Treatment of Painful Peripheral Neuropathies


Thomas H Brannagan III, MD
Peripheral Neuropathy Center
Weill Medical College of Cornell University


INTRODUCTION

Peripheral neuropathy is a common problem and for some patients, pain is a disabling component of the neuropathy. The first goal in evaluating patients with painful peripheral neuropathies is to identify the cause, with the hope of identifying treatment to reverse nerve damage. There are a large number of disorders that may be associated with a painful peripheral neuropathy. (Table 1) When neuropathic pain is present, it is appropriate to treat this concurrently, during the evaluation, as well as during the treatment of the neuropathy, if identified is in process. For some types of neuropathy, there are no available treatments for the underlying disorder, and therapy for neuropathic pain, if present may be the only treatment available.

Pain can be classified broadly as two types. Nociceptive pain is protective and is a normal response to tissue injury, serving to warn of the presence of injury. There is also sensitization of peripheral nociceptors and central nervous system changes, which protect the damaged area by avoiding contact.

Neuropathic pain is a pathologic or maladaptive pain, which results from damage to the nervous system, producing pain in the absence of stimulation of nociceptors or inappropriate response to stimulation of nociceptors. Nociceptive and neuropathic pain are not synonymous with acute and chronic pain. For instance, rheumatoid arthritis is a chronic pain, which is nociceptive pain. A herniated disc can cause acute sciatic pain, which is neuropathic.

Patients with neuropathic pain typically describe burning, lancinating, stabbing, cramping, aching and sometimes vice-like pain. It can be paroxysmal or constant. On examination hyperalgesia, allodynia or hyperpathia are sometimes elicited.

The normal pathways involved in the transmission of pain begin with stimulation of nociceptors including those that respond to chemical irritant stimuli such as VR1, DRASIC, P2X3 and noxious heat stimuli such as VR1 and VRL1. Signals resulting from intense mechanical and thermal stimulate A delta fiber nociceptors and intense mechanical ,thermal and chemical stimuli stimulate polymodal C nociceptors. Afferent fibers synapse in Rexed’s lamina I, II and V in the spinal cord, which is the first level of modulation. Opiate receptors and interneurons are present at the dorsal horn. There are are also descending inputs from the hypothalamus, periaqueductal gray. Opiods, norepineprhine and serotonin have modulatory effects on pain transmission.

MECHANISMS OF NEUROPATHIC PAIN

Why certain neuropathies cause pain is unknown, but there has been increasing knowledge about the mechanism of pain. The gate theory of pain proposed that the substantia gelatinosa acted as a gate allowing pain transmission to proceed and that the inhibitory affect of the substantia gelatinosa is increased by large diameter fibers. Central descending influence was also postulated. Though specifics of this theory have been shown to be incorrect, the basic premise of central modification of pain perception at the dorsal horn and other parts of the central nervous system is still held. Dyck et al, noted that neuropathic pain was related to the rate and kind of nerve fiber degeneration. It is not related simply to the ratio of remaining large and small nerve fibers. Brown et al noted that unmyelinated and small myelinated fibers were most prominently involved and unmyelinated nerve fiber sprouting was evident in painful diabetic neuropathy.

Animal models of pain have added to the understanding of neuropathic pain. There are 4 commonly used animal models, which include the chronic constrictive injury (CCI) model, the partial nerve transection, spinal nerve transection model and the spared nerve model. The chronic constrictive injury model is produced by a loosely constrictive ligature around the sciatic nerve. Almost all of A-β fibers die, as do the majority of Aδ fibers. A large percentage of C fibers persist.

The partial nerve transection model involves tightly ligating and transecting 1/3 to ½ of of the rat sciatic nerve. The spinal nerve transection model involves tight ligation and transection of the L5 and L6 nerve roots. This affects 50% of the sural and saphenous nerve fibers. The spared nerve model involves a lesion of 2 of the 3 terminal branches of the sciatic nerve (tibial and peroneal) intact, leaving the sural nerve intact. There is partial deafferentation but not differential involvement of nerve fibers, in these three models.

Heat hyperalgesia, mechano-hyperalgesia, mechano-allodynia and cold-allodynia may be observed and behavioral changes consistent with spontaneous pain such as limping and guarding the hind paw are seen. Both peripheral and central mechanisms have been observed in animal models.

After nerve injury there are spontaneous discharges of nerve fibers, neuromas and dorsal root ganglion. This has been seen in traumatic nerve injury models , as well as models of peripheral neuropathy from diabetes and heavy metal intoxication. Increased expression of sodium channels are seen in neuroma’s in humans and animal models. The density of sodium channel expression correlates with the degree of pain. Changes in sodium channel expression are seen and may contribute to nerve excitability and spontaneous pain..

These spontaneous discharges by themselves can cause pain, but they also have additional affects. C fiber afferents trigger cell death of neurons in the dorsal horn, where inhibitory interneurons are concentrated, possibly through an excitotoxic mechanism. This may result in increased pain transmission.. C fiber afferents release glutamate and synapse on 2nd order neurons and have excitatory affects. Glutamate synapses at AMPA receptors which depolarizes the membrane. This depolarization releases the inhibition of the NMDA receptor by the magnesium ions and there is an influx of calcium. Second order neurons are gradually depolarized and responses are amplified and this changes the response of neurons to subsequent input.

Two processes that are distinct occur at the dorsal horn which are designated “windup” and “central sensitization.” Windup up results from repetitive C fiber firing at low frequencies that result in a progressive buildup of the amplitude of the response of the dorsal horn neuron, only during the repetitive train. Central sensitization is an abnormal sensitivity with a spread of hypersensitivity to uninjured sites and pain resulting from stimulation of low threshold Ab mechanoreceptors. Central sensitization follows a brief high frequency input and the increased response to subsequent inputs is prolonged. Both can be blocked by NMDA receptor antagonists. Central sensitization can result from windup. This is a result of the calcium influx through the NMDA receptor following depolarization of the dorsal horn membrane. The intracellular calcium activated a number of kinases of which protein kinase C (PKC) is likely important. PKC enhances the NMDA receptor , which results in subsequent glutamate binding of the NMDA receptor generating an inward current. Though windup can result in central sensitization it is not necessary for central sensitization to occur.

Similar observations had previously been noted by Denny-Brown who described an enlarged and hypersensitive dermatomal region in primates when severing the surrounding nerve roots distal to the dorsal root ganglion compared with severing proximal to the DRG. This suggested plasticity of the dorsal horn neurons secondary to input from the DRG.

Nerve injury also results in sprouting of myelinated fibers into lamina II of the dorsal horn, which under normal circumstances receives only C fiber input. This may result in allodynia.

There is a genetic influence on the experience of neuropathic pain though it is poorly delineated. There are variations on the expression of pain behaviors seen in different strains of animals. A recent study noted that 56% of patients with painful diabetic neuropathy had a relative with painful diabetic neuropathy suggesting a genetic component.

The relative importance of these various mechanisms is not clearly known, however there is potentially multiple sites for intervention in treating painful neuropathies.

Medications

Tricyclic anti-depressants have been beneficial in controlled studies of neuropathic pain. They block the reuptake of norepinephrine and serotonin and are thought to modulate descending inhibitory pathways. There benefit with neuropathic pain is independent of their effect on depression. Patients who are not depressed can respond and lower doses than are used to treat depression are effective to treat neuropathic pain. Tricyclic anti-depressants can block voltage dependent sodium channels and this may contribute to their efficacy to treat neuropathic pain. They are typically started at a dose of 10-25 mg at night and titrated as tolerated up to a dose of 150 mg if necessary. Side effects include dry mouth, cardiac arrhythmias, urinary retention and sexual dysfunction. Venlafaxine has fewer side effects and has also been reported to benefit neuropathy pain.
The selective serotinin reuptake inhibitors (SSRI) have been less effective for treatment of neuropathic pain. Fluoxetine was not effective in clinical trials. Paroxetine and Citalopram been reported to be effective.

Anticonvulsants have also been studied in neuropathic pain. Both phenytoin (Dilantin) and carbamezapine (Tegretol) have been beneficial in trials of diabetic neuropathy. They act as sodium channel blockers. Both medications have frequent side effects that are well known.

Gapapentin (Neurontin), binding site alpha delta Ca channel. It is though to act at a spinal site of action. Controlled studies in diabetic neuropathy, post-herpetic neuralgia and other neuropathic pain states. Starting doses are usually100mg tid or 300mg qhs. Unless the drug is not tolerated or a dose of 600 mg tid has been tried it should not be considered a treatment failure. Often higher doses provide more benefit. Side effects include drowsiness, fogginess, leg edema. Gabapentin is cleared by the kidneys and there are not significant drug interactions. The dosing should be adjusted in renal insufficiency and failure.

Lamotrigine (Lamictal) is a sodium channel blocker, which also inhibits glutamate release. It has reduced cold allodynia in the CCI model. Lamotrigine is effective in controlled studies in diabetic, HIV associated painful neuropathy, post-herpetic neuralgia, and trigeminal neuralgia.. I’ve found lamotrigine to cause less drowsiness or dizziness than most other medications used for neuropathic pain and though these symptoms have been seen during epilepsy trials, they were not seen more commonly than placebo in 2 recent trials of neuropathy pain. Rash is a side effect and a Stevens Johnson syndrome may occur. This is less frequent when the medication is titrated slowly. The rash is less common with a slow titration. The following titration schedule has been used for neuropathic pain: 25mg qd for 2 weeks, 25 mg bid for the next 2 weeks, 50 mg bid for the next 2 weeks, 100mg bid for 2 weeks. If the patient is also taking valproate which inhibits the metabolism of lamotrigine, a dose of 25 mg qod is recommended as an initial dose.

Oxcarbazepine (Trileptal) is similar in structure to carbamezapine, but lacks the 10,11 epoxide, which is thought to be responsible for better tolerability. Autoinduction does not occur and rash and drug interactions are less frequent than with carbamezapine. Hyponatremia does occur.

Double blind placebo controlled studies of mexilitine have been negative, however subgroups with stabbing and burning pain had benefit. Another study noted benefit in night time pain with the 675 mg dose, but not the lower doses of 450 mg or 225 mg.

Topiramate (Topamax) showed benefit in animal models and preliminary studies and anecdotal reports of diabetic neuropathy and neuropathic pain. Though the results have not been published Johnson & Johnson announced that the results of clinical trials in diabetic neuropathy pain were not positive (Reuter 9/18/2001).

Dextromorthorphan is a low affinity NMDA antigonist. It has been beneficial in animal studies and painful neuropathies. Side effects are frequent and it is poorly tolerated. Dextromethorphan in combination with other medications as well as more selective NMDA receptor antagonists that may have less side effects are being pursued.

Clonidine an alpha-2 agonist, as a transdermal patch was successfully used in a subset of patients with painful diabetic neuropathy using an enrolled enrichment design. Tizanidine, also an alpha-2 agonist, reduces thermal hyperalgesia in the CCI rat model and has been successful in open label studies in neuropathic pain.

Tramodol (Ultram) has been effective in studies of painful diabetic neuropathy. Side effects include nausea, headache, constipation, somnolence and seizures. Lidoderm is beneficial for post-herpetic neuralgia. The dose of 1-3 patches for 12 hours, with 12 hours off the patch. It has been beneficial in other forms of neuropathic pain.

Based on small preliminary and anecdotal series, as well as animal data, other medications including zonisamide and levetiracetam may be beneficial.

HIV painful neuropathy is a particularly difficult syndrome to treat. Controlled studies of amitriptyline, mexilitine, and acupuncture have been negative. Gabapentin has been reported to be successful, though requiring higher doses. Phenytoin and carbamezapine as P450 inducers should be avoided as they can induce the metabolism of the protease inhibitors and make them ineffective. Lamotrigine has been successful in blinded placebo controlled studies. Two consecutive studies however provided conflicting data with the first suggesting that there was no benefit above placebo in patients taking neurotoxic dideoxynucleotide anti-retrovirals compared with those patient not taking neurotoxic antiretrovirals and the second study showed the opposite. Since several different mechanisms are involved in neuropathic pain, treatment directed against the mechanism of pain would be desirable and has been proposed. Unequivocally identifying mechanisms at work to date has been difficult in patients. For instance the symptom of allodynia may occur from peripheral sensitization or central sensitization. It is a commonly held belief that tricyclic anti-depressants are more effective against burning pain and anti-convulsants are more effective against paroxysmal stabbing pain. In clinical trials however this has not been demonstrated.. Selective affects in animal models have been demonstrated. For example dextrorphan reduces heat hyperalgesia, but has no effect on mechanical allodynia in the CCI model. How this correlates to treatment of people with neuropathic pain is still evolving. In the future cocktails targeting different mechanisms at work in patients with peripheral neuropathies may be possible.

Reference List

Ansari A. The efficacy of newer antidepressants in the treatment of chronic pain: A review of current literature. Harvard Review of Psychiatry 2000; 7: 257-277.
Backonja M, Beydoun A, Edwards KR et al. Gabapentin for the symptomatic treatment of painful neuropathy in patients with diabetes mellitus - A randomized controlled trial. Jama-Journal of the American Medical Association 1998; 280: 1831-1836.
Backonja MM. Use of anticonvulsants for treatment of neuropathic pain. Neurology 2002; 59: S14-S17.
Bennett GJ, Xie YK. A Peripheral Mononeuropathy in Rat That Produces Disorders of Pain Sensation Like Those Seen in Man. Pain 1988; 33: 87-107.
Boyce S, Wyatt A, Webb JK et al. Selective NMDA NR2B antagonists induce antinociception without motor dysfunction: correlation with restricted localisation of NR2B subunit in dorsal horn. Neuropharmacology 1999; 38: 611-623.
Brau ME, Dreimann M, Olschewski A, Vogel W, Hempelmann G. Effect of drugs used for neuropathic pain management on tetrodotoxin-resistant Na+ currents in rat sensory neurons. Anesthesiology 2001; 94: 137-144.
Brown MJ, Martin JR, Asbury AK. Painful Diabetic Neuropathy - Morphometric Study. Archives of Neurology 1976; 33: 164-171.
Chadda VS, Mathur MS. Double blind study of the effects of diphenylhydantoin sodium on diabetic neuropathy. J.Assoc.Physiol.India 26, 403. 1978.
Craner MJ, Klein JP, Renganathan M, Black, J. A., and Waxman, S. G. Changes of sodium channel expression in experimental painful diabetic neuropathy. Annals of Neurology 52, 786-792. 2002.
Decosterd I, Woolf CJ. Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain 2000; 87: 149-158.
Denny-Brown D, Kirk EJ, Yanagisawa N. Tract of Lissauer in Relation to Sensory Transmission in Dorsal Horn of Spinal-Cord in Macaque Monkey. Journal of Comparative Neurology 1973; 151: 175-199.
Devers A, Galer BS. Topical lidocaine patch relieves a variety of neuropathic pain conditions: An open-label study. Clinical Journal of Pain 2000; 16: 205-208.
Devor M, Raber P. Heritability of Symptoms in An Experimental-Model of Neuropathic Pain. Pain 1990; 42: 51-67.
Dyck PJ, Lambert EH, Obrien PC. Pain in Peripheral Neuropathy Related to Rate and Kind of Fiber Degeneration. Neurology 1976; 26: 466-471.
Edwards KR, Glantz MJ, Button J, Norton JA, Whittaker T, Cross N. Efficacy and safety of topiramate in the treatment of painful diabetic neuropathy: A double-blind, placebo controlled study. Neurology 2000; 54: A81.
Eisenberg E, Lurie Y, Braker C, Daoud D, Ishay A. Lamotrigine reduces painful diabetic neuropathy - A randomized, controlled study. Neurology 2001; 57: 505-509.
England JD, Happel LT, Kline DG et al. Sodium channel accumulation in humans with painful neuromas. Neurology 1996; 47: 272-276.
Galer BS, Gianas A, Jensen MP. Painful diabetic polyneuropathy: epidemiology, pain description, and quality of life. Diabetes Research and Clinical Practice 2000; 47: 123-128.
Harati Y, Gooch C, Swenson M et al. Double-blind randomized trial of tramadol for the treatment of the pain of diabetic neuropathy. Neurology 1998; 50: 1842-1846.
Hord AH, Chalfoun AG, Denson DD, Azevedo MI. Systemic tizanidine hydrochloride (Zanaflex (TM)) relieves thermal hyperalgesia in rats with an experimental mononeuropathy. Anesthesia and Analgesia 2001; 93: 1310-1315.
Hunter JC, Gogas KR, Hedley LR et al. The effect of novel anti-epileptic drugs in rat experimental models of acute and chronic pain. European Journal of Pharmacology 1997; 324: 153-160.
Kieburtz K, Simpson D, Yiannoutsos C et al. A randomized trial of amitriptyline and mexiletine for painful neuropathy in HIV infection. Neurology 1998; 51: 1682-1688.
Kim SH, Chung JM. An Experimental-Model for Peripheral Neuropathy Produced by Segmental Spinal Nerve Ligation in the Rat. Pain 1992; 50: 355-363.
Kirk EJ, Denny-Brown D. Functional Variation in Dermatomes in Macaque Monkey Following Dorsal Root Lesions. Journal of Comparative Neurology 1970; 139: 307-&.
Laird JMA, Bennett GJ. Dorsal-Root Potentials and Afferent Input to the Spinal-Cord in Rats with An Experimental Peripheral Neuropathy. Brain Research 1992; 584: 181-190.
Mannion RJ, Doubell TP, Coggeshall RE, Woolf CJ. Collateral sprouting of uninjured primary afferent A-fibers into the superficial dorsal horn of the adult rat spinal cord after topical capsaicin treatment to the sciatic nerve. Journal of Neuroscience 1996; 16: 5189-5195.
Max MB. Thirteen consecutive well-designed randomized trials show that antidepressants reduce pain in diabetic neuropathy and postherpetic neuralgia. Pain Forum 1995; 4: 248-253.
Max MB, Culnane M, Schafer SC et al. Amitriptyline Relieves Diabetic Neuropathy Pain in Patients with Normal Or Depressed Mood. Neurology 1987; 37: 589-596.
Max MB, Lynch SA, Muir J, Shoaf SE, Smoller B, Dubner R. Effects of Desipramine, Amitriptyline, and Fluoxetine on Pain in Diabetic Neuropathy. New England Journal of Medicine 1992; 326: 1250-1256.
Melzack R, Wall PD. Pain Mechanisms - A New Theory. Science 1965; 150: 971-&.
Nelson KA, Park KM, Robinovitz E, Tsigos C, Max MB. High-dose oral dextromethorphan versus placebo in painful diabetic neuropathy and postherpetic neuralgia. Neurology 1997; 48: 1212-1218.
Newshan G. HIV neuropathy treated with gabapentin. Aids 1998; 12: 219-221.
Potter D, Edwards KR. Potential role of topiramate in relief of neuropathic pain. Neurology 1998; 50: A255.
Romanelli F, Jennings HR, Nath A, Ryan M, Berger J. Therapeutic dilemma - The use of anticonvulsants in HIV-positive individuals. Neurology 2000; 54: 1404-1407.
Rull JA, Quibrera R, GONZALEZ.H, CASTANED.OL. Symptomatic Treatment of Peripheral Diabetic Neuropathy with Carbamazepine (Tegretol) - Double Blind Crossover Trial. Diabetologia 1969; 5: 215-&.
Seltzer Z, Dubner R, Shir Y. A Novel Behavioral-Model of Neuropathic Pain Disorders Produced in Rats by Partial Sciatic-Nerve Injury. Pain 1990; 43: 205-218.
Semenchuk MR, Sherman S. Effectiveness of tizanidine in neuropathic pain: An open-label study. Journal of Pain 2000a; 1: 285-292.
Semenchuk MR, Sherman S. Effectiveness of tizanidine in neuropathic pain: An open-label study. Journal of Pain 2000b; 1: 285-292.
Shlay JC, Chaloner K, Max MB et al. Acupuncture and amitriptyline for pain due to HIV-related peripheral neuropathy - A randomized controlled trial. Jama-Journal of the American Medical Association 1998; 280: 1590-1595.
Simpson DM, McArthur JC, Olney R, Ross D, Barrett P, Baird BJ. A multicenter, double-blind, randomized, placebo-controlled evaluation of lamotrigine in adult subjects with painful HIV-associated peripheral neuropathy. Neurology 2002; 58: A407.
Simpson DM, Olney R, McArthur JC, Kahn A, Godbold J, Ebel-Frommer KZ. A placebo-controlled trial of lamotrigine for painful HIV-associated neuropathy. Neurology 2000; 54: 2115-2119.
Sindrup SH, Bjerre U, Dejgaard A, Brosen K, Aaesjorgensen T, Gram LF. The Selective Serotonin Reuptake Inhibitor Citalopram Relieves the Symptoms of Diabetic Neuropathy. Clinical Pharmacology & Therapeutics 1992; 52: 547-552.
Sindrup SH, Gram LF, Brosen K, Eshoj O, Mogensen EF. The Selective Serotonin Reuptake Inhibitor Paroxetine Is Effective in the Treatment of Diabetic Neuropathy Symptoms. Pain 1990; 42: 135-144.
Sugimoto T, Bennett GJ, Kajander KC. Strychnine-Enhanced Transsynaptic Degeneration of Dorsal Horn Neurons in Rats with An Experimental Painful Peripheral Neuropathy. Neuroscience Letters 1989; 98: 139-143.
Sugimoto T, Bennett GJ, Kajander KC. Transsynaptic Degeneration in the Superficial Dorsal Horn After Sciatic-Nerve Injury - Effects of A Chronic Constriction Injury, Transection, and Strychnine. Pain 1990; 42: 205-213.
Tal M, Bennett GJ. Neuropathic Pain Sensations Are Differentially Sensitive to Dextrorphan. Neuroreport 1994; 5: 1438-1440.
Waxman SG, Cummins TR, Dib-Hajj S, Fjell J, Black JA. Sodium channels, excitability of primary sensory neurons, and the molecular basis of pain. Muscle & Nerve 1999; 22: 1177-1187.
Woolf CJ. Windup and central sensitization are not equivalent. Pain 1996; 66: 105-108.
Woolf CJ, Bennett GJ, Doherty M et al. Towards a mechanism-based classification of pain? Pain 1998; 77: 227-229.
Woolf CJ, Mannion RJ. Neuropathic pain: aetiology, symptoms, mechanisms, and management. Lancet 1999; 353: 1959-1964.
Woolf CJ, Salter MW. Neuroscience - Neuronal plasticity: Increasing the gain in pain. Science 2000; 288: 1765-1768.
Xiao, Y. K. and Bennett, G. J. Gabapentin has an anti-nociceptive effect mediated via a spinal site of action in a rat model of painful peripheral neuropathy. Analgesia 2, 267-273. 1996.
Zakrzewska JM, Patsalos PN. Oxcarbazepine-a new drug in the management of intractable trigeminal neuralgia. J Neurol Neurosurg Psychiatry 1989; 52: 472-476.
 
Table 1 - Painful peripheral neuropathies
Diabetes mellitus
HIV
Amyloidosis
Fabry’s disease
Celiac disease
Hereditary sensory neuropathy
Ideopathic
Toxic neuropathies
Dideoxynucleotide antiretrovirals
Antineoplastic agents (vincristine, taxol)
Isoniazid
Alcohol
Pyridoxine (B6)
Autoimmune
Anti-sulfatide antibody neuropathies
Neuropathy associated with monoclonal gammopathy
Sjogren’s disease
Lupus
Vasculitic neuropathy

Table 2 - Potential mechanisms of neuropathic pain
Sodium channel accumulation, redistribution, altered expression
Central sensitization
Peripheral sensitization
Α-receptor expression
Sympathetic sprouting
Increased transmission
Reduced inhibition

For original paper see:
Treatment of Painful Peripheral Neuropathies