Moka, P. Zis, P. Sykioti, and I. Siafaka.

2012. Pharmacological treatment of neuropathic cancer pain: A

comprehensive review of the current literature. Pain Practice 12:

219–251.

Vendel, A. C., M. D. Terry, A. R. Striegel, N. M. Iverson, V.

Leuranguer, C. D. Rithner, B. A. Lyons, G. E. Pickard, S.

A. Tobet, and W. A. Horne. 2007. Alternative splicing of the

voltage-gated Ca2? channel b4 subunit creates a uniquely folded

N-terminal protein binding domain with cell-specific expression

in the cerebellar cortex. Journal of Neuroscience 26, 2635–2644

(2006).

Vranken, J.H. 2009. Mechanisms and treatment of neuropathic pain.

Central Nervous System Agents in Medicinal Chemistry 9:

71–78.

Walczak, J.S., V. Pichette, F. Leblond, K. Desbiens, and P. Beaulieu.

2005. Behavioural, pharmacological and molecular character-

ization of the saphenous nerve partial ligation: A new model of

neuropathic pain. Neuroscience 132: 1093–1102.

Wallace, V.C., J. Blackbeard, A.R. Segerdahl, F. Hasnie, T. Pheby,

S.B. McMahon, and A.S. Rice. 2007. Characterization of rodent

models of HIV-gp120 and anti-retroviral-associated neuropathic

pain. Brain 130: 2688–2702.

Whittaker, C.A., and R.O. Hynes. 2002. Distribution and evolution of

von Willebrand/integrin A domains: Widely dispersed domains

with roles in cell adhesion and elsewhere. Molecular Biology ofthe Cell 13: 3369–3387.

Wodarski, R., A.K. Clark, J. Grist, F. Marchand, and M. Malcangio.

2009. Gabapentin reverses microglial activation in the spinal

cord of streptozotocin-induced diabetic rats. European Journalof Pain 13: 807–811.

Xiao, W., A. Boroujerdi, G.J. Bennett, and Z.D. Luo. 2007. Chemo-

therapy-evoked painful peripheral neuropathy: Analgesic effects

of gabapentin and effects on expression of the alpha-2-delta type-1

calcium channel subunit. Neuroscience 144: 714–720.

Verkhratsky, A., and F. Kirchhoff. 2007. Glutamate-mediated neu-

ronal-glial transmission. Anat 210: 651–660.

Yaksh, T.L. 2006. Calcium channels as therapeutic targets in

neuropathic pain. Journal of Pain 7(1 Suppl 1): S13–S30.

Yamama, Y., K. Nishikawa, T. Funao, T. Mori, and A. Asada. 2010.

Intrathecal gabapentin and clonidine synergistically inhibit

allodynia in spinal nerve-ligated rats. Life Sciences 87: 565–571.

Yang, R.H., W.T. Wang, J.Y. Chen, R.G. Xie, and S.J. Hu. 2009.

Gabapentin selectively reduces persistent sodium current in

injured type-A dorsal root ganglion neurons. Pain 143: 48–55.

Yang, R.H., J.L. Xing, J.H. Duan, and S.J. Hu. 2005. Effects of

gabapentin on spontaneous discharges and subthreshold mem-

brane potential oscillation of type A neurons in injured DRG.

Pain 116: 187–193.

Yang, J.L., B. Xu, S.S. Li, W.S. Zhang, H. Xu, X.M. Deng, and Y.Q.

Zhang. 2012. Gabapentin reduces CX3CL1 signaling and blocks

spinal microglial activation in monoarthritic rats. MolecularBrain 5: 18.

Yeh, C.Y., S.C. Chung, F.L. Tseng, Y.C. Tsai, and Y.C. Liu. 2011.

Biphasic effects of chronic intrathecal gabapentin administration

on the expression of protein kinase C gamma in the spinal cord

of neuropathic pain rats. Acta Anaesthesiologica Taiwanica 49:

144–148.

Yi, H., M.A. Kim, S.K. Back, J.S. Eun, and H.S. Na. 2011. A novel

rat forelimb model of neuropathic pain produced by partial

injury of the median and ulnar nerves. European Journal of Pain15: 459–466.

Yoshizumi, M., J.C. Eisenach, and K. Hayashida. 2012a. Riluzole and

gabapentinoids activate glutamate transporters to facilitate

glutamate-induced glutamate release from cultured astrocytes.

European Journal of Pharmacology 677: 87–92.

Yoshizumi, M., R.A. Parker, J.C. Eisenach, and K. Hayashida. 2012b.

Gabapentin inhibits c-amino butyric acid release in the locus

coeruleus but not in the spinal dorsal horn after peripheral nerve

injury in rats. Anesthesiology 116: 1347–1353.

Zakrzewska, J.M., and R. McMillan. 2011. Trigeminal neuralgia: The

diagnosis and management of this excruciating and poorly

understood facial pain. Postgraduate Medical Journal 87:

410–416.

Gabapentin in neuropathic pain 251

123