HOME JOURNALS CONTACT

Journal of Medical Sciences

Year: 2010 | Volume: 10 | Issue: 4 | Page No.: 93-109
DOI: 10.3923/jms.2010.93.109
Diabetic Neuropathic Pain: An Update and Novel Pharmacological Strategies for Relief of Pain
R. Taliyan and P.L. Sharma

Abstract: Diabetic neuropathy is one of the most painful complications of diabetes mellitus, involving progressive neuronal damage and dysfunction and up to 30% of patients with diabetes mellitus developed diabetic neuropathy. Pain caused by diabetic neuropathy is debilitating and often is refractory to classical analgesic, including morphine. The mechanisms underlying cause of diabetic neuropathic pain are complex and both peripheral and central components of the sensory systems are reported to be involved in progression and maintenance of neuropathy. This study summarises data on pathogenesis and on existing and new analgesics such as NSAIDS, opioids, anti-epileptic, membrane stabilising and anti-depressant drugs, that are the mainstay of treatment for alleviating diabetic neuropathic pain. In addition, novel pharmacological approaches and strategies for analgesics such as use of drug combination and their implications will be discussed.

Fulltext PDF Fulltext HTML

How to cite this article
R. Taliyan and P.L. Sharma, 2010. Diabetic Neuropathic Pain: An Update and Novel Pharmacological Strategies for Relief of Pain. Journal of Medical Sciences, 10: 93-109.

Keywords: analgesics, antidepressant, opioids, anti-epileptic and Diabetic neuropathy

INTRODUCTION

Diabetes Mellitus (DM) is reported to affect more than 100 million people worldwide in 2006 and is projected to affect more than 350 million by 2030 (Wild et al., 2004). Painful Diabetic Neuropathy (PDN) may constitute a considerable clinical problem and a burdensome condition worldwide (Hoffman et al., 2009). Various neuropathic symptoms, including hyperalgesia, allodynia, hypoesthesia and spontaneous pain, often develop in early stages, but may occur at any stage (Ramos et al., 2007). These symptoms of painful diabetic neuropathy are highly unpleasant for the individuals and affect their quality of life (Hoffman et al., 2009; Connor’O, 2009). Various classes of drug are currently under investigation to treat neuropathic pain but still there is no gold standard therapeutic approach or treatment to manage this difficult to treat pain (James et al., 2008; Ziegler, 2010). Therefore, there is an urgent need to search for some novel strategies based approaches or new drugs for alleviating PDN, especially those with novel mechanisms of action where a combination of beneficial- rather than side-effect may be achieved. Pain caused by diabetic neuropathy is debilitating and often is refractory to classical analgesics, including morphine (Raghavendra et al., 2004). The mechanisms involved in genesis of diabetes -induced neuropathy are complex and poorly understood (Tesfaye, 2009; Ziegler, 2010). Accumulating evidence indicates that one of the major causes of diabetic peripheral neuropathy is oxidative stress (Ozkul et al., 2010), formation of advanced glycation end product (Sugimoto et al., 2008), increased flux through the polyol pathway that leads to accumulation of sorbitol and fructose (Chen et al., 2010a), myo-inositol depletion and reduction in Na+-K+-ATPase activity (Pop-Busui et al., 2010), deficits in neurotrophism leading to reduced expression and depletion of neurotrophic factors such as nerve growth factor (Stuart et al., 2000), neurotrophin-3 and insulin-like growth factor (Shimoshige et al., 2010), as well as alterations in axonal transport (Kuwabara and Misawa, 2008), PARP over-activation (Negi et al., 2010) (Fig. 1). In addition to abnormalities of peripheral afferent nerves, altered sensory processing in the spinal cord may contribute to the development of diabetic neuropathic pain (Loseth et al., 2008; Morgado et al., 2009). Other factors, such as upregulation of spinal excitatory glutamate receptors and increased release of glutamate and substance P, are also implicated in the development of spinal hypersensitivity in diabetes (Anjaneyulu et al., 2008).

Fig. 1: Hyperglycemia induced generation of mitochondrial ROS, sorbitol and formation of AGEs initiates a vicious circle by activating stress-sensitive pathways such as NF-κB, p38 MAPK and Jak/STAT, polyol (sorbitol) and hexosamine pathways, PKC and RAGE. Hyperglycemias-induced glycation of protein, lipid, nucleic acid, increased sorbitol and pro-inflammatory cytokines exerts a positive feedback on ROS and RNS synthesis and potentiates PKC-mediated vascular dysfunction by altering gene expression as well as vascular function and structure. MAPK: Mitogen Activated Protein Kinase; PKC: Protein Kinase-C; NO: Nitric Oxide; AGE/RAGE: Advanced Glycation End product (receptor); ROS: Reactive Oxygen Species; TGF: Transforming Growth Factor; PAI: Plasminogen Activation Inhibitor; Nf-κB: Nuclear factor kappa-B

Therefore, a thorough understanding of molecular mechanism based therapeutic options and of the likely benefits and potential adverse effects of each option should be considered.

PATHOGENESIS OF PDN

Advanced glycation end-products pathway: Long standing hyperglycaemia has been reported to be involved in the formation of Advanced Glycation End-products (AGEs) (Lukic et al., 2008; Yamagishi, 2009). AGEs are heterogeneous modified intracellular and extracellular bio-molecules formed via a non-enzymatic reaction between reducing sugars and amine residues on proteins, lipids, or nucleic acids (Barbosa et al., 2008). Extracellular protein AGEs include plasma and matrix proteins that disrupt cellular adhesion and activate the receptor for AGEs (RAGE) (Yan et al., 2010). Activation of RAGE or AGE-RAGE interaction induces oxidative stress (Yamagishi, 2009), Protein Kinase C (PKC) and the transcription of nuclear factor kappa B (NF-κB) (Bierhaus et al., 2001; Toth et al., 2008). NF-κB is a pleiotrophic gene regulator that regulates genes involved in promoting inflammatory reactions and neuronal dysfunction (Bierhaus et al., 2001; Haslbeck et al., 2005). Hyperglycaemia is reported to increase the levels of AGE and RAGE in diabetic patients (Lukic et al., 2008). The RAGE expression in the peripheral nervous system rises cumulatively in diabetes patients and relates to progressive pathological changes (Toth et al., 2008; Yan et al., 2010). Diabetic mice lacking RAGE showed significant improvement in PDN and diminished expression of NF-κB and PKC as compared to wild type diabetic model (Haslbeck et al., 2005; Cameron and Cotter, 2008). Collectively, the biochemical damage induced by AGEs results in increase ROS, impaired nerve blood flow and diminished neurotrophic support contributes to neuronal injury (Loseth et al., 2008; Yan et al., 2010).

The polyol pathway: Increased flux, through the polyol-pathway leading to multiple biochemical abnormalities in the diabetic nerve, is thought to play a significant role in the pathogenesis of diabetic neuropathy (Sango et al., 2006; Takafumi et al., 2008). In polyol pathway, glucose is converted into sorbitol by Aldose Reductase (AR) and sorbitol dehydrogenase oxidises, sorbitol to fructose (Maria, 2005). Nicotinamide Adenosine Dihydrogen Phosphare (NADPH) is consumed by aldose reductase-mediated reduction of glucose to sorbitol (Srinivasan et al., 2007) and NADPH is required for regeneration of antioxidant enzyme glutathione (GSH) thus deficient amount of glutathione contributes to oxidative stress (Kaneto et al., 2001). Moreover, conversion of glucose to sorbitol induced osmatic stress and to restore osmotic equilibrium to cell, other osmolytes, particularly taurine and myo-inositol, are effluxes from cells. Depletion of taurine and myo-inositol in nerve cells are implicated in PDN (Sima et al., 1997; Trevor et al., 2009) and supplementation of taurine and myo-inositol prevented neuropathic deficits (Pop-Busui et al., 2001; Trevor et al., 2009). On the other hand, excess formation of fructose in polyol pathway promotes advanced glycation end product as well as depletes NADPH, further augmenting Reactive Oxygen Species (ROS) mediated damage of cellular protein, lipid and neuron (Srinivasan et al., 2007; Sugimoto et al., 2008).

Aldose-Reductase Inhibitors (ARI), block the increased activity of aldose reductase, the rate-limiting enzyme that converts glucose to sorbitol (Ramirez and Borja, 2008), reduces sorbitol level implicated in PDN (Takafumi et al., 2008). Transgenic mice over-expressing aldose reductase in Schwann cells shown severe nerve conduction velocity deficit and oxidative stress under hyperglycaemic stress (Song et al., 2003). On the contrary, aldose reductase deficiency or inhibitors improves nerve conduction velocity deficits, Wallerian degeneration and nerve regeneration in diabetic animals (Chen et al., 2010b). The first trials of ARIs in diabetic neuropathy were carried out 20 years ago and offer attractive therapeutic option to treat PDN (Ramirez and Borja, 2008). Later on, various compounds have been evaluated such as alrestatin, sorbinil, ponalrestat, tolrestat, epalrestat, zopolrestat and zenarestat for the treatment of PDN (Bril et al., 2009; Shimoshige et al., 2009). Long-term treatment with ranirestat (AS-3201), a potent aldose reductase inhibitor, has been reported to suppress diabetic neuropathy and cataract formation in rats (Takafumi et al., 2008). However, clinical trials with ARIs discomfited and shown lack of efficacy and potential toxicity. Therefore, subsequent clinical evaluation of ARI such as sorbinil tolrestat, ponalrestat and zopolrestat were halted due to lack of efficacy in PDN (Brown et al., 2004; SRTRG, 1993). Epalrestat is the only ARI drug approved and marketed in Japan and India for PDN (Manish et al., 2009).

Hexosamine pathway: The hexosamine pathway is activated when excess metabolite of glycolysis accumulated and was implicated in diabetes-induced oxidative stress and complications (James et al., 2008). Fructose-6 phosphate is a metabolic intermediate of glycolysis. However, during glucose metabolism some fructose-6 phosphate is shunted from the glycolytic pathway to the hexosamine pathway and is converted to glucosamine-6 phosphate by glutamine fructose-6 phosphate amidotransferase (GFAT) (Srinivasan et al., 2007). The end-product of this pathway, UDP-N acetyl-glucosamine (UDP-GlcNAc), is a substrate for the glycation of important intracellular factors including transcription factor, thereby affecting the expression of many genes including Plasminogen Activator-Inhibitor (PA-I) and Transforming Growth Factors (TGF) and leads to diabetic microvascular complications (Du et al., 2000; Kaneto et al., 2001). Inhibition of GFAT block the transcription of TGF and PA-I and are shown beneficial effect in PDN (Maria, 2005; Srinivasan et al., 2007). In addition,the hexosamine biosynthesis inhibitor azaserine prevents endothelial inflammation and dysfunction under hyperglycemic condition through antioxidant effects (Angana et al., 2009).

Protein kinase C pathway: The Protein Kinase C (PKC) pathway is an additional mechanism implicated in hyperglycemia induced neuropathy (Evcimen and King, 2007). Increased glucose levels stimulate diacylglycerol (DAG), which in turn activates PKC and PKC-β isoform which is known to contribute in PDN (Geraldes and King, 2010). Selective inhibitors of PKC-β such as ruboxistaurin ameliorated several neuropathic deficits in experimental diabetic neuropathy (Carolina et al., 2007a; Danis and Sheetz, 2009). In a Phase II-clinical trial, ruboxistaurin at a dose of doses of 32-64 mg day-1 attenuated neuropathic symptoms and deficits, including overall neurologic examination and patient global assessment (Carolina et al., 2007b). We await the results of ruboxistaurin trials, a selective PKC-β inhibitor and the possibility of the future trials with other isoform selective PKC drugs for the treatment of PDN.

OXIDATIVE STRESS AND PDN

Increased oxidative stress is a unifying mechanism in the causation of PDN (Vincent et al., 2010). Antioxidant treatment have proven benefits in PDN (Pazdro and Burgess, 2010; Skalska et al., 2010). Hyperglycaemia induced activation of polyol, hexosamine pathway and formation of advanced glycation end products are known to increase the production of Reactive Oxygen Species (ROS) that contribute to nerve injury (Johansen et al., 2005; Friederich et al., 2009). In normal neuron, ROS production is tightly regulated. The free radical superoxide is generated by mitochondrial electron transfer chain when nicotinamide adenine dinucleotide (NADH) is oxidised to NAD+. Superoxide produced in diabetic and hyperglycemic conditions rapidly combines with NO and the formed peroxynitrite causes protein nitration or nitrosylation, lipid peroxidation, DNA damage and cell death and has direct toxic effects on the nerve tissue leading to neuropathic pain (Obrosova et al., 2007a; Drel et al., 2010). Mitochondria in neuron is sensitive to oxidative damage-which results impaired energy regulatory function that leads to loss of neuronal function and the development of PDN (Friederich et al., 2009). Moreover, excess generation of mitochondrial ROS due to hyperglycemia initiates a vicious circle by activating stress-sensitive pathways such as NF-κB, p38 MAPK, Jak/STAT, PKC and pro-inflammatory cytokines that contribute to diabetic complications (Fabio et al., 2001; Ziegler, 2008). In addition, hyperglycaemia increases the formation of potent oxidant peroxynitrite, which is formed by the combination of superoxide anion radical with nitric oxide and the formed peroxynitrite has been documented to play a key role in experimental and clinical diabetic neuropathy (Obrosova et al., 2007b; Drel et al., 2010). Peroxynitrite causes nitration and nitrosylation of biomolecules including proteins, lipids, DNA and has a direct toxic effect on neurones leading to diabetic complication (Obrosova et al., 2007b; Drel et al., 2010).

Poly (ADP-ribose) polymerase pathway (PARP): Oxido-nitrosative stress has been implicated in DNA single-strand breakage, followed by over activation of PARP, which contribute to PDN (Obrosova et al., 2007a; Negi et al., 2010). PARP is a nuclear enzyme that acts as a DNA-nick sensor and facilitates DNA repair. The PARP acts by cleaving Nicotinamide Adenine Dinucleotide (NAD+) to nicotinamide and ADP ribose residues attached to nuclear proteins (Maureen and Bonner-Weir, 1999). This result in NAD+ depletion and the metabolic pathway that depends upon NAD+ such as glycolysis and mitochondrial respiration are impaired. Further, depletion of NAD+ leads to changes in gene transcription and expression, increased free radical and oxidant concentration and diversion of glycolytic intermediates to other pathogenic pathways such as PKC, AGE formation and nitro-sative stress (Maureen and Bonner-Weir, 1999). The PARP inhibition or gene deficiency shown to counteract intraepidermal nerve fiber loss and neuropathic pain in advanced diabetic neuropathy (Olga et al., 2006; Obrosova, 2009). Moreover, concurrent targeting of nitrosative stress-PARP pathway has been reported to corrects functional, behavioral and biochemical deficits in experimental diabetic neuropathy (Negi et al., 2010).

Involvement of cytokines and chemokines: Hyperglycaemia is known to increase inflammatory mediators including C-reactive protein, interleukin-2 (IL-2), IL-6 and TNF-α in both type-1 and type-2 DM (Goldberg, 2009; Lawrence and Catherine, 2010). Higher levels of pro-inflammatory cytokines correlate with the incidence of neuropathy (Yu et al., 2009). Spinal proinflammatory cytokines such as TNF-α, IFNγ and Ils are powerful pain-enhancing signals that contribute to neuropathic pain (Juan and Carmen, 2008; Doupis et al., 2009). In addition, hyperglycaemia induced ROS and AGEs activate intracellular inflammatory signalling to up-regulate NF-κB (Iwasaki et al., 2007; Toth et al., 2008). NF-κB is a pleotrophic gene regulator that regulates the expression of various diffusible factors including cytokines (Cameron and Cotter, 2008). Pro-inflammatory cytokines are also known to increase the expression of inducible Nitric Oxide Synthatase (iNOS) and like cytokines (Malin et al., 1996; Koch et al., 2007). The iNOS both induces and is induced by NF-κB, leading to a vicious cycle of inflammation (Iwasaki et al., 2007; Drel et al., 2010). The NO generated by iNOS directly modulates the blood supply to nerves and participates in macro and microvascular changes following injury (Obrosova et al., 2007a, b; Vareniuk et al., 2008). The NO has direct role in axon and myelin breakdown following an injury (Pamela and Benjamin, 1998) and also contributes to the development of hyperalgesia and allodynia (Grover et al., 2000; Chen et al., 2010b). Genetic knockout nitric oxide synthase mice failed to display nerve-injury induced mechanical hypersensitivity (Guan et al., 2007). Thus cytokines and NO are seems to play a key role in initiation and maintenance of neuropathic pain and inhibition or blockage of cytokines is an attractive therapeutic approach in PDN (Yu et al., 2009). Long-term treatment of diabetic rats with cyclosporine, an inhibitor of interleukin-2, attenuated STZ-induced neuropathic pain (Taliyan et al., 2010a).

Pharmacological treatment of PDN based on their mechanism of action: Various classes of drugs are being examined and used for the treatment of neuropathic pain and strict glycaemic control is remains the best preventive measure for neuropathy (Ziegler et al., 2009). The Diabetes Control and Complications Trial (DCCT) has reported that strict glycaemic control in patients with DM not only decreased the incidence of neuropathy but also slowed its progression by 50-55% (DCCT, 1993; Pop-Busui et al., 2010). However, more than 30-40% patients are unable to achieve complete pain relief, even after glycaemic control (Kaye et al., 2003; Ziegler, 2008). Chronic hyperglycemia is associated with the loss of myelinated and unmyelinated fibers, Wallerian degeneration and blunted nerve-fiber reproduction (Pamela and Benjamin, 1998). Further, hyperglycaemia is known to attenuate motor nerve conduction velocity and Nerve Blood Flow (NBF) (Saini et al., 2004). The patho-physiologic mechanisms that underlie these changes are not clearly understood, however, various mechanisms has been proposed (Fig. 1), such as oxidative stress, the formation of sorbitol and advanced glycosylation end products, activation of PKC, increased proinflammatory cytokines and iNOS, that consequently leads a cascade of various signalling mechanism that contribute to PDN (Tesfaye, 2009; Ziegler, 2010). Therefore, a thorough understanding of molecular mechanism based therapeutic approach and of the likely benefits and potential adverse effects of each option should be considered. Newer agents have been designed to favorably influence the underlying process, rather than for symptomatic pain relief (Brian and Kathie, 2007). Approaches to prevention or treatment of diabetic neuropathy include the intensive treatment of hyperglycaemia, aldose reductase inhibition, anti-oxidant, AGE-inhibitor, cytokines inhibitors/antagonist and various symptomatic treatments (Chong and Brandner, 2006; Ziegler, 2010) (Table 1).

Anti-depressants and anti-epileptics: The first line therapy of drugs used to treat PDN are antidepressants (tricyclic antidepressants) (TCAs), anti-epileptics and Selective Serotonin Reuptake Inhibitors (SSRI) drug (Wong, 2008; Sibilia et al., 2009). The TCA was studied for neuropathic pain in late 1950 and are known to act by inhibiting serotonin and noradrenaline reuptake. The first medication studied in a randomized, controlled trial for the treatment of PDN was amitriptyline in late 1977, that led to a plethora of synthetic drugs and various TCA agents tested for the treatment of PDN (Saarto and Wiffen, 2007; Bansal et al., 2009). Several double blind, placebo controlled, crossover clinical trials have demonstrated the efficacy of the TCAs such as amitryptiline, imipramine, clomipramine and desipramine (Max et al., 1991; Sindrup et al., 2003). Among them, amitriptyline/nortriptyline and desipramine are found effective and are considered first choice TCAs for treating painful diabetic neuropathy (Gilron et al., 2009). Although, amitriptyline and desipramine relieve pain in many patients with painful diabetic neuropathy, side effects often preclude effective treatment (Max et al., 1991). Moreover, second generation antidepressant, SSRIs include fluoxetine, paroxetine, sertraline and citalopram, have not yet been FDA approved to treat painful neuropathy because they have been found to be no more efficacious than placebo in several controlled trials. On the contrary, venlafaxine, a third generation TCA has been demonstrated to produce significant pain relief as compared with placebo in a double blind, placebo controlled study (Rowbotham et al., 2004; Kadiroglu et al., 2008).

Table 1: Mechanism based pharmacological treatment for PDN
MOA: Mode of action: NA: Nor-adrenaline, 5-HT: 5-hydroxytriptaline (serotonin), *Approved for PDN

Duloxetine (Cymbalta) is the first antidepressant drug, which equally inhibit reuptake of serotonin and nor-adrenaline manufactured by Ely-lily, approved by the FDA in 2004 for the treatment of diabetic neuropathic pain and fibromyalgia (Acuna, 2008; Sultan et al., 2008). Moreover, duloxetine is equally effective in the treatment of diabetic peripheral neuropathic pain as two anticonvulsants, gabapentin and pregabalin, which are pharmacologically unrelated to duloxetine (Goldstein et al., 2005; Sibilia et al., 2009). However, TCA treatment is known to be associated with various side effects such as dry mouth, sweating, dizziness and sedation (Hall et al., 2010). In addition, recently, a retrospective study, including 58,956 person years follow-up on TCA therapy, indicates severe cardiac toxicity and TCAs are contraindicated in heart disease, epilepsy and glaucoma patients (Table 1).

Anti-epileptics: Antiepeleptic drugs originally developed for preventing seizure are now in broad use for the treatment of PDN (Sindrup et al., 2005). Carbamazepine was first anti-epileptic agents tested in late 1969 for neuopathic pain (Rull et al., 1969; Chakrabarti and Samantaray, 1976). Carbamazepine and phenytoin are known to block the voltage gated sodium channels and both reduced PDN as compared to placebo (Jia et al., 2006), however, due to various side effect, their use for the treatment of PDN is not recommended (Yamada et al., 2002). Lamotrigine is another anticonvulsant reported to produce favourable results in the treatment of PDN (Vinik et al., 2007). Lamotrigine has multiple actions, blockade of voltage gated sodium channels, decreased presynaptic calcium currents to inhibit the release of glutamate and increased GABA levels in the brain (Pop-Busui, 2007). Gabapentin and pregabalin, both produces analgesia via binding to the α2-δ site of L-type voltage gated calcium channels and decreasing calcium influx (Sibilia et al., 2009). Various multicentre double blind, randomized, placebo controlled trial, have demonstrated that extended gabapentin or gabapentin at a dose from 900 to 3600 mg day-1, significantly reduced pain of PDN compared with placebo (Sandercock et al., 2009; Chou et al., 2009). Pfizer has marketed an anti-epileptic agents, gabapentin, under the brand name of neurotonin an adjunct for seizure in late 1983 and the benefits of this drug for other conditions, like neuropathy, soon became known (Kenneth et al., 1999). Another drug also developed by Pfizer, pregabalin (Lyica), is an analogue of Gamma-Amino Butyric Acid (GABA) was approved by Food and Drug Administration (FDA) in 2005,for use in neuropathic pain associated with diabetic peripheral neuropathy (Beghi, 2004; Stump, 2009) (Table 1).

NSAIDs: Nonsteroidal anti-inflammatory drugs (NSAIDs) have also been used for the treatment of PDN (Kellogg et al., 2008). An increase level of cyclooxygenase-2 (COX-2) protein in the lumbar spinal cord and sciatic nerve has been demonstrated in diabetic patients that implicated in diabetes induced endoneurial nerve blood flow deficits (Pop-Busui et al., 2002; Matsunaga et al., 2007). Cox-2 selective inhibition and/or COX-2 gene inactivation provide protection against various neuropathy symptoms and deficits associated with PDN (Kellogg et al., 2007). In addition, pro-inflammatory cytokines and inflammatory mediators are known to increase the expression of inducible nitric oxide and COX-2 enzyme (Taliyan et al., 2010b). Concurrent targeting of both COX-2 and iNOS may provide better result for the treatment of PDN. A few studies have been demonstrated ameloiarative potential of combination of iNOS with selective COX-2 inhibitors in neuropathic pain in rats (Dudhgaonkar et al., 2008). However, the effectiveness of COX-2 inhibitors or COX-2 with iNOS inhibitor is not yet proven in clinical studies. Great caution must be taken, when NSAIDS are used for the treatment of PDN due to their detrimental effects to Gastro-Intestinal Tract (GIT), renal and cardiac functions.

Opioids: Opioids such as morphine, fantanyl, oxycodone are strong analgesics with proven efficacy to manage chronic pain (Pergolizzi et al., 2008; Riley et al., 2008). However, their use in treatment of PDN is still a matter of debate, some studies indicating usefulness of opioids in PDN (Canta et al., 2009; Attal et al., 2010), whereas some other studies indicating inadequate efficacy of opioids in PDN (Karci et al., 2004; Christoph et al., 2010). Long term treatment with opioids are associated with side effect including constipation, urinary retention, impaired cognitive function, impaired immune function and many other issues such as analgesic tolerance and addiction (Raghavendra et al., 2004; Taliyan et al., 2010b). However, long acting opioids such as fentanyl, methadone and oxycodone were effective in PDN (Vallerand, 2003; Hays et al., 2005; Riley et al., 2008). Recently, various randomized, placebo controlled study demonstrated that slow release oxycodone 80 mg day-1 significantly relieved PDN (Gimbel et al., 2003; Hanna et al., 2008) and improved health related quality of life (Watson et al., 2003). Moreover, numerous clinical studies reported that pregablin and morphine/oxycodone prescribed together is more efficacious than one or either drug alone for alleviating PDN (Gilron et al., 2005; Zin et al., 2010).

Tramadol: Tramadol is a fruitful result of extensive studies in search of a desirable molecule which act by multiple mechanisms and is associated with fewer adverse effects (Harati et al., 2000; Ebell, 2007). Tramadol is a centrally acting weak μ-opioid receptor agonist that also inhibits the reuptake of norephinephrine and 5-HT very much like TCA, leading to enhancement of endogenous pain inhibitory pathway (Tzschentke et al., 2006; Reeves and Burke, 2008). In a randomized placebo, 6 week controlled study, tramadol significantly reduced pain and enhanced quality of life as compared with placebo and was well tolerated in the management of painful diabetic neuropathy (Christoph et al., 2007; Freeman et al., 2007). In addition, the risk of abuse with tramadol is less than that with strong opioids analgesics such as morphine, oxycodone, methadone and fentanyl (Kazuhisa et al., 2009; Wade and Spruill, 2009). The US Food and Drug administration approved tramadol for the management of moderate to severe pain in Nov-2008 (Wade and Spruill, 2009).

Alpha-lipioc acid: Alpha-Lipoic Acid (ALA), an anti-oxidant, has been reported to relive pain associated with neuropathy in diabetic patients (Singh and Jialal, 2008). Alpha-lipoic acid used as a non-prescription dietary supplement for the treatment of PDN (Huang and Gitelman, 2008; Shay et al., 2009). A randomized controlled trial that compared once-daily oral doses of 600 to 1800 mg of ALA to placebo, demonstrated that ALA significantly reduced symptoms of neuropathy in patients (Ziegler et al., 2006; Liu et al., 2007). In a meta-analysis comprising 1,258 patients, infusions of ALA (600 mg i.v per day) over a 3 week period, improved neuropathic symptoms and deficits (Alexander et al., 2003; Liu et al., 2007). More recently, a clinical trial (Sydney trial-2) has demonstrated, that oral lipoic acid (600 mg kg-1 for 5 weeks) markedly attenuated PDN as compared with placebo (Ziegler et al., 2006). These study provide evidence that treatment with alpha-lipoic acid improves nerve conduction velocity deficits, endoneurial blood flow and nerve Na+ K+ ATPase activity in experimental diabetes and in humans (Alexander et al., 2003; Tankova et al., 2005) and may improve positive neuropathic sensory symptoms by improving the imbalance between increased oxidative stress and depleted antioxidant defense even in patients with poor glycemic control (Huang and Gitelman, 2008; Skalska et al., 2010). In addition, lipoic acid has fewer side effects than traditional treatments for neuropathy, such as gabapentin (Neurontin) and pregabalin (Lyrica) (Foster, 2007; Vallianou et al., 2009) and are approved for PDN treatment in Germany (Ziegler et al., 1999).

Topical analgesics: As depicted in Table 1, Numerious topical analgesics such as capsaicin (Zostrix), mexilitine, lidocaine 5% patch (Lidoderm) or gel, doxepin (Zonalon) (Lorna et al., 2004; Wong, 2008) and colindine gel (Campbell et al., 2009) are gaining in popularity among pain specialists and can offer pain relief in PDN without potential systemic toxicity (Wong, 2008; Wolffa et al., 2010).

Capsaicin: It has been recognized for almost 150 years that the topical application of the capsaicin produced pain relief (Lorna et al., 2004). It is now recognized that capsacin causes reversible depletion of the neurotransmitter Substance-P (SP) from the sensory nerve endings, which are involved in hypersensitivity of neuron to painful stimuli (hyperalgesia) (De Felipe et al., 1998). Topical application of capsaicin has been shown to reduce the pain of a variety of conditions, including post herpetic neuralgia (Watson et al., 1993), painful diabetic neuropathy (Capsaicin Study Group, 1992; Tandan et al., 1992) and osteoarthritis (Rains and Bryson, 1995; McCleane, 2000a). A meta-analysis of four randomized, double-blind, placebo-controlled trials of capsaicin (0.75%) in diabetic neuropathy found capsaicin more effective than placebo (McCleane and McLaughlin, 1998; Walker and McCleane, 2002). In addition, another double-blind study demonstrated that capsaicin was as effective as amitriptyline (Biesbroek et al., 1995). The major limitation of capsaicin is that of burning discomfort and poor patient compliance because of the need for frequent applications and redness at application site (Rains and Bryson, 1995). Recently, it was reported that the burning discomfort associated with application of capsaicin is reduced by addition of glyceryl trinitrate (GTN) to capsaicin and improved compliance (McCleane, 2000b). The GTN is known to have an anti-inflammatory effect and this may augment the analgesia when combined with capsaicin (McCleane and McLaughlin, 1998; McCleane, 2000b). In a double blind, placebo controlled trial of 40 volunteers; the burning discomfort associated with application of capsaicin cream (0.025%) was compared to placebo, GTN cream (1.33%) and to the combination of capsaicin cream (0.025%) plus GTN cream 1.33%. This study demonstrated that after single application the addition of GTN to capsaicin significantly reduced the burning discomfort associated with application of capsaicin alone (McCleane and McLaughlin, 1998; Walker and McCleane, 2002).

Miscellaneous pharmacological agents: A large number of pharmacological agents including levodopa (Ertas et al., 1998), baclofen (Anghinah et al., 1994), nerve growth factor (Stuart et al., 2000), gamma-linolenic acid (Hounsom et al., 1998), acetyl-L Carnintine (Evans, 2008), methylcobalamin (Sun et al., 2005), mexilitine (Oskarsson et al., 1997). NMDA-antagonist, i.e., dextromethorphan, cannabinoids, ketamine and memantine have been tested and shown promising results for the treatment of PDN (Sang et al., 2002; Chen et al., 2009). However, clinical data for these agents are still insufficient.

Novel pharmacological approaches to treat PDN: Inadequate pain relief in diabetic patients associated with painful neuropathy remains a major cause of suffering. According to DCCT, strict glycemic control remains the best available treatment option for PDN, although other treatments are in development (DCCT, 1993; Pop-Busui et al., 2010). Because of various mechanism involved in initiation and maintenance of neuropathic pain, a combined strategy is necessary to manage difficult to treat PDN (Eyigor et al., 2009; Obrosova, 2009). Recently, combination of analgesics which act by different modes of action such as an opioid and a non opioid was fostered and constitute a valid approach to the treatment of chronic pain, where a reduction of individual doses and consequently of side effects, could be very important (Ziegler, 2008; Varrassi et al., 2010). It has been demonstrated that combining of opioids with NMDA receptor antagonist (L-Dextromethorphan, or ketamine), alpha-2 receptor ligands such as gabapentin/pregablin (Hanna et al., 2008; Gilron et al., 2005) and antidepressant such as amitryptaline/nortriptyline provide adequate pain relief in PDN (Gilron et al., 2009). However, some issues associated with use of combination of different drug, remain to be explored, such as the safety of poly-pharmacy and its effect on compliance, while clinical trials suggest that any improvement in efficacy may be outweighed by the increased incidence of side effects (Hanna et al., 2008; Varrassi et al., 2010).

Opioids are strong analgesics and WHO has recommended the use of opioids for treatment of chronic pain (Boulton, 2005). However, addiction and development of analgesic tolerance to morphine in nerve injury or diabetes induced pain is the major hurdle in clinical practice (Raghavendra et al., 2004; Carolina et al., 2007b). Activation of N-methyl-D-aspartate (Wen et al., 2008; Lin et al., 2010), opioid receptor internalization/desensitization has been reported to play a key role in development of analgesic tolerance to morphine (Nishikawa et al., 2004). Hyperglycaemia induced activation of MAPK (Cui et al., 2006), PKC (Geraldes and King, 2010), NO (Salvemini and Neumann, 2010) and increased productions of pro-inflammatory cytokines (Doupis et al., 2009) are well reported mechanisms that contribute to morphine analgesic tolerance (Berger and Whistler, 2010).

Once tolerance to the analgesic effect of the opioids are observed and in order to avoid unnecessary further development of tolerance, simultaneous administration of other receptor mediated analgesics and opioids rotation of a more potent ligand such as methadone, fentanyl or oxycodone is advocated (Berger and Whistler, 2010). Although, tolerance development may result in use of staggering doses of opioids, there is no reason to evade the use of such strong analgesic agents. Development of analgesic tolerance could by handled by various approaches such as the concept of multimodal analgesia, consisting of the simultaneous use of analgesics with a different mode of action, NMDA receptor blocker/antagonist and low dose μ-opioid receptor antagonist, can counteract tolerance development (Elizabeth et al., 2010; Berger and Whistler, 2010). In addition, splenectomy in diabetic animals have been reported to restore analgesic effect of morphine (Taliyan et al., 2010c).

Non-pharmacological therapies: Several non-pharmacological approaches such as pysiotheraphy, acupuncture and Transcutaneous Electrical Nerve Stimulation (TENS) have been reported to be effective for pain relief (Kalra et al., 2007). Acupuncture as a management strategy for painful diabetic neuropathy is also supported by research (Ahn et al., 2007; Zhang et al., 2010). A pilot randomised clinical study has shown beneficial effect of acupuncture (traditional Chinese medicine versus Japanese acupuncture) in management of painful diabetic neuropathy (Ahn et al., 2007). The TENS is another non-pharmacological approach, shown beneficial effect in the management for DNP (Rader and Barry, 2006). The TENS provides a mild electrical stimulation through the application of surface electrodes over the painful area (Goodnick et al., 2000). The mild electrical current, generates heat that serves to relieve stiffness, improve mobility, was thought to relieve pain via release of endothelium dependent relaxing factor and endogenous opioids (Wei-Hua et al., 2004; Kalra et al., 2007). Similarly, interferential therapy (IFT) uses the strong physiological effects of low frequency electrical stimulation of nerves and has proven benefits in the managements of PDN.

CONCLUSIONS

Advancement in medical science offers great hope for the treatment of patients with diabetes and its complication and glycemic control is remains the best treatment for diabetes and/or diabetes induced complications including neuropathy (Pop-Busui et al., 2010). In addition, anticonvulsants such as gabapentin, pregablin and lamotrigine, antidepressants such as venlafaxine, duloxetine and amitryptaline, ion channel blockers, N-Methyl-D-Aspartic acid (NMDA) receptor antagonists, opioids and tramadol are used to manage diabetic complication but they merely provide symptomatic relief and do not modify the course of the disease (James et al., 2008; Ziegler et al., 2009; Varrassi et al., 2010). Therefore, identification of underlying mechanisms is of greatest need to better understand the failures with existing treatments and develop new approaches for diagnosis, prevention and managements of PDN. Combinations of opioids with non-opioid drugs have been demonstrated to offer benefits over single agent for treatment of PDN (Gilron et al., 2005). However, safety and tolerability of combination in clinical practice is yet to be proven. Anticonvulsants such as gabapentin and antidepressants such as duloxetine are the most successful pharmacological agents and are common treatment options for PDN, but evidence of the long-term efficacy of mono-therapy with these drugs in human is still lacking. Thus, developing a drug, which can act by multiple mechanism and/or modify the underlying pathogenesis of painful diabetic neuropathy, would meet a critical unmet need. Tramadol is perhaps the best example that has opioid analgesic (μ-agonist) and serotonergic properties, theoretically, make it an attractive medication for diabetic neuropathy (Reeves and Burke, 2008). Similarly, another agent AVP-923 (Neurodex), is a combination of dextromethorphan and an inhibitor of its metabolizing enzyme CYP2D6 quinidine, underwent a Phase III trial for PDN and demonstrated significantly better pain relief than placebo (Olney and Rosen, 2010). Although, many experimental and clinical studies with novel pharmacological agents have been performed to determine the most optimal strategy for preventing and treating diabetic neuropathy, but the pharmacologic treatment for PDN remains a challenge for the clinician. Safety and tolerability remains a major aspect in any treatment decision. More studies on different drug mechanism, drug combinations, are needed and ideally, non-pharmacological methods for attenuating PDN should also be included.

ACKNOWLEDGMENT

This study is dedicated, in found memory of, Late Prof Manjeet Singh. The author’s are also thankful to Mr. Parveen Garg for their valuable suggestion.

REFERENCES

  • Wild, S., G. Roglic, A. Green, R. Sicree and H. King, 2004. Global prevalence of diabetes: Estimates for the year 2000 and projections for 2030. Diabetes Care, 27: 1047-1053.
    CrossRef    PubMed    Direct Link    


  • Hoffman, D.L., A. Sadosky and J. Alvir, 2009. Cross-national burden of painful diabetic peripheral neuropathy in Asia, Latin America and the Middle East. Pain Pract., 9: 35-42.
    PubMed    


  • Ramos, K.M., Y. Jiang, C.I. Svensson and N.A. Calcutt, 2007. Pathogenesis of Spinally Mediated Hyperalgesia in Diabetes. Diabetes, 56: 1569-1576.
    CrossRef    


  • O'Connor, A.B., 2009. Neuropathic pain: Quality-of-life impact, costs and cost effectiveness of therapy. Pharmacoeconomics, 27: 95-112.
    CrossRef    PubMed    


  • Edwards, J.L., A.M. Vincent, H.T. Cheng and E.L. Feldman, 2008. Diabetic neuropathy: Mechanisms to management. Pharmacol. Ther., 120: 1-34.
    CrossRef    PubMed    Direct Link    


  • Ziegler, D., 2010. Can diabetic polyneuropathy be successfully treated? MMW Fortschr Med., 152: 64-68.
    PubMed    


  • Raghavendra, V., M.D. Rutkowski and J.A. DeLeo, 2004. Attenuation of morphine tolerance, withdrawal-induced hyperalgesia and associated spinal inflammatory immune responses by propentofylline in rats. Neuropsychopharmacology, 29: 327-334.
    CrossRef    


  • Tesfaye, S., 2009. Advances in the management of diabetic peripheral neuropathy. Curr. Opin Support Palliat Care., 3: 136-143.
    PubMed    


  • Ozkul, A., M. Ayhan, C. Yenisey, A. Akyol, E. Guney and F.A. Ergin, 2010. The role of oxidative stress and endothelial injury in diabetic neuropathy and neuropathic pain. Neuro Endocrinol. Lett., 31: 261-264.


  • Sugimoto, K., M. Yasujima and S. Yagihashi, 2008. Role of advanced glycation end products in diabetic neuropathy. Curr. Pharm. Dis., 14: 953-961.
    PubMed    


  • Eyigor, C., M. Uyar, S. Pirildar and M. Coker, 2009. Combination therapy in treatment of peripheral diabetic neuropathy with severe pain in an adolescent patient. Paediatr Anaesth., 19: 193-194.
    PubMed    


  • Pop-Busui, R., K.A. Sullivan, C. van Huysen, L. Bayer, X. Cao, R. Towns, M.J. Stevens, 2001. Depletion of taurine in experimental diabetic neuropathy: Implications for nerve metabolic, vascular and functional deficits. Exp. Neurol., 168: 259-272.
    PubMed    


  • Stuart, C.A., S. Sherwin, T.A. Bruce, R. Freeman and B. Victor et al., 2000. Polyneuropathy: A randomized controlled trial growth factor in patients with diabetic polyneuropathy. JAMA., 284: 2215-2221.
    CrossRef    


  • Shimoshige, Y., R. Enomoto,T. Aoki, N. Matsuoka and S. Kaneko, 2010. The involvement of aldose reductase in alterations to neurotrophin receptors and neuronal cytoskeletal protein mRNA levels in the dorsal root ganglion of streptozotocin-induced diabetic rats. Biol. Pharm. Bull., 33: 67-71.
    PubMed    


  • Kuwabara, S. and S. Misawa, 2008. Pharmacologic intervention in axonal excitability: In vivo assessment of nodal persistent sodium currents in human neuropathies. Curr. Mol. Pharmacol., 1: 61-67.
    PubMed    


  • Negi, G., A. Kumar and S.S. Sharma, 2010. Concurrent targeting of nitrosative stress-PARP pathway corrects functional, behavioral and biochemical deficits in experimental diabetic neuropathy. Biochem. Biophys. Res. Commun., 391: 102-106.
    PubMed    


  • Loseth, S., E. Stalberg, R. Jorde and S.I. Mellgren, 2008. Early diabetic neuropathy: Thermal thresholds and intraepidermal nerve fibre density in patients with normal nerve conduction studies. J. Neurol., 255: 1197-1202.
    PubMed    


  • Morgado, C., P.P. Terra and I. Tavares, 2009. Neuronal hyperactivity at the spinal cord and periaqueductal grey during painful diabetic neuropathy: Effects of gabapentin. Eur. J. Pain.,


  • Anjaneyulu, M., A. Berent-Spillson and J.W. Russell, 2008. Metabotropic glutamate receptors (mGluRs) and diabetic neuropathy. Curr. Drug Targets, 9: 85-93.
    PubMed    


  • Lukic, I.K., P.M. Humpert, P.P. Nawroth and A. Bierhaus, 2008. The RAGE pathway: Activation and perpetuation in the pathogenesis of diabetic neuropathy. Ann. N. Y. Acad. Sci., 1126: 76-80.
    PubMed    


  • Yamagishi, S., 2009. Advanced glycation end products and receptor-oxidative stress system in diabetic vascular complications. Ther. Apher. Dial., 13: 534-539.
    PubMed    


  • Barbosa, J.H., S.L. Oliveira and L.T. Seara, 2008. The role of advanced glycation end-products (AGEs) in the development of vascular diabetic complications. Arq. Bras. Endocrinol. Metabol., 52: 940-950.
    PubMed    


  • Yan, S.F., R. Ramasamy and A.M. Schmidt, 2010. The RAGE axis: A fundamental mechanism signaling danger to the vulnerable vasculature. Circ. Res., 106: 842-853.
    PubMed    


  • Toth, C., L.L. Rong, C. Yang, J. Martinez and F. Song et al., 2008. Receptor for advanced glycation end products (RAGEs) and experimental diabetic neuropathy. Diabetes, 57: 1002-1017.
    CrossRef    


  • Bierhaus, A., S. Schiekofer, M. Schwaninger, M. Andrassy and P.M. Humpert et al., 2001. Diabetes-associated sustained activation of the transcription factor nuclear factor-κB. Diabetes, 50: 2792-2808.
    CrossRef    Direct Link    


  • Haslbeck, K.M., E. Schleicher, A. Bierhaus, P. Nawroth, M. Haslbeck, B. Neundorfer and D. Heuss, 2005. The AGE/RAGE/NF-κB pathway may contribute to the pathogenesis of polyneuropathy in impaired glucose tolerance (IGT). Exp. Clin. Endocrinol. Diabetes, 113: 288-291.
    CrossRef    


  • Cameron, N.E. and M.A. Cotter, 2008. Pro-inflammatory mechanisms in diabetic neuropathy: Focus on the nuclear factor kappa B pathway. Curr. Drug Targets, 9: 60-67.
    PubMed    


  • Sango, K., T. Suzuki, H. Yanagisawa, S. Takaku, H. Hirooka, M. Tamura and K. Watabe, 2006. High glucose-induced activation of the polyol pathway and changes of gene expression profiles in immortalized adult mouse Schwann cells IMS3. J. Neurochem., 98: 446-458.
    PubMed    


  • Takafumi, M., O. Yoshiyuki, K. Akemi, T. Kaoru, U. Yoshinaka and B. Vera, 2008. Long-term treatment with ranirestat (AS-3201), a potent aldose reductase inhibitor, suppresses diabetic neuropathy and cataract formation in rats. J. Pharmacol. Sci., 107: 340-348.
    CrossRef    


  • Srinivasan, V., N. Sandhya, R. Sampathkumar, S. Farooq, V. Mohan and M. Balasubramanyam, 2007. Glutamine fructose-6-phosphate amidotransferase (GFAT) gene expression and activity in patients with type 2 diabetes: Inter-relationships with hyperglycaemia and oxidative stress. Clin. Biochem., 40: 952-957.
    PubMed    


  • Maria, G.B., 2005. Hexosamines, insulin resistance and the complications of diabetes: current status. Am. J. Physiol. Endocrinol. Metabial., 290: E1-E8.


  • Sima, A.A., A.J. Dunlap, P.E. Davidson, J.T. Wiese, L.R. Lightle, A.D. Greene and A.M. Yorek, 1997. Supplemental myo-inositol prevents L-fucose-induced diabetic neuropathy. Diabetese, 46: 301-306.
    CrossRef    


  • Trevor, A., Z. Wei, C.E. Margaret and J.S. Martin, 2009. Oxidative stress and dysregulation of the taurine transporter in high-glucose-exposed human Schwann cells: Implications for pathogenesis of diabetic neuropathy. Am. J. Physiol. Endocrinol. Metabial., 297: E620-E628.


  • Song, Z., D.T. Fu, Y.S. Chan, S. Leung, S.S. Chung and S.K. Chung, 2003. Transgenic mice overexpressing aldose reductase in Schwann cells show more severe nerve conduction velocity deficit and oxidative stress under hyperglycemic stress. Mol. Cell Neurosci., 23: 638-647.
    PubMed    


  • Ramirez, M.A. and N.L. Borja, 2008. Epalrestat: An aldose reductase inhibitor for the treatment of diabetic neuropathy. Pharmacotherapy, 28: 646-655.
    PubMed    


  • Bril, V., T. Hirose, S. Tomioka, R. Buchanan and Ranirestat Study Group, 2009. Ranirestat for the management of diabetic sensorimotor polyneuropathy. Diabetes Care, 32: 1256-1260.
    PubMed    


  • Shimoshige, Y., K. Minoura, N. Matsuoka, S. Takakura, S. Mutoh and M. Kamijo, 2009. Thirteen-month inhibition of aldose reductase by zenarestat prevents morphological abnormalities in the dorsal root ganglia of streptozotocin-induced diabetic rats. Brain Res., 1247: 182-187.
    PubMed    


  • Brown, M.J., S.J. Bird, S. Watling, H. Kaleta and L. Hayes et al., 2004. Natural progression of diabetic peripheral neuropathy in the Zenarestat study population. Diabetes Care, 27: 1153-1159.
    CrossRef    


  • SRTRG., 1993. The sorbinil retinopathy trial: Neuropathy results. Neurology, 43: 1141-1149.
    Direct Link    


  • Manish, M., R. Girish, N. Venkataswamy, R.S. Hariharan and S.R. Lohati, 2009. Efficacy, safety and tolerability of Epalrestat compared to Methylcobalamine in patients with diabetic neuropathy. Int. J. Dev. Countries, 29: 28-34.


  • Kaneto, H., G. Xu, K.H. Song, K. Suzuma, S. Bonner-Weir, A. Sharma and G.C. Weir, 2001. Activation of the hexosamine pathway leads to deterioration of pancreatic β-cell function through the induction of oxidative stress. J. Biol. Chem., 276: 31099-31104.
    CrossRef    Direct Link    


  • Du, X.L., D. Edelstein, L. Rossetti, I.G. Fantus and H. Goldberg et al., 2000. Hyperglycemia-induced mitochondrial su-peroxide overproduction activates the hexosamine pathway and induces plasminogen activator inhibitor-1 expression by increasing Sp1 glycosylation. Proc. Natl. Acad. Sci. USA., 97: 12222-12226.
    Direct Link    


  • Angana, G.R., M. Xi-Fen, M.C. Joao and Y. Zhihong, 2009. The hexosamine biosynthesis inhibitor azaserine prevents endothelial inflammation and dysfunction under hyperglycemic condition through antioxidant effects. Am. J. Physiol. Heart Circ. Physiol., 296: H815-H822.
    CrossRef    


  • Evcimen, N.D and G.L. King, 2007. The role of protein kinase C activation and the vascular complications of diabetes. Pharmacol. Res., 55: 498-510.
    CrossRef    PubMed    Direct Link    


  • Geraldes, P. and G.L. King, 2010. Activation of protein kinase C isoforms and its impact on diabetic complications. Circ. Res., 106: 1319-1331.
    PubMed    


  • Danis, R.P. and M.J. Sheetz, 2009. Ruboxistaurin: PKC-beta inhibition for complications of diabetes. Expert Opin Pharmacother., 10: 2913-2925.
    PubMed    


  • Carolina, M.C., M.B. Patricia, L.R. Amanda, C. Melissa and S. Kathryn et al., 2007. A 6-Month, randomized, double-masked, placebo-controlled study evaluating the effects of the protein kinase c-β inhibitor ruboxistaurin on skin microvascular blood flow and other measures of diabetic peripheral neuropathy. Diabeties Care, 30: 896-902.
    CrossRef    


  • Vincent, A.M., J.L. Edwards, L.L. McLean, Y. Hong and F. Cerri et al., 2010. Mitochondrial biogenesis and fission in axons in cell culture and animal models of diabetic neuropathy. Acta Neuropathol.,


  • Pazdro, R. and J.R. Burgess, 2010. The role of vitamin E and oxidative stress in diabetes complications. Mech. Ageing Dev., 131: 276-286.
    PubMed    


  • Skalska, S., P. Kucera, Z. Goldenberg, M. Stefek and Z. Kyselova et al., 2010. Neuropathy in a rat model of mild diabetes induced by multiple low doses of streptozotocin: Effects of the antioxidant stobadine in comparison with a high-dose α-lipoic acid treatment. Genet. Physiol. Biophys., 29: 50-58.
    PubMed    


  • Johansen, J.S., A.K. Harris, D.J. Rhychly and A. Ergul, 2005. Oxidative stress and the use of antioxidants in diabetes: Linking basic science to clinical practice. Cardiovasc. Diabetol., 4: 5-9.
    CrossRef    


  • Friederich, M., P. Hansell and F. Palm, 2009. Diabetes, oxidative stress, nitric oxide and mitochondria function. Curr. Diabetes Rev., 5: 120-144.
    PubMed    


  • Obrosova, I.G., V.R. Drel, C.L. Oltman, N. Mashtalir and J. Tibrewala et al., 2007. Role of nitrosative stress in early neuropathy and vascular dysfunction in streptozotocin-diabetic rats. Am. J. Physiol. Endocrinol. Metab., 293: E1645-E1655.
    CrossRef    PubMed    Direct Link    


  • Drel, V.R., S. Lupachyk, H. Shevalye, I. Vareniuk and W. Xu et al., 2010. New therapeutic and biomarker discovery for peripheral diabetic neuropathy: PARP inhibitor, nitrotyrosine and tumor necrosis factor α. Endocrinology, 151: 2547-2555.
    CrossRef    


  • Fabio, F., L. Annarosa, C. Daniela, L. Federica and S. Bijan et al., 2001. Hyperglycemia activates p53 and p53-regulated genes leading to myocyte cell death. Diabetes, 50: 2363-2375.
    CrossRef    


  • Maureen, J. C and S. Bonner-Weir, 1999. Implicating PARP and NAD+ depletion in type I diabetes. Nature Med., 5: 269-270.
    CrossRef    


  • Ilnytska, O., V.V. Lyzogubov, M.J. Stevens, V.R. Drel and N. Mashtalir et al., 2006. Poly(ADP-Ribose) polymerase inhibition alleviates experimental diabetic sensory neuropathy. Diabetes, 55: 1686-1694.
    CrossRef    Direct Link    


  • Obrosova, I.G., W. Xu, V.V. Lyzogubov, O. Ilnytska and N. Mashtalir et al., 2007. PARP inhibition or gene deficiency counteracts intraepidermal nerve fiber loss and neuropathic pain in advanced diabetic neuropathy. Free Radic. Biol. Med., 44: 972-978.
    PubMed    


  • Goldberg, R.B., 2009. Cytokine and cytokine-like inflammation markers, endothelial dysfunction, and imbalanced coagulation in development of diabetes and its complications. J. Clin. Endocrinol. Metabial., 94: 3171-3182.
    CrossRef    PubMed    


  • Lawrence, L. and M.C. Catherine, 2010. TNF-α and neuropathic pain-a review, J. Neuroinflammation, 7: 27-27.
    CrossRef    


  • Yu, L.N., X.S. Yang, Z. Hua and W. Xie, 2009. Serum levels of pro-inflammatory cytokines in diabetic patients with peripheral neuropathic pain and the correlation among them. Zhonghua Yi Xue Za Zhi, 89: 469-471.
    PubMed    


  • Navarro-Gonzalez, J.F. and C. Mora-Fernandez, 2008. The role of inflammatory cytokines in diabetic nephropathy. J. Am. Soc. Nephrol., 19: 433-442.
    CrossRef    


  • Doupis, J., T.E. Lyons, S. Wu, C. Gnardellis, T. Dinh and A. Veves, 2009. Microvascular reactivity and inflammatory cytokines in painful and painless peripheral diabetic neuropathy. J. Clin. Endocrinol. Metab., 94: 2157-2163.
    CrossRef    PubMed    Direct Link    


  • Iwasaki, Y., M. Kambayashi, M. Asai, M. Yoshida, T. Nigawara and K. Hasimoto, 2007. High glucose alone, as well in combination with proinflammatory cytokines, stimulates nuclear factor κB-mediated transcription in hepatocytes in vivo. J. Diabetes Complications, 21: 56-62.
    CrossRef    


  • Malin, F., W. Nils and L.E. Decio, 1996. Cytokines activate the nuclear factor κB (NF-κB) and induce nitric oxide production in human pancreatic islets. FEBS Lett., 385: 4-6.
    CrossRef    


  • Koch, A., K. Zacharowski, O. Boehm, M. Stevens and P. Lipfert et al., 2007. Nitric oxide and pro-inflammatory cytokines correlate with pain intensity in chronic pain patients. Biomed. Life Sci., 56: 32-37.
    CrossRef    


  • Vareniuk, I., I.A. Pavlov and I.G. Obrosova, 2008. Inducible nitric oxide synthase gene deficiency counteracts multiple manifestations of peripheral neuropathy in a streptozotocin-induced mouse model of diabetes. Diabetologia, 51: 2126-2133.
    PubMed    


  • Chen, Y., M.K. Boettger, A. Reif, A. Schmitt, N. Uceyler and C. Sommer, 2010. Nitric oxide synthase modulates CFA-induced thermal hyperalgesia through cytokine regulation in mice. Mol. Pain, 63: 13-13.
    CrossRef    


  • Grover, V.S., A. Sharma and M. Singh, 2000. Role of nitric oxide in diabetes-induced attenuation of antinociceptive effect of morphine in mice. Eur. J. Pharmacol., 399: 161-164.
    CrossRef    PubMed    Direct Link    


  • Chen, Y.S., S.S. Chung and S.K. Chung, 2010. Aldose reductase deficiency improves Wallerian degeneration and nerve regeneration in diabetic thy1-YFP mice. J. Neuropathol. Exp. Neurol., 69: 294-305.
    PubMed    


  • Guan, Y., M. Yaste, S.N. Raja and Y.X. Tao, 2007. Genetic knockout and pharmacologic inhibition of neuronal nitric oxide synthase attenuate nerve injury-induced mechanical hypersensitivity in mice. Mol. Pain, 3: 29-29.
    CrossRef    


  • Taliyan, R., M. Singh and P.L. Sharma, 2010. Beneficial effect of cyclosporine in experimental diabetes induced neuropathic pain in rats. Int. J. Pharmacol., 6: 393-399.
    CrossRef    Direct Link    


  • Ziegler, D., W. Rathmann, T. Dickhaus, C. Meisinger and A. Mielck and KORA Study Group, 2009. Neuropathic pain in diabetes,prediabetes and normal glucose tolerance: the MONICA/KORA Augsburg Surveys S2 and S3. Pain Med., 10: 393-400.
    PubMed    


  • Pop-Busui, R, W.H. Herman, E.L. Feldman, P.A. Low and C.L. Martin et al., 2010. DCCT and EDIC studies in type 1 diabetes: lessons for diabetic neuropathy regarding metabolic memory and natural history. Curr. Diabetes Rep.,


  • DCCT Research Group, 1993. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N. Engl. J. Med., 329: 977-986.
    CrossRef    PubMed    Direct Link    


  • Kaye, G., A. Wollitzer and L. Jovanovic, 2003. Comfort and support improve painful diabetic neuropathy, whereas disappointment and frustration deteriorate the metabolic and neuropathic status despite an intensive diabetes care program. Diabetes Care, 26: 2470-2480.


  • Ziegler, D., 2008. Treatment of diabetic neuropathy and neuropathic pain: How far have we come? Diabetes Care, 31: S255-S261.
    CrossRef    PubMed    Direct Link    


  • Pamela, B.J. and B.G. Benjamin, 1998. Increased vulnerability to demyelination in streptozotocin diabetic rats. J. Comparative Neurol., 373: 55-61.
    CrossRef    


  • Saini, A.K., K.H.S. Arun, C.L. Kaul and S.S. Sharma, 2004. Acute hyperglycemia attenuates nerve conduction velocity and nerve blood flow in male sprague-dawley rats: Reversal by adenosine. Pharmacol. Res., 50: 593-599.
    PubMed    


  • Brian, C.C. and L.H. Kathie, 2007. Current concepts in targeted therapies for the pathophysiology of diabetic microvascular complication. Vascular Health Risk Manage., 3: 823-832.
    Direct Link    


  • Chong, M.S. and B. Brandner, 2006. Neuropathic agents and pain; New strategise. Biomed. Pharmacother., 60: 318-332.


  • Wong, M.C., 2008. TCAs, anticonvulsants, opioids and capsaicin cream are effective for diabetic neuropathy. Evid Based Med., 13: 21-21.
    CrossRef    


  • Sibilia, Q., C. Jeremy, L. Mickael, S. Dominique and S. Gerard et al., 2009. Meta-analysis of duloxetine vs. pregabalin and gabapentin in the treatment of diabetic peripheral neuropathic pain. BMC Neurol., 9: 6-6.
    CrossRef    


  • Saarto, T. and P.J. Wiffen, 2007. Antidepressants for neuropathic pain. Cochrane Database Syst. Rev.,
    CrossRef    


  • Bansal, D., A. Bhansali, D. Hota, A. Chakrabarti and P. Dutta, 2009. Amitriptyline vs. pregabalin in painful diabetic neuropathy: A randomized double blind clinical trial. Diabetic Med., 26: 1019-1026.
    CrossRef    


  • Max, M.B., R.K. Kishore, S.C. Schafer, B. Meister, R.H. Gracely, B. Smoller and R. Dubner, 1991. Efficacy of desipramine in painful diabetic neuropathy: a placebo-controlled trial. Pain, 45: 3-9.
    PubMed    


  • Sindrup, S.H., F.W. Bach, C. Madsen, L.F. Gram and T.S. Jensen, 2003. Venlafaxine versus imipramine in painful polyneuropathy, a randomized, controlled trial. Neurology, 60: 1284-1289.
    Direct Link    


  • 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.
    PubMed    


  • Rowbotham, M.C., V. Goli, N.R. Kunz and D. Lei, 2004. Venlafaxine extended release in the treatment of painful diabetic neuropathy: a double-blind, placebo-controlled study. Pain, 110: 697-706.
    PubMed    


  • Kadiroglu, A.K., D. Sit, H. Kayabasi, A.K. Tuzcu, N. Tasdemir and M.E. Yilmaz, 2008. The effect of venlafaxine HCl on painful peripheral diabetic neuropathy in patients with type 2 diabetes mellitus. J. Diabetes Complications, 22: 241-245.
    PubMed    


  • Acuna, C., 2008. Duloxetine for the treatment of fibromyalgia. Drugs Today, 44: 725-734.
    CrossRef    PubMed    


  • Sultan, A., H. Gaskell, S. Derry and R.A. Moore, 2008. Duloxetine for painful diabetic neuropathy and fibromyalgia pain: Systematic review of randomised trials. BMC Neurol., 8: 29-29.
    CrossRef    


  • Goldstein, D.J., Y. Lu, M.J. Detke, T.C. Lee and S. Iyengar, 2005. Duloxetine vs. placebo in patients with painful diabetic neuropathy. Pain, 116: 109-118.
    PubMed    


  • Hall, J.A., F. Wang, T.M. Myers Oakes, B.G. Utterback, A.Crucitti and N. Acharya, 2010. Safety and tolerability of duloxetine in the acute management of diabetic periphera neuropathic pain: Analysis of pooled data from three placebo-controlled clinical trials. Expert Opin Drug Saf.,


  • Sindrup, S.H., M. Otto, N.B. Finnerup and T.S. Jensen, 2005. Antidepressants in the treatment of neuropathic pain. Basic Clin. Pharmacol. Toxicol., 96: 399-409.
    PubMed    


  • Rull, J.A., R. Quibrera, H. Gonzalez-Millan and O. Lozano Castaneda, 1969. Symptomatic treatment of peripheral diabetic neuropathy with carbamazepine (Tegretol®): Double blind crossover trial. Diabetologia, 5: 215-218.
    CrossRef    


  • Chakrabarti, A.K. and S.K. Samantaray, 1976. Diabetic peripheral neuropathy: nerve conduction studies before, during and after carbamazepine therapy. Aust. N. Z. J. Med., 6: 565-568.
    PubMed    


  • Jia, H.Y., L. Qi-Fu, P. Song, A.N. Zhen-Mei and Y.P. Liu et al., 2006. Effects of venlafaxine and carbamazepine for painful peripheral diabetic neuropathy: A randomized, double-blind and doubledummy, controlled multi-center trial chin. J. Evid-Based Med., 6: 321-327.


  • Yamada, N., K. Kaneko, K. Saito and I. Tatsuno, 2002. Anticonvulsant hypersensitivity syndrome with marked eosinophilia in treatment of diabetic neuropathy. Diabetes Care, 25: 1099-1100.
    CrossRef    


  • Vinik, A.I., M. Tuchman, B. Safirstein, C. Corder and L. Kirby et al., 2007. Lamotrigine for treatment of pain associated with diabetic neuropathy: Results of two randomized, double-blind, placebo-controlled studies. Pain, 128: 169-179.
    CrossRef    


  • Pop-Busui, R., 2007. Does lamotrigine alleviate the pain in diabetic neuropathy?. Nature Rev. Neurol., 3: 424-425.
    CrossRef    


  • 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 tolerability in a double-blind, randomized, controlled clinical trial. Diabetes Care, 32: e20-e20.
    PubMed    


  • 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. J. Genet. Int. Med., 24: 178-188.
    PubMed    


  • Kenneth, C.G., S. Cecilia, H. Robert and H.R. Allan, 1999. Gabapentin in the treatment of painful diabetic neuropathy: A placebo controlled, double blind, crossover trial. J. Neurol. Neurosurg Psychiatry, 66: 251-252.
    CrossRef    


  • Beghi, E., 2004. Efficacy and tolerability of the new antiepileptic drugs: Comparison of two recent guidelines. Lancet Neurol., 3: 618-621.
    PubMed    


  • Stump, P., 2009. Pregabalin-profile of efficacy and tolerability in neuropathic pain. Drugs Today, 45: 19-27.
    PubMed    


  • Kellogg, A.P., H.T. Cheng and R. Pop-Busui, 2008. Cyclooxygenase-2 pathway as a potential therapeutic target in diabetic peripheral neuropathy. Curr. Drug Targets, 9: 68-76.
    PubMed    


  • Taliyan, R., M. Singh and P.L. Sharma, 2010. Possible mechanism of hyperglycemia induced decrease antinociceptive effect of analgesics in Rats. IJPSR., 1: 99-107.
    Direct Link    


  • Pop-Busui, R., V. Marinescu, C. van Huysen, F. Li and K. Sullivan et al., 2002. Dissection of metabolic, vascular and nerve conduction interrelationships in experimental diabetic neuropathy by cyclooxygenase inhibition and acetyl-l-carnitine administration. Diabetese, 51: 2619-2628.
    CrossRef    


  • Kellogg, A.P., T.D. Wiggin, D.D. Larkin, J.M. Hayes, M.J. Stevens and R. Pop-Busui, 2007. Protective effects of cyclooxygenase-2 gene inactivation against peripheral nerve dysfunction and intraepidermal nerve fiber loss in experimental diabetes. Diabetes, 56: 2997-3005.
    CrossRef    


  • Matsunaga, A., M. Kawamoto, S. Shiraishi, T. Yasuda, S. Kajiyama, S. Kurita and O. Yuge, 2007. Intrathecally administered COX-2 but not COX-1 or COX-3 inhibitors attenuate streptozotocin-induced mechanical hyperalgesia in rats. Eur. J. Pharmacol., 554: 12-17.
    PubMed    


  • Dudhgaonkar, S.P., S.K. Tandan, D. Kumar, R. Arunadevi and V.R. Prakash, 2008. Synergistic interaction between meloxicam and aminoguanidine in formalin-induced nociception in mice. Eur. J. Pain, 12: 321-328.
    PubMed    


  • Pergolizzi, J., R.H. Boger, K. Budd, A. Dahan and S. Erdine et al., 2008. Opioids and the management of chronic severe pain in the elderly: consensus statement of an International Expert Panel with focus on the six clinically most often used World Health Organization Step III opioids (buprenorphine, fentanyl, hydromorphone, methadone, morphine, oxycodone). Pain Pract., 8: 287-313.
    PubMed    


  • Riley, J., E. Eisenberg, G. Muller-Schwefe, A.M. Drewes and L. Arendt-Nielsen, 2008. Oxycodone: A review of its use in the management of pain. Curr. Med. Res. Opin, 24: 175-192.
    PubMed    


  • Canta, A., A. Chiorazzi, C. Meregalli, V. Carozzi and N. Oggioni et al., 2009. Continuous buprenorphine delivery effect in streptozotocine-induced painful diabetic neuropathy in rats. J. Pain, 10: 961-968.
    CrossRef    PubMed    Direct Link    


  • Attal, N., G. Cruccu, R. Baron, M. Haanpaa, P. Hansson, T.S. Jensen and T. Nurmikko, 2010. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2009 revision. Eur. J. Neurol.,
    CrossRef    


  • Karci, A., A. Tasdogen, Y. Erkin, G. Aktaş and Z. Elar, 2004. The analgesic effect of morphine on postoperative pain in diabetic patients. Acta Anaesthesiol. Scand., 48: 619-624.
    CrossRef    


  • Christoph, T., J. de Vry and T.M. Tzschentke, 2010. Tapentadol, but not morphine, selectively inhibits disease-related thermal hyperalgesia in a mouse model of diabetic neuropathic pain. Neurosci. Lett., 470: 91-94.
    PubMed    


  • Hays, L., C. Reid, M. Doran and K. Geary, 2005. se of methadone for the treatment of diabetic neuropathy. Diabetes Care, 28: 485-487.
    PubMed    


  • Gimbel, J.S., P. Richards and R.K. Portenoy, 2003. Controlled-release oxycodone for pain in diabetic neuropathy: A randomized controlled trial. Neurology, 60: 927-934.
    PubMed    


  • Watson, C.P., D. Moulin, J. Watt-Watson, A. Gordon and J. Eisenhoffer, 2003. Controlled-release oxycodone relieves neuropathic pain: A randomized controlled trial in painful diabetic neuropathy. Pain, 105: 71-78.
    PubMed    


  • Hanna, M., G. O'Brien and M.C. Wilson, 2008. Prolonged-release oxycodone enhances the effects of existing gabapentin therapy in painful diabetic neuropathy patients. Eur. J. Pain, 12: 804-813.


  • Vallerand, A.H., 2003. The use of long-acting opioids in chronic pain management. Nurs Clin. North Am., 38: 435-445.
    PubMed    


  • Gilron, I., J.M. Bailey, Tu D, R.R. Holden, D.F. Weaver and R.L. Houlden, 2005. Morphine, gabapentin, or their combination for neuropathic pain. N. Engl. J. Med., 352: 1324-1334.
    PubMed    


  • Zin, C.S., L.M. Nissen, J.P. O'Callaghan, S.B. Duffull, M.T. Smith and B.J. Moore, 2010. A randomized, controlled trial of oxycodone versus placebo in patients with postherpetic neuralgia and painful diabetic neuropathy treated with pregabalin. J. Pain, 11: 462-471.
    PubMed    


  • Harati, H., G. Gooch, M. Swenson, S.V. Edelman and D. Greene et al., 2000. Maintenance of the long-term effectiveness of tramadol in treatment of the pain of diabetic neuropathy. J. Diabetes Complications, 14: 65-70.


  • Ebell, M., 2007. Tramadol relieves neuropathic pain. Am. Fam. Physician., 75: 1335-1336.
    PubMed    


  • Tzschentke, T.M., J. de Vry, R. Terlinden, H.H. Hennies and C. Lange et al., 2006. Tapentadol hydrochloride: Analgesic mu-opioid receptor agonist noradrenaline reuptake inhibitor. Drugs Future, 31: 1053-1061.
    CrossRef    


  • Reeves, R.R. and R.S. Burke, 2008. Tramadol: basic pharmacology and emerging concepts. Drugs Today (Barc)., 44: 827-836.
    PubMed    


  • Olney, N. and H. Rosen, 2010. AVP-923, a combination of dextromethorphan hydrobromide and quinidine sulfate for the treatment of pseudobulbar affect and neuropathic pain. Drugs, 13: 254-265.
    PubMed    


  • Christoph, T., B. Kogel, W. Strassburger and S.A. Schug, 2007. Tramadol has a better potency ratio relative to morphine in neuropathic than in nociceptive pain models. Drugs R. D., 8: 51-57.
    PubMed    


  • Freeman, R., P. Raskin, D.J. Hewitt, G.J. Vorsanger and D.M. Jordan et al., 2007. Randomized study of tramadol/acetaminophen versus placebo in painful diabetic peripheral neuropathy. Curr. Med. Res. Opin., 23: 147-161.
    PubMed    


  • Kazuhisa, M., H. Minoru, I. Hisanori, N. Atsushi and T. Takako et al., 2009. Morphine, oxycodone, and fentanyl exhibit different analgesic profiles in mouse pain models. J. Pharm. Sci., 111: 60-72.
    CrossRef    


  • Wade, W.E. and W.J. Spruill, 2009. Tapentadol hydrochloride: A centrally acting oral analgesic. Clin. Therapeutics, 31: 2804-2818.
    CrossRef    


  • Huang, E.A. and S.E. Gitelman, 2008. The effect of oral alpha-lipoic acid on oxidative stress in adolescents with type 1 diabetes mellitus. Pediatr Diabetes, 9: 69-73.
    PubMed    


  • Shay, K.P., R.F. Moreau, E.J. Smith, A.R. Smith and T.M. Hagen, 2009. Alpha-lipoic acid as a dietary supplement: Molecular mechanisms and therapeutic potential. Biochimica Biophysica Acta-General Subjects, 1790: 1149-1160.
    CrossRef    PubMed    Direct Link    


  • Parenti. F, 1986. Oral treatment with alpha-lipoic acid improves symptomatic diabetic polyneuropathy: The SYDNEY 2 trial. J. Hosp. Infect, 7: 79-83.
    CrossRef    PubMed    Direct Link    


  • Liu, F., Y. Zhang, M. Yang, B. Liu, Y.D. Shen, W.P. Jia and K. Xiang, 2007. Curative effect of alpha-lipoic acid on peripheral neuropathy in type 2 diabetes: A clinical study. HHZhonghua Yi Xue Za Zhi, 87: 2706-2709.
    PubMed    


  • Alexander, S.A., A. Barinov, P.J. Dyck, R. Hermann and N. Kozlova et al., 2003. The sensory symptoms of diabetic polyneuropathy are improved with α-lipoic acid the sydney trial. Diabetes Care, 26: 770-776.
    PubMed    


  • Tankova, T., S. Cherninkova and D. Koev, 2005. Treatment for diabetic mononeuropathy with alpha-lipoic acid. Int. J. Clin. Pract., 59: 645-650.
    PubMed    


  • Foster, T.S., 2007. Efficacy and safety of alpha-lipoic acid supplementation in the treatment of symptomatic diabetic neuropathy. Diabetes Educ., 33: 111-117.
    CrossRef    


  • Vallianou, N., A. Evangelopoulos and P. Koutalas, 2009. Alpha-lipoic Acid and diabetic neuropathy. Rev. Diabetes Stud., 6: 230-236.
    PubMed    


  • Ziegler, D., M. Reljanovic, H. Mehnert and F.A. Gries, 1999. Alpha-lipoic acid in the treatment of diabetic polyneuropathy in Germany: Current evidence from clinical trials. Exp. Clin. Endocrinol. Diabetes, 107: 421-430.
    PubMed    


  • Lorna, M., R.A. Moore, D. Sheena, J.E. Edwards and H.J. McQuay, 2004. Systematic review of topical capsaicin for the treatment of chronic pain. Br. Med. J., 328: 991-991.


  • Campbell, C., J. Campbell, W. Schmidt, K. Brady and B. Stouch, 2009. Topical clonidine gel reduces pain caused by diabetic neuropathy: Results of a multicenter, placebo-controlled clinical trial. J. Pain, 10: 55-55.
    CrossRef    


  • Wolffa, R.F., M.M. Bala, M. Westwood, A.G. Kessels and J. Kleijnen, 2010. 5% lidocaine medicated plaster in painful Diabetic Peripheral Neuropathy (DPN): A systematic review. Swiss Med. Wkly, 140: 297-306.
    Direct Link    


  • De Felipe, C., J.F. Herrero, J.A. O'Brien, J.A. Palmer and C.A. Doyle et al., 1998. Altered nociception, analgesia and aggression in mice lacking the receptor for substance P. Nature, 392: 394-397.
    PubMed    


  • Watson, C.P.N., K.L. Tyler, D.R. Bickers, L.E. Millikan, S. Smith and E. Coleman, 1993. A randomized vehicle-controlled trial of topical capsaicin in the treatment of post herpetic neuralgia. Clin. Therapeutics, 15: 510-526.
    PubMed    


  • Capsaicin Study Group, 1992. Effect of treatment with capsaicin on daily activities of patients with painful diabetic neuropathy. Diabetes Care, 15: 159-165.
    PubMed    


  • Tandan, R., G.A. Lewis, P.B. Krusinski, G.B. Badger and T.J. Fries, 1992. Topical capsaicin in painful diabetic neuropathy. Controlled study with long-term follow-up. Diabetes Care, 15: 8-14.
    PubMed    


  • Rains, C. and H.M. Bryson, 1995. Topical capsaicin. A review of its pharmacological properties and therapeutic potential in post-herpetic neuralgia, diabetic neuropathy and osteoarthritis. Drugs Aging., 7: 317-328.
    PubMed    


  • McCleane, G., 2000. The analgesic efficacy of topical capsaicin is enhanced by glyceryl trinitrate in painful osteoarthritis: a randomized, double blind, placebo controlled study. Eur. J. Pain, 4: 355-360.
    PubMed    


  • McCleane, G.J. and M. McLaughlin, 1998. The addition of GTN to capsaicin cream reduces the discomfort associated with application of capsaicin alone. A volunteer study. Pain, 78: 149-151.
    PubMed    


  • Walker, R.A. and G.J. McCleane, 2002. The addition of glyceryltrinitrate to capsaicin cream reduces the thermal allodynia associated with the application of capsaicin alone in humans. Neurosci. Lett., 323: 78-80.
    CrossRef    


  • Biesbroek, R., V. Bril, P. Hollander, U. Kabadi and S. Schwartz et al., 1995. A double-blind comparison of topical capsaicin and oral amitriptyline in painful diabetic neuropathy. Adv. Ther., 12: 111-120.
    PubMed    


  • McCleane, G., 2000. Topical application of doxepin hydrochloride, capsaicin and a combination of both produces analgesia in chronic human neuropathic pain: a randomized, double-blind, placebo-controlled study. Br. J. Clin. Pharmacol., 49: 574-579.


  • Ertas, M., A. Sagduyu, N. Arac, B. Uludag and C. Ertekin, 1998. Use of levodopa to relieve pain from painful symmetrical diabetic polyneuropathy. Pain, 75: 257-259.
    PubMed    


  • Anghinah, R., A.S. Oliveira and A.A. Gabbai, 1994. Effect of baclofen on pain in diabetic neuropathy. Muscle Nerve, 17: 958-959.
    PubMed    


  • Hounsom, L., D.F. Horrobin, H. Tritschler, R. Corder and D.R. Tomlinson, 1998. A lipoic acid-gamma linolenic acid conjugate is effective against multiple indices of experimental diabetic neuropathy. Diabetologia, 41: 839-843.
    PubMed    


  • Evans, J.D., 2008. Role of acetyl-L-carnitine in the treatment of diabetic peripheral neuropathy. Ann. Pharmacothep., 42: 1686-1691.
    CrossRef    


  • Sun, Y., M.S. Lai and C.J. Lu, 2005. Effectiveness of vitamin B12 on diabetic neuropathy: Systematic review of clinical controlled trials. Acta Neurol. Taiwan, 14: 48-54.
    PubMed    


  • Oskarsson, P., J.G. Ljunggren and P.E. Lins, 1997. Efficacy and safety of mexiletine in the treatment of painful diabetic neuropathy Mexiletine Study Group. Diabetes Care, 20: 1594-1597.
    CrossRef    


  • Sang, C., S. Booher, I. Gilron, S. Parada ans M. Max, 2002. Dextromethorphan and memantine in painful diabetic neuropathy and postherpetic neuralgia: efficacy and dose-response trials. Anesthesiology, 96: 1053-1061.
    Direct Link    


  • Chen, S.R., G. Samoriski and H.L. Pan, 2009. Antinociceptive effects of chronic administration of uncompetitive NMDA receptor antagonists in a rat model of diabetic neuropathic pain. Neuropharmacology, 57: 121-126.
    CrossRef    PubMed    Direct Link    


  • Obrosova, I.G., 2009. Diabetes and the peripheral nerve. Biochim. Biophys. Acta, 1792: 931-940.
    PubMed    


  • Varrassi, G., F. Marinangeli, A. Piroli, S. Coaccioli and A. Paladini, 2010. Strong analgesics: Working towards an optimal balance between efficacy and side effects. Eur. J. Pain, 14: 340-342.
    CrossRef    


  • Boulton, A.J.M., 2005. Opioids for painful diabetic neuropathy. Curr. Diabetes Rep., 5: 407-408.
    CrossRef    


  • Carolina, M., C. Salvatore, M.N. Michael, M. Vincenzo and P. Frank et al., 2007. Therapeutic manipulation of peroxynitrite attenuates the development of opiate-induced antinociceptive tolerance in mice. J. Clin. Invest., 117: 3530-3539.
    CrossRef    


  • Wen, Z.H., G.J. Wu, L.C. Hsu, W.F. Chen and J.Y. Chen et al., 2008. N-Methyl-D-aspartate receptor antagonist MK-801 attenuates morphine tolerance and associated glial fibrillary acid protein up-regulation: A proteomic approach. Acta Anaesthesiol. Scand., 52: 499-508.
    PubMed    


  • Lin, S.L., R.Y. Tsai, C.H. Shen, F.H. Lin, J.J. Wang, S.T. Hsin and C.S. Wong, 2010. Coadministration of ultra-low dose naloxone attenuates morphine tolerance in rats via attenuation of NMDA receptor neurotransmission and suppression of neuroinflammation in the spinal cords. Pharmacol. Biochem. Behav.,


  • Nishikawa, K., K. Tanobe, H. Hinohara, T. Okamoto, S. Saito and F. Goto, 2004. Molecular mechanism of morphine tolerance and biological approaches to resolve tolerance. Masui, 53: 502-507.
    PubMed    


  • Cui, Y., Y. Chen, J.L. Zhi, R.X. Guo, J.Q. Feng and P.X. Chen, 2006. Activation of p38 mitogen-activated protein kinase in spinal microglia mediates morphine antinociceptive tolerance. Brain Res., 1069: 235-243.
    PubMed    


  • Salvemini, D. and W. Neumann, 2010. Targeting peroxynitrite driven nitroxidative stress with synzymes: A novel therapeutic approach in chronic pain management. Life Sci., 86: 604-614.
    CrossRef    


  • Berger, A.C. and J.L. Whistler, 2010. How to design an opioid drug that causes reduced tolerance and dependence. Ann. Neurol., 67: 559-569.
    PubMed    


  • Elizabeth, K.J., B.R. David and D.L. Jon, 2010. Shared mechanisms for opioid tolerance and a transition to chronic pain. J. Neurosci., 30: 4660-4666.
    CrossRef    


  • Taliyan, R., M. Singh and P.L. Sharma, 2010. Effect of hyperglycemia on antinociceptive effect of cannabinoids-agonit, Win 55, 212-2, in STZ induced neuropathic pain in rats. Med. Chem. Res., 19: 103-103.


  • Kalra, S., K. Bharti and K. Naresh, 2007. Prevention and management of diabetes: the role of the physiotherapist. Diabetes Voice, 52: 12-14.
    Direct Link    


  • Ahn, A.C., T. Bennani, R. Freeman, O. Hamdy and T.J. Kaptchuk, 2007. Two styles of acupuncture for treating painful diabetic neuropathy-a pilot randomised control trial. Acupunct Med., 25: 11-17.
    PubMed    


  • Zhang, C., Y.X. Ma and Y. Yan, 2010. Clinical effects of acupuncture for diabetic peripheral neuropathy. J. Trad. Chin. Med., 30: 13-14.
    CrossRef    


  • Rader, A.J. and T.P. Barry, 2006. A proposed mechanism for pain relief following NIR or MIRE therapy. Diabetic Microvascular Complications Today, pp: 27-28.


  • Wei-Hua, Q., Q. Hong, W. Tong, B. Yan-Hui, L. Lan and Q. Liang-Cai, 2004. Clinical research on acupuncture treatment of diabetic peripheral neuropathy. J. Acupuncture Tuina Sci., 2: 12-14.
    CrossRef    


  • Goodnick, P.J., K. Breakstone, W. Xue-Lan and A. Kumar, 2000. Acupuncture and neuropathy. Am. J. Psychiatry, 157: 1342-1343.
    Direct Link    


  • Singh, U. and I. Jialal, 2008. α-lipoic acid supplementation and diabetes. Nutr. Rev., 66: 646-657.
    PubMed    

  • © Science Alert. All Rights Reserved