6
AIDS and AIDS-treatment Neuropathies Derek Williams, R-PA, Anthony Geraci, MD, and David M. Simpson, MD Address Mount Kisco Medical Group, 90 South Bedford Road, Mount Kisco, NY 10549, USA. E-mail: [email protected] Current Pain and Headache Reports 2002, 6:125–130 Current Science Inc. ISSN 1531-3433 Copyright © 2002 by Current Science Inc. Introduction The face of AIDS is changing. The life expectancy of indi- viduals infected with HIV has increased dramatically due to a better understanding of HIV infection, which in turn has led to advances in medications used to treat HIV and associated opportunistic infections. Along with increased survival, there has been a decrease in the incidence of some neurologic complications of HIV infection. In the era of highly active antiretroviral therapy (HAART), there has been a decline in the incidence of neurologic diseases, such as AIDS dementia, toxoplasmosis, and cytomegalovirus (CMV) infections [1,2]. Evidence that HAART improves both neurocognitive and sensory nerve function in HIV-infected patients is encouraging [3••,4], and provides a rationale for future studies on the nervous system effects of aggressive antiretroviral therapy. The neuromuscular complications of HIV are common (Table 1) [5]. The most common peripheral nerve disorder is distal symmetrical polyneuropathy (DSP), which occurs in more than 30% of patients with AIDS [6,7•]. Asymptomatic DSP (signs of peripheral nerve dysfunction without subjective pain) may occur in more than 50% of patients with less advanced disease [8], and can be demon- strated by abnormal nerve conduction studies (NCS) [7•]. Current anti-HIV therapies incorporating nucleoside analogue derivatives have well-documented dose-depen- dent toxicities, including DSP [9]. The relative benefit versus toxicity of these medications when used for aggressive HIV viral suppression in those predisposed to developing symptomatic DSP is a subject of investigation. Clinical Presentation and Diagnosis The cardinal feature of DSP is pain. Classically, patients present with symmetrical paresthesias, dysthesias, and distal numbness in the lower extremities. In advanced DSP, there may be similar symptoms in the upper extremities, as well as weakness of distal intrinsic muscle groups [10]. Symptoms usually have an insidious onset, except when DSP is related to antiretroviral drugs, in which case the symptoms may appear more rapidly [11]. The pain of DSP is often described as burning, and patients may complain of hypersensitivity in their feet. Often, the degree of pain reported by the patient is disproportionate to objective findings on neurologic examination. Patients may have an antalgic gait and be unable to tolerate even light contact with clothing or bedsheets. Wearing socks or shoes increases their discomfort. Neurologic examination demonstrates decreased or absent ankle reflexes, elevated vibratory threshold, and reduction in pain sensation or hyperalgesia in a stock- ing and glove distribution. There may also be a decrease in pain and temperature sensation, but joint proprioception is typically relatively normal [10]. Depressed or absent ankle reflexes are the most common sign of DSP [6,7•], and may be the only sign of neuropathy. Relatively normal ankle reflexes, together with hyperactive knee reflexes, might indicate coexisting myelopathy, particularly when extensor plantar responses are present. The diagnosis of DSP is usually straightforward and can be made based on signs and symptoms in conjunction with a comprehensive history and neurologic examination. Marra et al. [8] demonstrated that trained nonphysician clinicians, such as physician assistants and nurse practit- ioners, can diagnose DSP with a sensitivity that is com- parable to a neurologist’s examination. Confirmation of DSP can be made by nerve conduction studies (NCS). The primary pathologic feature of DSP is axonal degen- eration, as evidenced by a disproportionate loss of large myelinated fibers of both sensory and motor nerves. [7•,12,13]. There is a percentage of patients with DSP, however, who have normal NCS, as well as those without clinical signs and symptoms of DSP with abnormal NCS. AIDS and AIDS-treatment neuropathies are common in individuals infected with HIV. As patients live longer due to improved antiretroviral therapies, the impact of painful neuropathy on patients’ lives may increase. Several anti- retroviral medications are known to cause toxic neuro- pathy in patients with AIDS, but this may be outweighed by the beneficial effects of viral suppression. Current theories on the pathogenesis of AIDS neuropathies include mito- chondrial toxicity secondary to γ-DNA polymerase inhibition and subsequent abnormal mitochondrial DNA synthesis. Treatment of AIDS neuropathies is directed toward relief of symptoms; however, new evidence suggests that aggressive antiretroviral therapy may also be effective.

AIDS and AIDS-treatment neuropathies

Embed Size (px)

Citation preview

Page 1: AIDS and AIDS-treatment neuropathies

AIDS and AIDS-treatment NeuropathiesDerek Williams, R-PA, Anthony Geraci, MD, and David M. Simpson, MD

AddressMount Kisco Medical Group, 90 South Bedford Road, Mount Kisco, NY 10549, USA. E-mail: [email protected]

Current Pain and Headache Reports 2002, 6:125–130Current Science Inc. ISSN 1531-3433Copyright © 2002 by Current Science Inc.

IntroductionThe face of AIDS is changing. The life expectancy of indi-viduals infected with HIV has increased dramatically dueto a better understanding of HIV infection, which in turnhas led to advances in medications used to treat HIV andassociated opportunistic infections. Along with increasedsurvival, there has been a decrease in the incidence of someneurologic complications of HIV infection. In the eraof highly active antiretroviral therapy (HAART), there hasbeen a decline in the incidence of neurologic diseases, suchas AIDS dementia, toxoplasmosis, and cytomegalovirus(CMV) infections [1,2]. Evidence that HAART improvesboth neurocognitive and sensory nerve function inHIV-infected patients is encouraging [3••,4], and providesa rationale for future studies on the nervous system effectsof aggressive antiretroviral therapy.

The neuromuscular complications of HIV are common(Table 1) [5]. The most common peripheral nerve disorderis distal symmetrical polyneuropathy (DSP), whichoccurs in more than 30% of patients with AIDS [6,7•].Asymptomatic DSP (signs of peripheral nerve dysfunctionwithout subjective pain) may occur in more than 50% ofpatients with less advanced disease [8], and can be demon-strated by abnormal nerve conduction studies (NCS) [7•].Current anti-HIV therapies incorporating nucleoside

analogue derivatives have well-documented dose-depen-dent toxicities, including DSP [9]. The relative benefit versustoxicity of these medications when used for aggressive HIVviral suppression in those predisposed to developingsymptomatic DSP is a subject of investigation.

Clinical Presentation and DiagnosisThe cardinal feature of DSP is pain. Classically, patientspresent with symmetrical paresthesias, dysthesias, anddistal numbness in the lower extremities. In advanced DSP,there may be similar symptoms in the upper extremities,as well as weakness of distal intrinsic muscle groups [10].Symptoms usually have an insidious onset, exceptwhen DSP is related to antiretroviral drugs, in which casethe symptoms may appear more rapidly [11]. The pain ofDSP is often described as burning, and patients maycomplain of hypersensitivity in their feet. Often, the degreeof pain reported by the patient is disproportionate toobjective findings on neurologic examination. Patientsmay have an antalgic gait and be unable to tolerate evenlight contact with clothing or bedsheets. Wearing socks orshoes increases their discomfort.

Neurologic examination demonstrates decreased orabsent ankle reflexes, elevated vibratory threshold,and reduction in pain sensation or hyperalgesia in a stock-ing and glove distribution. There may also be a decrease inpain and temperature sensation, but joint proprioceptionis typically relatively normal [10]. Depressed or absentankle reflexes are the most common sign of DSP [6,7•],and may be the only sign of neuropathy. Relatively normalankle reflexes, together with hyperactive knee reflexes,might indicate coexisting myelopathy, particularly whenextensor plantar responses are present.

The diagnosis of DSP is usually straightforward and canbe made based on signs and symptoms in conjunctionwith a comprehensive history and neurologic examination.Marra et al. [8] demonstrated that trained nonphysicianclinicians, such as physician assistants and nurse practit-ioners, can diagnose DSP with a sensitivity that is com-parable to a neurologist’s examination. Confirmation ofDSP can be made by nerve conduction studies (NCS).

The primary pathologic feature of DSP is axonal degen-eration, as evidenced by a disproportionate loss oflarge myelinated fibers of both sensory and motor nerves.[7•,12,13]. There is a percentage of patients with DSP,however, who have normal NCS, as well as those withoutclinical signs and symptoms of DSP with abnormal NCS.

AIDS and AIDS-treatment neuropathies are common in individuals infected with HIV. As patients live longer due to improved antiretroviral therapies, the impact of painful neuropathy on patients’ lives may increase. Several anti-retroviral medications are known to cause toxic neuro-pathy in patients with AIDS, but this may be outweighed by the beneficial effects of viral suppression. Current theories on the pathogenesis of AIDS neuropathies include mito-chondrial toxicity secondary to γ-DNA polymerase inhibition and subsequent abnormal mitochondrial DNA synthesis. Treatment of AIDS neuropathies is directed toward relief of symptoms; however, new evidence suggests that aggressive antiretroviral therapy may also be effective.

Page 2: AIDS and AIDS-treatment neuropathies

126 Neuropathic Pain

Tagliati et al. [7•] documented that 12 of 63 patients(19%) with clinical DSP had normal results of NCS, and26 of 103 patients (25%) without clinical DSP hadabnormal results. A negative examination in the presenceof clinical disease could be a result of an abnormality ofsmall nerve fibers [3••,7•], which are not examined byconventional NCS. The most common NCS abnormalityseen in patients with DSP is reduced or absent sural nerveaction potentials [7•]. In spite of the predominance of sen-sory complaints, motor fibers are usually affected as well.Generally, however, motor nerve conduction velocities areonly mildly reduced in relation to the reduction in ampli-tude [14]. NCS of the upper extremities may reveal reducedsensory and motor amplitudes of the median or ulnarnerves. F-wave and H-reflex latencies may be prolonged[7•]. Diagnostic modalities, such as quantitative sensorytesting and punch skin biopsy, are generally reserved forthe research setting [15,16]. Invasive studies, such as suralnerve biopsy, are rarely necessary in diagnosing DSP, butmay be helpful in complicated cases with atypical features.The electrophysiologic abnormalities in drug-induced andHIV-related DSP are indistinguishable [17].

In an ongoing attempt to develop more sensitive screen-ing examinations for DSP, some investigators have employedother tests or combinations of standard tests. Bouhassiraet al. [18] compared quantitative sensory testing (QST) withestablished electrodiagnostic studies, such as NCS. Theyfound that in NCS-confirmed painful and nonpainful DSP,QST revealed increases in thermal and mechanical thresh-olds, as well as pain thresholds. They concluded that QST canbe helpful in detecting sensory alterations in patients withpainful neuropathies, which might prove useful in quantify-ing DSP clinically, and for following the effects of treatmentson neuropathic pain [18].

PathophysiologySeveral observations support multifactorial etiologies forthe development and severity of DSP. Tagliati et al. [7•]

reported that older age and advanced HIV infection, asreflected by reduced CD4 lymphocyte counts, are assoc-iated with DSP. Low hemoglobin levels also correlatedwith DSP in that study, likely as a surrogate for advanceddisease and a relatively debilitated state. Plasma HIV RNAis an important predictor of neurologic outcomes. Individ-uals with plasma HIV RNA levels greater than 10,000copies/mL have a 2.3-fold greater hazard of sensory neur-opathy than those with less than 500 copies/mL [19••].Accordingly, aggressive antiretroviral therapy and suppres-sion of plasma HIV-1 viral burden improves sensory func-tion in HIV-infected patients, as measured by quantitativesensory testing [3••]. There are no data corroborating thisobservation or to indicate whether effective antiretroviraltherapy leads to clinical improvement in DSP. Althoughthe specific pathogenesis of this neuropathy has not yetbeen delineated, it has been linked to the use of nucleosideanalogue reverse transcriptase inhibitors (NRTIs)[9,17,20•]. Commonly referred to as d-drugs, didanosine(ddI), zalcitabine (ddC), and stavudine (d4T) are theNRTIs most implicated in DSP. Combinations of thesemedications with each other or with hydroxyurea [9,20•]and with other medications used to treat HIV-relatedconditions, such as isoniazid (INH) [21], vincristine [22],and thalidomide [23], are also known to cause DSP. Puta-tive contributing factors to the development of neuropathyinclude acetyl-cartinine and vitamin B12 deficiencies[5,24], and neurotoxicity caused by cytokines [25].

Antiretroviral Agents and Mitochondrial ToxicityDistal symmetrical polyneuropathy related to nucleosideanalogue toxicity is dose-dependent [26]. The dideoxy-nucleoside analogues ddI, ddC, and d4T produce well-recognized peripheral neurotoxicity (Table 2) [9]. Periph-eral neuropathy was first described in patients receivingddC [27] and is clinically and electrophysiologicallysimilar to HIV-related peripheral neuropathy. It has beentheorized that the neurotoxin is actually unmasking anexisting, silent peripheral neuropathy. Clinical and electro-physiologic abnormalities are similar to those in HIV-related neuropathy and indicate distal axonopathy[7•,17,27]. Stavudine may also cause peripheral neuro-toxicity. In a study of 36 patients taking a maximum d4Tdosage of 2 mg/kg/day, 20 (55%) developed dose-limitingperipheral neuropathy with identical features to ddC- andddI-induced neuropathy [28].

The occurrence of dideoxynucleoside toxic neuropathyis related to both daily and cumulative dosage [17], andappears to be most frequent with ddC. In a clinicaltrial comparing four different treatment regimens (ZDValone, ddI alone, ZDV plus ddI, ZDV plus ddC), the 6%incidence of HIV-related neuropathy was highest in theZDV plus ddC arm [29]. Risk factors for dideoxynucleosidetoxic neuropathy include a history of neuropathy, older

Table 1. Neuromuscular complications of HIV infection

Neuropathy Common Distal symmetrical polyneuropathy HIV-related Nucleoside analogue-related Less common Inflammatory polyneuropathy Chronic inflammatory demyelinating polyneuropathy Neuropathy due to opportunistic infections Progressive polyradiculopathy (cytomegalovirus) Mononeuritis multiplex (cytomegalovirus, hepatitis C)Myopathy HIV ? Nucleoside analogue-related

Page 3: AIDS and AIDS-treatment neuropathies

AIDS and AIDS-treatment Neuropathies • Williams et al. 127

age, poor nutrition, and more advanced HIV disease[7•,30]. Based on data from the Moore Clinic at JohnsHopkins University, the risk of dideoxynucleoside toxicneuropathy is additive or even synergistic for regimenscontaining ddI plus d4T combination when comparedwith ddI or d4T alone [20•]. We anticipate that as combi-nation dideoxynucleoside therapy is employed morewidely and for longer periods, the incidence of drug-induced neuropathy will probably rise in patients withadvanced HIV disease [20•].

Nucleoside analogue-related neuropathy may be due,in part, to their effects on mitochondrial DNA synthesis[27,31,32]. There are several mechanisms by which NRTIsare believed to interfere with mitochondrial DNA synthe-sis. These may be linked by the fact that they provide analternative substrate for mitochondrial g-DNA polymerasebecause they are derivatives of the endogenous nucleo-sides: adenosine, guanosine, thymidine, and cytidine [31].The five major human cellular DNA polymerases are alpha(a), beta (b), delta (d), epsilon (e), and gamma (g). Onlyg-DNA polymerase is located in mitochondria, whereasthe others reside in the nucleus [31]. Inhibition of g-DNApolymerase activity would be expected to result in mito-chondrial (mt)DNA depletion. However, although inhibi-tion of g-DNA polymerase occurs with nucleosideanalogue use, tissue-specific effects are not well understood[33], and there are no data that establish an association ofmitochondrial dysfunction with peripheral neuropathy inpatients taking these medications. Brinkman et al. [34•]have hypothesized that mitochondrial toxicity due to

nucleoside analogue reverse transcriptase inhibitors is, inpart, responsible for antiretroviral-related lipodystrophy.This syndrome bears resemblance to multiple symmetricallipomatosis, which includes peripheral neuropathy as aprominent feature. Several investigators have demon-strated point mutations and multiple or single mtDNAdeletions in this disorder and other inherited mito-chondrial diseases, leading to impaired cytochrome Coxidase function [35]. This in turn leads to impaired oxida-tive phosphorylation, anaerobic glycolysis, and formationof lactate. Other theories on the causes of mitochondrialtoxicity include the development of oxygen-free radicalsthat damage deoxyguanosine [36], involvement of theother DNA nucleosides [32], and mitochondrial-inducedapoptosis [31,37].

HydroxyureaHydroxyurea is an antimetabolite that is used for salvagetherapy in patients failing multiple antiretroviral regimens.When this agent is added to nucleoside analogue therapy,the incidence of peripheral neuropathy is increased [38].When combined with didanosine and/or stavudine,hydroxyurea increases the risk of DSP by several fold [39].The combination of ddI plus d4T also increases the risk ofneuropathy, but not to the same extent as when hydrox-yurea is added [20•,38]. It is thought that hydroxyureasupports the integration of ddI, and possibly also d4T, intothe mitochondrial matrix, thereby increasing the neuro-toxicity of the latter two medications [20•].

Table 2. Nucleoside analogue-related neuropathy

AgentDideoxynucleoside analogues Stavudine Didanosine Zalcitabine Synergistic effect from hydroxyureaDiagnosis Improvement after withdrawal of offending drug ? Elevated serum lactate levels Treatment Dosage Nonsteroidal anti-inflammatory agents Variable dose, depending on agent Acetaminophen 500–1000 mg every 4 to 6 hours Amitriptyline Up to 150 mg/day, at bedtime Carbamazepine 200–400 mg three times daily Phenytoin 100 mg three times daily Gabapentin 300–1200 mg three times daily Lamotrigine 200–250 mg twice daily Topical analgesics Lidoderm patch, ointment As needed Narcotic analgesics Oxycodone, morphine, fentanyl Variable dose

Page 4: AIDS and AIDS-treatment neuropathies

128 Neuropathic Pain

Plasma HIV Viral Load and CD4+ Cell CountThere is increasing evidence that elevated plasma-HIV RNAlevels may be a risk factor for the development of DSP[3••,19••]. In a retrospective review of 1604 AIDS-freeHIV-positive men from the Multicenter AIDS CohortStudy, Childs et al. [19••] showed that as HIV viral loadincreased, so too did the incidence of sensory neuropathy,and that individuals with viral loads greater than 10,000copies/mL had a greater than twofold increased hazard forDSP [19••]. There was also a trend for increased neuro-pathy risk by over 100% in men with CD4+ counts of lessthan 750 cells/mm3, but the relationship was much weakerthan for plasma HIV viral load.

TreatmentCurrently, there are no treatment modalities that reversethe pathology of HIV-associated DSP. The management ofpolyneuropathy in patients infected with HIV focuses onrelief of pain [40]; therefore, therapy is aimed largely atsymptom reduction. To this end, management of DSPemploys various pain-relieving modalities that includemedications to directly control the pain and approachesdirected at the source of the pain. Additionally, as notedearlier, HAART is effective in improving nerve function inDSP [3••] and decreasing the incidence of DSP [19••],even when neurotoxic drugs are part of the regimen. Themost recent debate in this area centers on the question ofthe utility of antiretroviral therapy for HIV RNA suppres-sion at the expense of neurotoxicity. Data from the AIDSClinical Trials Group (ACTG) 291 study of recombinanthuman nerve growth factor indicate that in a cohort of 236patients, suppression of plasma HIV RNA by nucleosideanalogue drugs resulted in lower maximum and globalpain scores [41]. These data suggest that virologic controlmight outweigh the potential for dideoxynucleosidetoxicity. Prospective studies are needed to furtherassess this hypothesis.

Discontinuation of neurotoxic agentsAs a first step, metabolic causes of neuropathy, such asunderlying diabetes mellitus, vitamin B12 deficiency, andother forms of neuropathy seen in HIV disease, should beidentified and corrected where possible. Identification andtreatment of alcohol abuse is imperative. Next, consider-ation should be given to neurotoxic medications such asddI, ddC, and d4T as a cause of DSP, and a trial of dosereduction or discontinuation of the offending agent(s)may be beneficial. If the decision is made to stop a nucleo-side analogue medication, the clinician and patient shouldbe aware that it may take 4 to 8 weeks for symptoms tosubside, and during this time the pain may actuallyincrease. This is referred to as the "coasting period" [17]. Insome instances, resolution of pain may take as long as 16weeks. Only about two thirds of patients will improve withantiretroviral drug discontinuation [42].

All patients on nucleoside analogue drug therapy forHIV should be assessed frequently for signs or symptomsof DSP, as this complication may be most easily reversedwhen it is diagnosed early [8]. DSP can often be preventedby modifying the drug dose or by changing to less neuro-toxic antiretroviral agents [42].

AnalgesicsAnalgesics are commonly prescribed for the treatment ofDSP. Clinicians might follow the step-wise recom-mendations of the World Health Organization for themanagement of chronic pain [43]. Some patients with mildsymptoms may respond to nonsteroidal anti-inflammatorydrugs or acetaminophen [9]. For refractory pain, treatmentwith topical lidocaine or capsaicin 0.075% may bebeneficial. A 5% lidocaine patch (Lidoderm; ENDO Phar-maceuticals, Chadds Ford, PA) was recently approved by theFood and Drug Administration for treatment of postherpeticneuralgia, and it has also shown efficacy in treating DSP-related pain [44]. In a small, open-label study of 30 patientswith DSP, application of either a 5% lidocaine patch orlidocaine gel applied directly to painful sites demonstratedthat this medication was effective in providing pain relief[44]. Although use of a topical anesthetic appears to besafe in treating the pain due to DSP, it will not be useful inpatients for whom pain is not a primary symptom. Analysisof data from a larger, double-blind, placebo-controlledstudy is underway. Ultimately, some patients with DSP maybe in such severe and debilitating pain that narcotic anal-gesia is required to provide relief.

Adjuvant therapySeveral medications with putative effects on nerve mem-branes have been investigated for the treatment of DSP. Tri-cyclic antidepressants, such as amitriptyline or desipramine,at doses of up to 150 mg/day can be used. The anti-arrhythmic drug, mexilitine, was investigated due to itsinhibitory effects on sodium conductance at the axonalmembrane. However, neither amitriptyline nor mexilitinewere superior to placebo in a placebo-controlled, double-blind trial [45]. Anticonvulsants, such as phenytoin and car-bamazepine, reduce neuropathic pain in patientswith diabetic neuropathy, and anecdotal experiencesuggests that they may also be effective in the treatmentof DSP. Oxcarbazepine, the dihydroketo analogue of carb-amazepine, potentially has a better pharmacokineticprofile in patients with HIV infection who are taking antiret-rovirals with extensive hepatic metabolism. Oxcarbazepinehas no autoinduction and low protein binding (NovartisPharmaceuticals, unpublished data). Clinical trials to assessthe efficacy of this drug are being planned. Lamotrigine andgabapentin are newer anticonvulsants that show promise inthe treatment of DSP. Gabapentin is an attractive therapy forpainful neuropathy due to its favorable pharmacokineticprofile and sparse drug-drug interactions. Reports of its effec-tiveness in AIDS-associated DSP are anecdotal [46], but it

Page 5: AIDS and AIDS-treatment neuropathies

AIDS and AIDS-treatment Neuropathies • Williams et al. 129

has been used successfully in other neuropathic conditions,particularly diabetes [47].

Lamotrigine, however, appears to be more effective intreating DSP-related pain, as demonstrated in a pilot studyby Simpson et al. [48]. The most common side effectreported from this study was the occurrence of an allergicrash, which resulted in premature discontinuation of studydrug in 25% of patients [10,48]. We have recently concludeda large, multicenter, double-blind, placebo-controlled studyof lamotrigine. In this study, the drug was gradually intro-duced over an 8-week period until a dosage of 400 mg/daywas attained. Although the data collected in this study arebeing analyzed, it is believed that lamotrigine will be bettertolerated when introduced gradually [10].

Recombinant human nerve growth factorRecombinant human nerve growth factor (rhNGF) hasbeen investigated as a therapy that might reverse or arrestthe neuropathic changes of DSP. Ample laboratory dataand studies of animal models point to rhNGF as a poten-tial treatment for peripheral neuropathy, in particulardiabetic and HIV-related neuropathies [49].

In a placebo-controlled ACTG study of 270 patientswith painful HIV neuropathy, subcutaneously adminis-tered rhNGF improved Gracely Pain Scale scores and sensi-tivity to pinprick. However, in that study, most secondarymeasures of nerve regeneration, including quantitativesensory tests and epidermal skin biopsies, did not improve[50]. In spite of these encouraging results, future develop-ment of rhNGF is uncertain, because Phase III studies ofrhNGF in diabetic neuropathy were negative.

Other therapiesSmall studies of capsaicin for painful HIV neuropathy havebeen generally unsuccessful [51]. A more recent study byPaice et al. [52] concluded that capsaicin is ineffective, as itshowed no significant differences in pain relief, sensoryperception, quality of life, or mood [52]. Acupuncture hasshown no significant difference in effectiveness in HIV-associated painful DSP, as compared with sham acupunc-ture points, which were used as placebo [53].

ConclusionsDistal symmetrical polyneuropathy is a frequent neuro-logic complication of HIV infection. Recent advances inantiretroviral therapy have led to an increasingly large poolof individuals who are surviving long enough to become atincreased risk for neurologic complications, such as DSP.Additionally, neurotoxic medications, such as the dideoxy-nucleoside drugs that are widely used in combinations ofHAART regimens, increase the risk of development of pain-ful neuropathy. Current research focuses on identifyingindividuals most at risk for DSP while taking neurotoxicHAART regimens, and on mechanisms of mitochondrialinjury that can lead to neuronal damage.

References and Recommended ReadingPapers of particular interest, published recently, have been highlighted as:• Of importance•• Of major importance

1. d’Arminio Monforte A, Duca PG, Vago L, et al.: Decreasing incidence of CNS AIDS-defining events associated with antiretroviral therapy. Neurology 2000, 54:1856–1859.

2. Clifford DB, Yiannoutsos C, Glicksman M, et al.: HAART improves prognosis in HIV-associated progressive multifocal leukoencephalopathy. Neurology 1999, 52:623–625.

3.•• Martin C, Solders G, Sonnerborg A, et al.: Antiretroviral therapy may improve sensory function in HIV-infected patients: a pilot study. Neurology 2000, 54:2120–2127.

This pilot study of 49 HIV-infected patients was the first to demon-strate that lowering plasma HIV RNA viral load with antiretroviral therapy improves objective parameters of sensory nerve function.

4. Sacktor NC, Lyles RH, Skolasky RL, et al.: Combination anti-retroviral therapy improves psychomotor speed performance in HIV-seropositive homosexual men. Multicenter AIDS Cohort Study (MACS). Neurology 1999, 52:1640–1647.

5. Wulff EA, Simpson DM: Neuromuscular complications of the human immunodeficiency virus type 1 infection. Semin Neurol 1999,19:157–164.

6. Bacellar H, Munoz A, Miller EN, et al.: Temporal trends in the incidence of HIV-1-related neurologic diseases: Multicenter AIDS Cohort Study, 1985–1992. Neurology 1994, 44:1892–1900.

7.• Tagliati M, Grinnell J, Godbold J, Simpson DM: Peripheral nerve function in HIV infection: clinical, electrophysio-logical, and laboratory findings. Arch Neurol 1999, 56:84–89.

This study comprehensively assessed 251 consecutive HIV-infected individuals and evaluated the multifactorial etiologies of painful polyneuropathy.

8. Marra CM, Boutin P, Collier A, et al.: Screening for distal sensory peripheral neuropathy in HIV-infected persons in research and clinical settings. Neurology 1998, 51:1678–1681.

9. Simpson DM, Tagliati M: Nucleoside analogue-associated peripheral neuropathy in human immunodeficiency virus infection. J Acquir Immune Defic Syndr Hum Retrovirol 1995, 9:153–161.

10. Wulff EA, Wang AK, Simpson DM: HIV-associated peripheral neuropathy: epidemiology, pathophysiology and treatment. Drugs 2000, 59:1251–1260.

11. Williams D, Shah NR, Simpson DM: HIV-associated neuro-muscular syndromes. In Approach Towards Management of Human Immunodeficiency Virus Infection and Its Related Complications. Edited by Singh N, Swindells. 2001, in press.

12. de la Monte SM, Gabuzda DH, Ho DD, et al.: Peripheral neuropathy in the acquired immunodeficiency syndrome. Ann Neurol 1988, 23:485–492.

13. Mah V, Vartavarian LM, Akers MA, et al.: Abnormalities of peripheral nerve in patients with human immunodeficiency virus infection. Ann Neurol 1988, 24:713–717.

14. Cornblath DR, McArthur JC: Predominantly sensory neuropathy in patients with AIDS and AIDS-related complex. Neurology 1988, 38:794–796.

15. McCarthy BG, Hsieh ST, Stocks A, et al.: Cutaneous innervation in sensory neuropathies: evaluation by skin biopsy. Neurology 1995, 45:1848–1855.

16. McArthur JC, Stocks EA, Hauer P, et al.: Epidermal nerve fiber density: normative reference range and diagnostic efficiency. Arch Neurol 1998, 55:1513–1520.

17. Berger AR, Arezzo JC, Schaumburg HH, et al.: 2’,3’-dideo-xycytidine (ddC) toxic neuropathy: a study of 52 patients. Neurology 1993, 43:358–362.

18. Bouhassira D, Attal N, Willer J, et al.: Painful and painless peripheral sensory neuropathies due to HIV infection: a comparison using quantitative sensory evaluation. Pain 1999, 80:265–272.

Page 6: AIDS and AIDS-treatment neuropathies

130 Neuropathic Pain

19.•• Childs EA, Lyles RH, Selnes OA, et al.: Plasma viral load and CD4 lymphocytes predict HIV-associated dementia and sensory neuropathy. Neurology 1999, 52:607–613.

This large analysis of over 1600 HIV-seropositive men without AIDS from the Multicenter AIDS Cohort demonstrated a relationship between elevated plasma HIV-RNA viremia and a greater than twofold increase in the hazard for decreased sensory perception.20.• Moore RD, Wong WM, Keruly JC, et al.: Incidence of neuro-

pathy in HIV-infected patients on monotherapy versus those on combination therapy with didanosine, stavudine and hydroxyurea. AIDS 2000, 14:273–278.

This study demonstrated the synergistic effect of combination antiretroviral therapy versus monotherapy on the incidence of decreased sensory perception.21. Figg WD: Peripheral neuropathy in HIV patients after

isoniazid therapy initiated. DICP 1991, 25:100–101.22. Gill P, Rarick M, Bernstein-Singer M, et al.: Treatment of

advanced Kaposi’s sarcoma using a combination of bleomy-cin and vincristine. Am J Clin Oncol 1990, 13:315–319.

23. Ochonisky S, Verroust J, Bastuji-Garin S, et al.: Thalidomide neuropathy incidence and clinico-electrophysiologic findings in 42 patients. Arch Dermatol 1994, 130:66–69.

24. Famularo G, Moretti S, Marcellini S, et al.: Acetyl-carnitine deficiency in AIDS patients with neurotoxicity on treatment with antiretroviral nucleoside analogues. AIDS 1997, 11:185–190.

25. Yoshioka M, Shapshak P, Srivastava AK, et al.: Expression of HIV-1 and interleukin-6 in lumbosacral dorsal root ganglia of patients with AIDS. Neurology 1994, 44:1120–1130.

26. Moyle GJ, Sadler M: Peripheral neuropathy with nucleoside antiretrovirals: risk factors, incidence and management. Drug Safety 1998, 19:481–494.

27. Dubinsky RM, Yarchoan R, Dalakas M, et al.: Reversible axonal neuropathy from the treatment of AIDS and related disorders with 2’,3’-dideoxycytidine (ddC). Muscle Nerve 1989, 12:856–860.

28. Browne MJ, Mayer KH, Chafee SB, et al.: 2’,3’-didehydro-3’-deoxythymidine (d4T) in patients with AIDS or AIDS-related complex: a phase I trial. J infect Dis 1993, 167:21–29.

29. Simpson DM, Katzenstein DA, Hughes MD, et al.: Neuro-muscular function in HIV infection: analysis of a placebo-controlled combination antiretroviral trial. AIDS Clinical Group 175/801 Study Team. AIDS 1998, 12:2425–2432.

30. Pike IM, Nicaise C: The didanosine expanded access program: safety analysis. Clin Infect Dis 1993, 16(suppl 1):S63–S68.

31. Kakuda TN: Pharmacology of nucleoside and nucleotide reverse transcriptase inhibitor-induced mitochondrial toxicity. Clin Ther 2000, 22:685–708.

32. Pan-Zhou XR, Cuil JR, Zhou XJ, et al.: Differential effects of antiretroviral nucleoside analogues on mitochondrial function in HepG2 cells. Antimicrob Agents Chemother 2000, 44:496–503.

33. Brinkman K, Hofstede HJ, Burger DM, et al.: Adverse effects of reverse transcriptase inhibitors: mitochondrial toxicity as common pathway. AIDS 1998, 12:1735–1744.

34.• Brinkman K, Smeitink JA, Romijn JA, et al.: Mitochondrial tox-icity induced by nucleoside-analogue reverse-transcriptase inhibitors is a key factor in the pathogenesis of anti-retroviral-therapy-related lipodystrophy. Lancet 1999, 354:1112–1115.

This article proposes mitochondrial toxicity as the etiology of antiretroviral (ARV)-related lipodystrophy and presents evidence that may support the developing hypothesis that neuropathy is included in the spectrum of disorders that may result from ARV-induced mitochondrial toxicity.

35. Larsson NG, Tulinius MH, Holme E, et al.: Segregation and manifestations of the mtDNA tRNA(Lys) A--->G(8344) mutation of myoclonus epilepsy and ragged-red fibers (MERRF) syndrome. Am J Hum Genet 1992, 51:1201–1212.

36. Mhiri C, Baudrimont M, Bonne G, et al.: Zidovudine myopathy: a distinctive disorder associated with mito-chondrial dysfunction. Ann Neurol 1991, 29:606–614.

37. Hashimoto KI, Tsunoda R, Okamoto M, et al.: Stavudine selectively induces apoptosis in HIV type 1-infected cells. AIDS Res Hum Retroviruses 1997, 13:193–199.

38. Cepeda JA, Wilks D: Excess peripheral neuropathy in patients treated with hydroxyurea plus didanosine and stavudine for HIV infection. AIDS 2000, 14:332–333.

39. Moyle G: Clinical manifestations and management of anti-retroviral nucleoside analog-related mitochondrial toxicity. Clin Ther 2000, 22:911–936.

40. Wulff EA, Simpson DM: HIV-associated neuropathy: recent advances in management. HIV Adv Res Ther 1998, 8:23–29.

41. Katzenstein D, Haidich AB, Simpson D, et al., for the ACTG 291 Study Team: HIV-1 RNA levels, antiretroviral therapy, and distal sensory polyneuropathy in participants of ACTG 291 [abstract]. J Neurovirol 2000, 6:428.

42. Blum AS, Dal Pan GJ, Feinberg J, et al.: Low-dose zalcitabine-related toxic neuropathy. Neurology 1996, 46:999–1003.

43. Grond S, Zech D, Schug SA, et al.: Validation of World Health Organization guidelines for cancer pain relief during the last days and hours of life. J Pain Symptom Management 1991, 6:411–422.

44. Dorfman D, Dalton A, Khan A, et al.: Treatment of painful distal sensory polyneuropathy in HIV-infected patients with a topical agent: results of an open-label trial of 5% lidocaine gel. AIDS 1999, 13:1589–1590.

45. Kieburtz K, Simpson D, Yiannoutsos C, et al.: A randomized trial of amitriptyline and mexiletine for painful neuropathy in HIV infection. AIDS Clinical Trial Group 242 Protocol Team. Neurology 1998, 51:1682–1688.

46. Newshan G: HIV neuropathy treated with gabapentin. AIDS 1998, 12:219–221.

47. Rosner H, Rubin L, Kestenbaum A: Gabapentin adjunctive therapy in neuropathic pain states. Clin J Pain 1996, 12:56–58.

48. Simpson DM, Olney R, McArthur JC, et al.: A placebo-controlled trial of lamotrigine for painful HIV-associated neuropathy. Neurology 2000, 54:2115–2119.

49. Rask CA: Biological actions of nerve growth factor in the peripheral nervous system. Eur Neurol 1999, 41:14–19.

50. McArthur JC, Yannoutsos C, Simpson DM, et al.: A phase II trial of nerve growth factor for sensory neuropathy assoc-iated with HIV infection. AIDS Clinical Trials Group Team 291. Neurology 2000, 54:1080–1088.

51. Simpson DM, Olney RK: Peripheral neuropathies associated with human immunodeficiency virus infection. In Peripheral Neuropathies: New Concepts and Treatments. Edited by Dyck PJ. Philadelphia: WB Saunders;1992:685–711.

52. Paice JA, Estwing-Ferrans C, Lashley FR, et al.: Topical capsaicin in the management of HIV-associated peripheral neuropathy. J Pain Symptom Management 2000, 19:45–51.

53. Shlay JC, Chaloner K, Max MB, et al.: Acupuncture and amitriptyline for pain due to HIV-related peripheral neuro-pathy: a randomized controlled trial. Terry Beirn Community Programs for Clinical Research on AIDS. JAMA 1998, 280:1590–1595.