CRITICAl REVIEW

Transcutaneous Electrical Nerve Stimulation: Basic Science Mechanisms and Clinical Effectiveness

Kathleen A. Sluka* and Deirdre Walsh t

Abstract: Transcutaneous electrical nerve stimulation (TENS) is used clinically by a variety of health care professionals for the reduction of . Clinical effectiveness of TENS is controversial, with some studies supporting whereas others refute its clinical use. Although used by health professionals for decades, the mechanisms by which TENS produces analgesia or reduces pain are only recently being elucidated. This article describes the basic science mechanisms behind different frequencies of TENS stimulation. Specifically, we describe the literature that supports the use of different frequencies and intensities of TENS. We further describe theories that support the use of TENS such as the gate control theDly and the release of endogenous opioids. The literature that supports or refutes each of these theories is described. We also review the clinical literature on TENS effectiveness and elucidate the problems with clinical research studies to date. In wnclusion, TENS is a noninvasive modality that is easy to apply with relatively few contraindications. However, the clinical efficacy of TENS will remain equivocal until the publication of sufficient numbers of high quality, randomized, controlled clinical trials. © 2003 by the American Pain Society Key words: Pain, , analgesia, electrical stimulation

What Is TENS? demonstrated that high frequency (100 Hz) stimulation at an intensity that activates large afferent fibers re­ ranscutaneous electrical nerve stimulation (TENS) is duced neuropathic pain in 8 patients. Although used defined by the American Physical Therapy Associa­ clinically for more than 30 years, the mechanisms by Ttion as the application of electrical stimulation to which TENS produces pain relief were not known. Sev­ the skin for pain control. TENS is noninvasive, inexpen­ eral theories support the use of TENS including the gate sive, safe, and easy to use.' Electricity has been used for control theory of pain and release of endogenous opi­ thousands of years for relief of pain, with the first writ­ aids. This review will focus on the current and previous ten documentation by Aristotle'S literature that has begun to elucidate the basic science In the mid-1800s and early 1900s a number of physi­ mechanisms of TENS and how these mechanisms can be cians and dentists reported the use of electricity as an applied to the clinic. We will also review the clinical lit­ and anesthetic. However, electrical stimulation erature on TENS and elu.cidate the problems with clinical for pain relief was not fully accepted by the medical field research studies to date. until the publication by Wall and Sweet"O in 1967 in re­ Clinically, TENS is applied at varying frequencies, inten­ sponse to the gate theory of pain." Wall and Sweet sities, and pulse durations of stimulation. Frequency of stimulation is broadly classified as high frequency (>50 Hz), low frequency «10 Hz), or burst (bursts of high Received September 12, 2002; Revised November 13, 2002; Accepted No­ frequency stimulation applied at a much lower fre­ vember 13, 2002 From the "'Physical Therapy and Rehabilitation Sciences Graduate Pro­ quency) TENS. Intensity is determined by the response of gram, Neuroscience Graduate Program, Pain Research Program, Univer­ the patient as either sensory level TENS or motor level sity of Iowa, Iowa City, lA, and'RehabHitation Sciences Research Group, School of Rehabilitation Sciences, University of Ulster, Ulster, United TENS. In addition, some clinicians use stimulation below Kingdom. a sensory intensity termed microcurrent electrical stimu­ Supported by the Arthritis Foundation and K02 AR02201 lation. To date, there are no data to support microcur­ Address reprint requests to Kathleen A. Sluka, PT, PhD, 100 Medical Ed~ u(ation Building, 1-242, Physical Therapy and Rehabilitation Science rent electrical stimulation. With sensory level TENS the Graduate Program, University of Iowa, Iowa City, fA 52242. E~mail: voltage (ie, amplitude) is increased until the patient feels [email protected] a comfortable tingling (perceived with high frequency) © 2003 by the American Pain Society 1526-5900/2003 $30.00 +- 0 or tapping (perceived with low frequency) sensation doi: 10.1 054/jpai,2003.434 without motor contraction. This amplitude is referred to

The Journal of Pain, Vol 4, No 3 (April), 2003: pp 109-121 109 110 TENS review as low intensity. With motor level TENS the intensity is ned as acute pain models, inflammatory pain models, increased to produce a motor contraction. Usually the and neuropathic pain models. TENS has been used in all intensity is increased to the maximal level before becom­ of these conditions. Acute pain models have been used ing noxious. This is referred to as high intensity TENS. In for decades as screening tools to test the efficacy of phar­ general, high frequency TENS is applied at low intensities macologic agents and do not produce tissue injury'» The and is referred to as conventional TENS. In contrast, low response to noxious heat, mechanical or electrical stim~ frequency TENS is typically applied at high intensities so ulation is assessed in acute pain models. Models oftissue that a motor contraction is produced. This mode of stim­ injury were developed later to more directly measure ulation is referred to as strong, low rate, or acupuncture­ pain that might be similar to clinical syndromes. Hyper­ like TENS. Stimulus strength duration curves for applica­ algesia, an increased response to a , oc­ tion of TENS to the skin demonstrate that sensory level curs in response to tissue injury.95 Hyperalgesia can occur TENS occurs with the lowest amplitude, followed by mo­ at the site of injury, termed primary hyperalgesia, and is 63 91 tor contraction and then noxious sensation. • thought to reflect changes in primary afferent fibers, Numerous studies have attempted to determine the although central neuronal changes will influence pri­ effectiveness otTENS treatment for people with a variety mary hyperalgesia 95 Secondary hyperalgesia develops of pain conditions. 52.5~V:d,62.,85 However, the clinical Ilter­ outside the site of injury and is thought to reflect an ature on TENS is controversial. Although the majority of increase in central neuron excitability."-' Both primary studies support the use of TENS, a number of studies and secondory hyperalgesia occur in response to heat refute its effectiveness. Several factors may contribute to and mechanical stimuli after tissue injury.26,41,43,61.81 this controversy. Many early studies did not use a placebo Carrageenan COn be injected into the paw or knee joint control but rather compared their results to patients to produce an acute inflammatory event resulting in hy­ 8 who did not receive any treatment. TENS itself has a peralgesia ' The carrageenan model has been well char­ significant placebo effect. A number of studies com­ acterized neurophysiologically with increased firing and pared the effectiveness of TENS to other treatments in­ sensitivity of , increased receptive field size of cluding modalities, exercise, and various pharmacologic spinal neurons, and increased firing and sensitivity of treatments. Although TENS may not be more effective spinal dorsal horn neurons.69 Injection of complete than these treatments. it may be equally effective. Stim­ Freund's adjuvant, either systemically or into a joint, is a ulation parameters, ie, frequency, intensity, and pulse model of chronic inflammation similar to rheumatoid duration, are commonly not specified or not kept con­ arthritis.'· Several models of neuropathic pain have stant among patients within a given study. Placement of been developed and are used extensively. The 2 most electrodes varies considerably between studies such that common models are the Bennett model induced by mak­ some studies place electrodes at the site of injury, some ing loose ligations around the sciatic nerve5 and the within the dermatome, and others proximal to the site of Chung model induced by making tight ligations around injury. Furthermore, patient populations vary between the spinal nerves40 Each of these neuropathic pain mod­ studies and within studies, making it difficultto interpret els produces 0 measurable long-lasting hyperalgesia and the appropriate patient population who would benefit changes in the central nervous system. from TENS treatment. Last, a variety of different out­ Effects of TENS were analyzed in several animal mod­ come measures are used to assess the effectiveness of els. Early studies used acute tests such as the tail flick TENS including subjective pain rating scales, joint func­ response to noxious heat and hot plate test to examine 96 tion, analgesic intake, primary hyperalgesia (increased the effects of TENS. Specificolly, Woolf et 01 ,97 demon­ responsiveness to nociceptive stimuli at the site of inju­ strated that the tail flick latency to heat increased (ie, ry), secondary hyperalgesia (increased responsiveness to analgesia) after treatment with electrical stimulation at nociceptive stimuli outside the site of injury), and various high frequencies that activate A fibers (Fig 1A). Inhibi­ questionnaire outcomes measures. It is entirely possible tion by TENS still occurs in animals that have been spinal­ 73 that TENS is effective on some measures of pain or func­ ized to remove descending inhibition. •97 However, the tion and ineffective for others. To overcome many of inhibition ofthe tail flick reflex by high frequency TENS is these shortcomings in the clinical research design, animal not as great in spinalized animals os compared to intact models of pain have been used to assess effects of vary­ animals, suggesting both segmental and descending in­ ing parameters and different outcome measures. Animal hibition are involved in the analgesia produced by high models minimize the placebo effect, control the extent frequency TENS 97 (Fig 1A). and type of injury, and maintain application of TENS Another measure of nociceptive activity is to record constant between subjects. In addition, animal models of activity of ventral roots or of neurons located in the spi­ pain allow one to assess the neurobiologic mechanisms nal cord. Neurons in the that respond to nox­ by which TENS produces a reduction in pain behaviors. ious stimuli include (1) high threshold neurons, which exclusively respond to noxious stimuli, and (2) wide dy­ TENS in Animal Models namic range neurons, which respond to both innocuous and noxious stimulation95A-fiber conditioning stimula­ Several animal models of pain exist, are used to mea­ tion (TENS) reduces (1) activation of ventral roots by C­ sure effectiveness of pharmaceutical agents, and mimic fiber stimulation" and (2) activity of dorsal horn neu­ 4 clinical conditions.'· These models can broadly be classi- rons.2 ,25,44 cells transmit pain and CLINICAL REVIEW/Sluka et al 111

150 A INT.6£T SPINAL A 40

~ '"0- E 100

D€GREE OF ANALGf:SIA 1%1 50 100 150 Time...,..., (5) " 110Hz, ,30mA , Pinch

,l, B 0 ....L CONTROL STiM 'dORPHINE CONTRct. 'TN ",;p- SOH, 7,5I!l9f~9 SOH, 1,5m;/k; O,Z5mS O,25mS N. 10 10 6

150 rlTACT RATS PCPAPRETRU,TED B 1350lf\9h;l lNTACTRATS

50 100 150 200 250 Time Is) 100 ~ ~ ----­25Hz 15Hz ----­10Hz 5Hz DEGREEOf' spon~ ANALGESIA Figure 2. Oscilloscope trace of action potentials from a taneously firing dorsal horn neuron that responds to noxious "" stimuli. Spontaneous firing of the neuron is reduced by TENS. Increasing frequency of stimulation results in a greater inhibi~ " tion of spontaneous activity. The bottom figure shows the re~ sponse of a dorsal horn neuron that responds to pinch. An in~ crease in the number of action potentials occurs when a noxious mechanical stimulus is applied to the skin (pinch, long bar). TENS application to the receptive reduces the pinch response of the neuron. Once TENS is removed, the pinch response returns. .Re~ 0 printed from Garrison DW, Foreman RD: Decreased activity of tONTRCl sr. MOIlI'lH: ,ONTllOL sr. MOOPH'" spontaneous and noxiously evoked dorsal horn cells during SOH, 7.511l9/~Q SOH, 7,5 mtV kq O,25mS 0,"'" transcutaneous electrical nerve stimulation (TENS). Pain 58:309~ 315,1994 with permission of Elsevier Science Publishers. N' 10 10 • 12 , Figure 1. (A) Bar graphs represent the analgesia produced by 24 44 TENS and in the for animals that are quency TENS. ,25A4 However, Lee et al demonstrated intact and thosethat were spinalized. Fifty~hertz electrical stim­ that low frequency TENS produced a greater inhibition ulation produced an increase in the tail flick latency similar to than high frequency TENS. Further, intensity at 3X the that of systemic morphine. Spinal transection reduced the amount of inhibition by electrical stimulation or morphine by threshold to activate All fibers was ineffective. but in­ approximately 50%. (B) Bar graphs represent the analgesia pro~ creasing to a strength that activated AS nociceptors pro­ duced by 50~Hz electrical stimUlation or morphine in the tail duced a greater inhibition.44 In contrast, Garrison and flick test in intact animals. Animals pretreated with para~chlo~ Foreman25 recorded from dorsal horn neurons in cats rophenylalanine (PCPA) to deplete the neurotransmitter sero~ tenin (5~HT) showed a significant reduction in the amount of and examined the effect of varying frequency, intensity, analgesia produced by either electrical stimulation or mor­ and pulse duration on the inhibition of dorsal horn cell phine. Control animals did not receive electrical stimulation or activity by TENS (Fig 2). Specifically, increasing intensity, morphine but were still spinalized or pretreated with PCPA. Reprinted from Woolf CJ, Mitchell D, Barrett GD: Antinocicep~ frequency, or pulse duration increases the amount of tive effect of peripheral segmental electrical stimulation in the inhibition of dorsal horn neurons produced by TENS. In rat. Pain 8:237-252, 1980 with permission of Elsevier Science addition. the effects of TENS on dorsal horn cells are Publishers. short lasting, returning to normal after removal of the TENS. Similarly, increasing intensity of stimulation to activate A8 fibers increases the inhibition of the flex­ temperature information from the spinai cord to the ion reflex with either iow or high frequency stimula­ thalamus and are both high threshold and wide dynamic tion parameters."·74 These data suggest that high and range neurons.95 low frequency TENS are effective, increasing intensity The spontaneous activity and noxious input to spino­ increases inhibition, and the effects of TENS are short thaiamic tract cells are inhibited by low and high fre- lasting. 112 TENS review In one study, effect of electrode placement was evalu­ ated by placing electrodes within the receptive field for a Central Control spinothalamic tract neuron, outside the receptive field of the neuron but on the same limb, and at the mirror Large Fiber site." The greatest degree of inhibition of spinothalamic tract cell activity occurred with electrodes placed within the receptive field for the neuron, and only minimal in­ Input ~TCel1 _ Action hibition occurred when placed on the same hind limb but System outside the receptive field.44 Behaviorally, in animals without tissue injury, TENS applied to the knee joint has no e'flect on the paw withdrawal latencyn These data Small fiber suggest that electrode placement is important and that the greatest effect will occur if given at the site of injury Figure 3. Diagram showing the gate control theory of pain as at which one would be expected to affect the receptive originally described by Melzack and Wall, 1965. T cell is an as~ fields of sensitized neurons. cending neuron that could be from the spinothalamic, spinore~ ticular, or spinomesencephalic tract. SG is a neuron in the sub~ TENS, however, is not given to people without pain so stantia gelatinosa (laminae II) of the spinal cord dorsal horn. later studies began to use well-established animal mod­ Open triangles represent excitatory synapses and closed trian­ els of pain to test TENS effectiveness. After injection of gles represent inhibitory synapses. Large Fiber represents input carrageenan into the paw a localized acute inflamma­ from large diameter primary afferent fibers in the peripheral nervous system, and Small fiber represents input from small tory event occurs. Response to heat and mechanical stim­ diameter nociceptive primary afferent fibers in the peripheral uli on the paw at the site of the inflammation is used to nervous system. The general concept is that small diameter fi· measure primary hyperalgesia. Modulation of frequency bel's excite cells in the spinal cord that send information to (4 vs 100 Hz), intensity (sensory vs motor), or pulse dura­ higher centers for the perception of pain. Large diameter fiber input reduces noxious input of nociceptors by activation of in~ tion (100 vs 2S0 ILSec) demonstrated a frequency, but not hibitory neurons in the substantia gelatinosa of the spinal cord. intensity or pulse duration, dependent effect on primary Reproduced from Melzack R, Wall PD: Pain mechanisms: A new hyperalgesia to mechanical and heat stimuli in animals theory. Science 150:971 ~978, 1965 with permission of the Amer~ ican Association of the Advancement of Science. with carrageenan paw inflammation.'" In this study only animals treated with high frequency TENS at the site of inflammation showed a reduction in primary hyperalge­ sia, and this reduction was minima!.28 In contrast, injec­ dorsal horn neurons before and after application of low tion of kaolin and carrageenan into the knee joint is used frequency, motor intensity TENS and compared these ef­ to measure secondary hyperalgesia on the paw. Treat­ fects to those from animals without tissue injury. TENS ment of the infiamed knee joint with either high or low reduced the responsiveness to noxious mechanical stim­ frequency TENS at sensory intensity produced an equai ulation of dorsal horn neurons in both normal and neu­ and dramatic reversai of heat and mechanical hyperalge­ ropathic animals. However, the responsiveness of spinal 41 sia. ,n Increasing intensity did not further increase the neurons to innocuous mechanical stimulation was only analgesia produced by either high or low frequency inhibited by TENS in neuropathic animals,45 TENS." Interestingly, there is a long-lasting reduction in hyperalgesia that persists for 12 to 24 hours for both primary and secondary hyperalgesia models with carra­ Theories of TENS 41 geenan inflammation. ,77 The increased responsiveness Several theories are used to support the use of TENS. of dorsal horn neurons to innocuous and noxious me­ The gate control theory of pain is most commonly used chanical stimuli that occurs after inflammation is equally to explain the inhibition of pain by TENS (Fig 3). Accord­ reduced after either high or low frequency TENS treat­ ing to the gate control theory of pain, stimulation of ment applied to the inflamed paw.'6 This reduction in large diameter afferents by TENS inhibits nociceptive fi­ sensitization of high threshold and wide dynamic range ber evoked responses in the dorsal horn. 51 The gate con­ dorsal horn neurons parallels the effects of TENS on sec­ trol theory is thought to involve segmental inhibition by ondary hyperalgesia46 When measuring secondary hy­ using neurons located in the substantia gelatinosa of the peralgesia or dorsal horn neuron activity, responses are spinal cord dorsal horn. However, the original theory did reduced back toward preinflammation responses by suggest that descending inhibitory pathways might exist TENS but not beyond basal responses. and that these spinal neurons are under descending in­ With the Bennett model of neuropathic pain, Somers fluences. Specific neurotransmitters or their receptors and Clemente"' demonstrated that high frequency, low were not suggested at the time because we were only (sensory) intensity TENS stimulation over the paraspinal beginning to understand the pharmacology of the ner­ musculature reduced heat but not mechanical hyperal­ vous system. Thus, the gate control theory can be inter­ gesia that normally occurs in this model. This inhibition preted broadly, There are now much more detailed data of heat hyperalgesia only occurs if TENS was started the on mechanisms of actions of TENS that include anatomic first day after injury but not if started 3 days after inju­ pathways, neurotransmitters and their receptors, and ryB' Following spinal nelve ligation (Chung model of the types of neurons involved in the inhibition, Several neuropathic pain), Leem et al45 recorded responses of studies support segmentally mediated inhibition mecha- CLlI~IC:AL !'[VIEW/Sluka et al 113 nisms in TENS analgesia. High frequency TENS inhibition is partially prevented by spinalization, which removes Thalamus descending inhibitory influences.97 However, a signifi­ cant amount of inhibition remains after spinalization. Thus, TENS appears to produce both segmental and de­ scending inhibition. Alternatively, Campbell and Taub B suggested that high frequency stimulation by TENS results in conduction block or fatigue of M fibers. However, Janko and Tron­ tel?' and Lee et al 44 demonstrated that afferent barrage evoked by painful stimuli is intact during and after TENS. Thus, even high frequency TENS stimulation was unable to block input from the peripheral site to the central nervous system. Further, the antihyperalgesic effects of TENS outlast the stimulation time by 8 to 24 hours, sug­ ~DRG gesting mechanisms other than blockade of input from the periphery. A role for adenosine in large fiber stimulation by vibra­ tion analgesia has been suggested by Salter and Henry."7 Because TENS presumably activates large fibers, it fol­ lows that adenosine may playa role. In support of aden­ osine, if human subjects were given caffeine (which d1j~ blocks adenosine receptors) before TENS, the analgesia Injury produced by TENS was significantly reduced compared to placeboso Figure 4. Diagram representing the descending inhibitory Last, release of endogenous opioids has been used to pathways. The PAG sends projections to the RVM, which then explain the actions of TENS, particularly low frequency sends seronergic (5~Hn projections to the spinal cord dorsal stimulation. Recent data support this theory for low fre­ horn, Application of opioids {*} into the PAG, RVM, or spinal cord results in analgesia and reduces pain. DRG, dorsal root quency TENS as well as for high frequency TENS stimula­ ganglion. tlon.37,79 23 There are 3 types o'f opioid receptors, 1-1" &, and K. These are located peripherally, in the spinal cord and in areas involved in descending inhibition including the nu­ several opiolds and their receptors might be involved in cleus raphe magnus in the rostral ventral medulla (RVM) relief of pain by TENS. and the periaqueductal gray (PAG)." The PAG sends pro­ In an animal model of knee joint inflammation, sec­ jection to the RVM, which in turn sends projections to the ondary hyperalgesia is reversed completely by either low spinal dorsal horn" (Fig 4). Stimulation ofthe PAG orthe frequency (4 Hz) or high frequency (100 Hz) TENS at sen­ RVM produces inhibition of dorsal horn neurons includ­ sory intensities." To test the role of opioid receptors in ing spinothalamic tract cells. 23 It is commonly accepted the reduction of hyperalgesia produced by TENS, opioid that opioid mediated inhibition produces its effects receptor antagonists were delivered directly to the spi­ through activation of the PAG-RVM pathway." Further, nal cord to block ~~ (naloxone), 8 (naltrindole), or K (nor­ the RVM pathways use serotonin as a neurotransmit­ BNI) opioid receptors" (Fig 5). Low frequency, sensory ter." Another common inhibitory pathway is from the intensity TENS antihyperalgesia was prevented by the pontine noradrenergic cells groups, A6 (locus caeruleus) blockade of I), opioid receptors with naloxone, and high and A7 (locus subcaeruleus)." These pontine neurons frequency, sensory intensity TENS antihyperalgesia was use the neurotransmitter noradrenaline and activate n~-2 prevented by blockade of5 opioid receptors with naltrin­ receptors spinally to produce inhibition of dorsal horn dole79 (Fig 5), Further studies tested the role of opioid neurons.23 receptors in areas of descending inhibition. Blockade of Concentrations of ~-endorphins increase in the blood­ opioid receptors in the RVM showed a similar effect as stream and cerebrospinal fluid of healthy subjects after observed by spinal blockade." Specifically, blockade of 11 31 administration of either high or lowfrequency TENS. ,•• opioid receptors in the RVM prevented the antihyperal­ Increased concentrations of methionine enkephalin, a B gesia by low frequency, sensory intensity TENS, and opioid agonist, and dynorphin A, a " opioid agonist, are blockade of 8 opioid receptors prevented the antihyper­ observed in the lumbar cerebrospinal fluid after treat­ algesia produced by high frequency, sensory intensity ment of patients with either low or high frequency TENS, TENS" (Fig 5), These data thus suggest that specific and respectively.'9 This suggests that at the spinal level there different opioid receptors are activated by different fre­ are different opioids released with different stimulation quencies of TENS such that fl. opioid receptors are in­ frequencies and thus possibly different opioid receptors volved in the antlhyperalgesia produced by high fre­ activated to produce analgesia with high or low fre­ quencies and 8 oploid receptors are involved in the quency TENS. Taken together, these data indicate that antihyperalgesia produced by low frequency TENS, 114 TENS review transmitter located in the PAG-RVM pathway. Depletion Spinal blockade 150 of serotonin reduces the antinociceptive effect of high frequency stimulation in the intact but not in the spinal­ ized animaL97 Electrical 5timulation induced antinoci­ ~ 100 j------t-,-­ ception is significantly enhanced by administration of ,e ~ L-S-hydroxytryptophan, a serotonin precursor, and abol­ .S 50 ished by the opiate receptor antagonist, naioxone, and n the serotonin receptor antagonist, methysergide ,97 i Thus, TENS inhibition involves activation of descending "- 0 inhibitory pathways involving the RVM and using sero­ tonin and opioids to reduce pain and hyperalgesia. -50 .L--_~----~-_--~-- TENS could potentially have local peripheral or auto­ no TENS Low High nomic effects. Electrical stimulation in intensities that could activate Ao fibers modifies local blood flow and vascular resistance. 6B Transient increases in blood flow with low frequency, burst mode (2 Hz) TENS were ob­ RVM blockade c=::J Saline _ Naloxone served at the area of stimulation if fntensity was 25% 150 lZZ2ZI Naltrindole above the motor threshold but not just below (sensory intensity) or just above motor threshold?'Similarly, high ~ 100 t------+-h-,,------i~- frequency TENS stimulation at intensities just above or

~ below motor threshold did not affect local blood flow.'2 :E E 50 With laser Doppler imaging, increases in blood flow C were observed with either low or high frequency TENS at @ m an intensity that was felt but not painful (10 to 15 mA)."4 "- 0 In rats with nerve injury, low frequency, motor intensity TENS reduced mechanical hyperalgesia and cold allo­

-50 ~---~---~- dynia. The effect of TENS on cold , but not me­ no TENS Low High chanical hyperalgesia, wa5 reduced by systemic phentol­ amine to biock alpha-adrenergic receptors, suggesting Figure 5. Bar graphs represent the percent inhibition of hyper~ activation of sympathetic noradrenergic receptors may algesia following blockade of IJ.. opioid receptors with naloxone mediate TENS effects.54 However, effects of phentol­ or 0 OpiDid receptors with naltrindole in the spinal cord (top amine could block central receptors, and future studies panel) or RVM (bottom panel). Animals received no TENS, low frequency TENS (low), or high frequency TENS (high) at sensory should address this issue. Substance Pin dorsal root gan­ intensities. Secondary hyperalgesia was induced by intra~articu~ glia neurons and spinal cord dorsal horn is reduced by far injection of kaolin and carrageenan into the knee joint. A full high frequency TENS in animals injected with the inflam­ reduction in hyperalgesia is 100% inhibition. and no change in matory irritant, formalin. 64 Thus, evidence is beginning hyperalgesia is 0% inhibition. In animals that did not receive TENS there was no change in the degree of hyperalgesia. Low to emerge that some olthe analgesic effects otTENS may and high frequency TENS with saline injected into either the be mediated through actions on primary afierent fibers spinal cord or RVM resulted in approximately 100% inhibition and modulation of autonomic activity. of hyperalgesia. Blockade of fJ. opioid receptors with naloxone, but not 0 opioid receptors with naltrindole, in either the spinal cord or RVM prevented the antihyperalgesic effects of [ow fre­ What Does This Mean in the Clinic and quenqr TENS, In contrast, blockade of 1) opioid receptors, but not 11, opioid receptors, prevented the antihyperalgesic effect of Suggestions for Future Studies high frequency TENS, Clinically, TENS will more than likely not be the only treatment the patient is receiving. TENS is a complemen­ tary and adjunct treatment to control pain. Medically, Early clinical and basic studies support this view. Sjol­ the patient will more than likely be taking prescription und and Eriksson"used naloxone systemically to block medications such as nonsteroidal anti-inflammatories, opioid receptors in human subjects treated with either opioids (fentanyl, oxycodone hydrochloride), alpha-ad­ high or low frequency TENS. Doses used were at a range renergic agonists (c1onidine), or muscle relaxants (cyclo­ sufficient to block fL opioid receptors. Their data showed benzaprine). The most common procedural interven­ that low frequency, but not high frequency, TENS was tions in physical therapy are therapeutic exercise and blocked by naloxone. This was followed by 2 animal stud­ functional training.' Physical therapists who treat pain, ies, in which higher doses of naloxone could be adminis­ particularly , use a combination of exercise tered to nonselectively block opioid receptors. These and functional training. Electrotherapeutic modalities, studies showed that high frequency TENS was reduced or TENS, are used by physical therapists as an adjunct to by systemic naloxone in doses sufficient to block fL, 0, and modulate and reduce pain, and use of TENS in the ab­ Ie opioid receptors.30,97 sence of other interventions is not considered physical A role for descending inhibitory pathways is further therapy.' However, in some conditions and patients, supported by studies on serotonin, which is the neuro- pain limits the ability of a patient to perform an ade-

----_._------._---_.._- ---- CLINICAL REVIEW/Sluka et al 115 quate exercise program. Once the pain is controlled, the Because low frequency TENS works by activation of Ii patient should be better able to perform an active exer­ opioid receptors, this enhanced antihyperalgesia is prob­ cise program, activities of daily Jiving, or return to work. ably a result of synergistic interaction between alpha Understanding the mechanisms will better assist the cli­ 2-adrenergic receptors and endogenous opioids. Use of nician in the appropriate choice of pain control treat­ TENS in combination with morphine or c10nidine should ment. Parameters of stimulation can be based on the reduce the dosage of morphine or clonidine necessary to basic knowledge, and use ofa particular modality such as reduce hyperalgesia and thus reduce side effects of mor­ electrical stimulation can be used in a more educated phine and increase analgesia. In fact, clinically, intake of manner. Specific examples will be given belowto address opioids is reduced in patients using TENS.2'l·G5.82,8.3,92,93 these issues. Further, there is a reduction in nausea, dizziness, and Use of TENS (in combination with other therapies) will pruritus associated with morphine intake.92 allow patients to increase activity level, reduce hospital Animal studies suggest that TENS would be more ef­ stay, and improve function. Indeed, treatment with TENS fective for or secondary hyperalgesia than increases joint function in patients with arthri­ for primary hyperalgesia. This has yet to be determined 2 ti5. ,1.12,4.'7,48,98 In patients with chronic low , im­ clinically. In clinical studies, one should use a number of provements on the physical and mental component sum­ assessments for measuring the effectiveness of TENS. It mary on the SF-36 quality of life survey occur with seems clear that there are a variety of measurements TENS." Postoperative TENS treatment in patients after that could be assessed in the pain patient and that TENS thoracic surgery reduces recovery room stay and im­ may not work equally well on all of these measures. proves pulmonary function as measured by postopera­ Therefore, understanding the neurotransmitters and tive P02, vital capacity, and functional residual capacity pathways involved in TENS antihyperalgesia could help 3 when compared to sham controls. ,92 Thus, decreasing explain conflicting data with respect to the patient pop­ pain with TENS increases function and allows the patient ulation studies and TENS. Itwill further assist the clinician to tolerate other therapies and activities, resulting in an in the treatment choice for a particular patient. The clin­ improved quality of life. ical use of TENS and further clinical outcome studies One should be aware of the medication a person is should be carefully evaluated with respect to the current taking and the effects of these medications on the ef­ medication olthe patient. fects of TENS. By understanding the mechanisms of ac­ Thus, future clinical studies need to be performed to tion of TENS, more appropriate treatment strategies can confirm these animal data to provide a solid evidence be tried. If a patient is taking opioids, currently those base for the use of TENS. Clearly all patients might not available that activate 1,1, opioid receptors, high fre­ respond to TENS treatment. Some considerations to take quency TENS may be more appropriate. Repeated appli­ into account would be which frequency of TENS to use. cation of opioid produces tolerance to the opioid such Combinations of commonly administered pharmaceuti­ that a higher dose is necessary to produce the same ef­ cal agents and TENS should be addressed in a clinical fect. This is based on the fact that low frequency TENS, population. but not high frequency, is ineffective if given in animals tolerant to morphine.ao Clinically, Solomon et al8.3 dem­ onstrated that in patients who had taken enough opi­ The Clinical Efficacy of TENS oids to become tolerant to morphine, TENS was ineffec­ Many of the early publications on TENS were either tive in reducing postoperative pain. However, anecdotal or case reports and did not involve random­ parameters of stimulation were not given, and this needs ized controlled trials (RCTs). An RCT is a trial in which to be more fully addressed. Furthermore, it follows that patients are randomly allocated to different treatment repeated treatment with the same frequency of TENS regimens, eg, active treatment, placebo, or control.'9 would produce tolerance to its analgesic effects. Indeed, Blinding is a very important factor in the design of an daily treatment with either low frequency or high fre­ RCT. This refers to whether the participants, those ad­ quency TENS in animals with knee joint inflammation ministering the interventions, and those assessing the produces tolerance to TENS and a cross-tolerance to spi­ outcomes are blinded to group assignment.53 A single nally administered Ii or ~ opioid agonists, respectively.13 blind trial is one in which 1 group of individuals involved Thus, TENS is ineffective if morphine tolerance is present in the trial do not know the identity of the intervention and shows opioid tolerance with repeated use. that is given to the participant; this is usually the partic­ It might be possible to enhance the effects of TENS ipants or the investigators assessing the outcomes. A clinically if given in combination with certain agonists or double blind trial is one in which 2 groups of individuals antagonists. High frequency TENS only partially reduces involved in the trial do not know the intervention given primary hyperalgesia, and low frequency TENS is ineffec­ to each participant; usually these 2 groups are the par­ tive on primary hyperalgesia.'s However, either high or ticipants and the investigators who are assessing the out­ lowfrequency TENS is more effective in reducing primary comes.34 Although the number of clinical trials on TENS hyperalgesia if given in combination with acute admin­ has increased considerably during the past several years, istration of morphine76 or clonidine.78 Synergism be­ there is still a need for rigorously conducted RCTs to tween alpha-adrenergic agonists and opioid agonists (IJ. determine its efficacy for acute and chronic pain condi­ and ~) has been shown in pharmacologic studies.,o.21 tions and, indeed, its nonanalgesic effects. The RCT is

~~~~~~~~---_._._--_ .. ------116 TENS review

TABLE 1. Summary of Outcomes of TENS Systematic Reviews

NUMBER OF STUDIES CONDITION AUTHORS INCLUDED OUTCOME

Chronic pain Carroll el ai, 2001 19 Inconclusive Chronic Milne el ai, 2002 5 No evidence to support TEhlS PI"imary dysillenorrhea Proctor et ai, 2001 9 High frequency TENS more effective than placebo; low frequency TENS no more effective than placebo Labor pain Carroll et a!, 1997 10 TENS has no signifiGmt effect Po~;l operative pain Carroll et ai, 1996 17 In 15 of 17 RCTs, TENS had no henefit over pk;cebo Kne(~ o51eoarthl'iti~, Osiri et ai, 2001 7 Conventional TE.NS and acupuncture-like TENS effective over placebo Post-stroke ~,houlder pain Price and Pandyan, 2000 4 Inconclusive rEf'~S, tl'ilmcutaneous electrical nerve stimulation; HCTs, randomized controlled trials, regarded as the gold standard in clinical trials of effica­ ity for chronic pain conditions, and a wide range of sur­ cy70 and should therefore be viewed as the method of veys provide evidence to support this belief.'2,36.87 The choice for evaluating a modality such as TENS. systematic review by Carroll et al" included 19 RCTs on One of the quickest methods of reviewing the clinical TENS and chronic pain that covered a broad range of research on TENS is to read a recent systematic review. conditions including rheumatoid arthritis, myofascial Systematic reviews should ideally provide an objective pain, diabetic neuropathy. and low back pain. Chronic summary of the current literature on the chosen topic pain was defined as pain of at least 3 months' duration. However, some concerns have been raised about the The most common outcome measure used in these stud­ methods involved in determining their outcome; there­ ies was the 10-cm (VAS). The au­ fore, perhaps the best way to regard these reviews is that thors thoughtthat the methodologic quality of the stud­ "they may not be correct all of the time but they give a ies was generally poor and reported that the results of good guide most of the time."16 Systematic reviews in­ their review were inconclusive. Several problems were volve the retrieval of relevant studies that have been highlighted such as inadequate reporting of the results selected according to certain inclusion criteria and using and indeed the TENS treatments; the latter makes any predefined criteria lists such as the Jadad, Delphi, or future replication very difficult. Carroll et al suggested Maastricht to score the quality of the study.17.33.89 The that we need large, randomized, multicenter, controlled following items are used to rate the study's method­ trials in chronic pain. ologic quality: randomization. blinding, withdrawals, analysis and bias. However, the reader should be aware Chronic Low Back Pain that different criteria lists do not always provide similar Only five RCTs were eligible for inclusion in the system­ results when applied to the same trials. Verhagen et al 88 atic review published by Milne et al'2 on TENS and compared the outcome of 3 criteria lists on a data set of chronic low back pain. Chronic low back pain was de­ 21 studies and highlighted several differences between fined as low back pain over 3 months' duration. Interest­ them that affected their respective ranking of the stud­ ingly, the application of TENS in the studies varied ies. With this warning in mind, Table 1 provides a sum­ greatly, ranging from 1 treatment per day for 2 consec­ mary of the key systematic reviews on the effectiveness utive days to 3 treatments per day for 4 weeks. This vari­ of TENS published during the past several years, each of ation in the actual method of applying TENS is a very which will be discussed in more detail below. good example of the lack of standardization generally observed across the RCTs published on TENS. Outcome Chronic Pain measures included assessment of pain, function, well­ Berman and Bausell" conducted a survey on the use of being, disability, and satisfaction of care. The reviewers nonpharmacologic therapies by pain specialists in 2000; concluded that there was no evidence to support the use they surveyed a sample of members olthe International of TENS for the management of chronic low back pain Association for the Study of Pain and reported that 77% but that there was a lack of data on type of application, of respondents indicated that they used TENS/other elec­ treatment duration, and optimal frequencies and inten­ tromagnetic applications. The cost-effectiveness of TENS sities. One study included in the review determined the as a technique has been highlighted best electrode placement site for each individual patient by Chabal et al,12 who interviewed a sample of 376 pa­ before the trial commenced49 This is a technique that is tients with chronic pain who were long-term users of used in clinical practice and should also be used more TENS. After applying a cost simulation model to their frequently in clinical trials rather than selecting fixed data, they concluded that costs could be reduced up to points for all patients. Although this study did not score SS% for pain medication and up to 69% for physical well overall in terms of the Jadad criteria list used, the therapy/occupational therapy treatments. TENS application technique was commendable. Similar TENS is viewed by many clinicians as primarily a modal- findings were shown in a meta-analysis by Brosseau et CLINICAL REVIEW/Sluka et al 117 aI.' With the same S RCTs as Milne et al 52 they found no short time period (eg, 30 minutes), but a few used much difference between active TENS and sham TENS. They longer periods (8 hours). This factor may therefore affect concluded that there was no evidence to support the use subsequent evaluation of the treatment. or nonuse of TENS for the treatment of chronic low back pain and future studies should include standardized out­ Knee Osteoarthritis come measures. Seven RCTs were eligible for inclusion in the systematic review by Osiri et al 57 on osteoarthritis and TENS. Both Post-Stroke Shoulder Pain conventional and acupuncture-like modes of TENS were Price and Pandyan S9 published a systematic review on used in these studies. The length of treatment varied the effect of electrical stimulation for post-stroke shoul­ from one single 3D-minute application to several appli­ der pain. They included various types ofsurface electrical cations daily for up to 6 weeks. Assessments of pain, stimulation including functional electrical stimulation stiffness, joint circumference, and muscle strength were and TENS; the stimulation parameters were different; used as outcome measures. The authors concluded that therefore they were not comparing "like with like." both conventional and acupuncture-like TENS were Measurements of pail\ range of passive humeral lateral more effective than placebo for relief of pain but that rotation, motor score, and spasticity score were the out­ the studies were heterogenous with different study de­ come measures used in the 4 included studies. The au­ signs and outcomes used. The reviewers called for 5tan~ thors expressed disappointment that so many of the dardized treatment protocols to be adopted for further published studies were case reports or else they used TENS studies that would include electrode placement, nonstandard outcome measures. They concluded that treatment time, and parameters. However, the point they were unable to make any definite conclusion about raised with the low back pain studies applies to this con­ the 4 studies included in this review and emphasized the dition also, ie, electrode placement should ideally be de­ need for adequately powered RCTs to examine the role termined for each individual placement as performed in of electrical stimulation for this application. clinical practice. A recent article by Cheing et ai, 15 which was not included in the systematic review by Osiri et a!, Primary Dysmenorrhea has investigated the effect of TENS or isometric exercise on osteoarthritic knee pain. TENS was applied for 60 min­ Primary dysmenorrhea is the occurrence of painful utes S days per week for 4 weeks. The results showed a menstrual cramps that are associated with ischemia of significant cumulative reduction in VAS in the TENS the uterus. The ischemia is believed to be linked to the group and placebo group. The authors also reported presence of prostaglandins in the menstrual fluid that in that the decrease in pain was maintained at the 4-week turn cause hypercontractility of the myometrium. In the follow-up only in the TENS and TENS plus exercise treatment of this condition, the TENS electrodes are typ­ groups. Previous work by this research group has also ically placed over the abdomen or thoracic spine in areas reported a cumulative beneficial effect of daily applica­ related to the spinal nerve roots that receive nociceptive tion of TENS for both experimental pain and chronic low information from the uterus. In some cases, acupuncture 14 58 back pain. . points have been used, eg, 821, 829, ST36, and SP6.55 Nine RCTs were selected forthe systematic review carried out by Proctor et a1,',0 which examined the effect ofTENS Acute Postoperative Pain and acupuncture on primary dysmenorrhea. Four of the Postoperative pain is an example of an acute pain con­ studies used a crossover design that involved the subjects dition in which TENS has been used with some success. In receiving all of the treatments during different men­ the postoperative situation, TENS is typically used as an strual cycles. The outcome measures typically included adjunct to routine medication rather than as an isolated pain measurement and a record of menstrual symptoms. treatment. Application involves positioning sterile elec­ The findings of this review were that high frequency trodes parallel to the incision with additional electrodes TENS was more effective than placebo and low fre­ sometimes placed over the corresponding thoracic spinal quency TENS was no more effective than placebo. In ad­ nerves. The obvious advantages of controlling pain post­ dition, there was insufficient evidence to compare high operatively include earlier mobilization, more effective frequency with low frequency TENS. The reviewers que­ deep breathing/coughing, which will lead to earlier dis­ ried whether the different approach to electrode place­ charge. Seventeen studies were included in the system­ ment in the studies contributed to treatment outcome, atic review by Carroll et al. '1 Analgesic consumption and ie, the use of specific acupuncture points versus placing VAS were the 2 most common methods used to evaluate the electrodes over the site of pain. They also raised an­ the treatments. This review is an important one because other interesting point with regard to the physiologic it clearly highlights the effect of lack of randomization in effects of TENS. Low frequency TENS typically produces clinical trials. Of the 17 randomized studies that were muscle contractions, which may make it difficult for the included, lS reported no beneficial effect of TENS, user to wear TENS as a portable unit and to carry out whereas 17 of the 19 nonrandomized trials that were daily activities, an issue that is not associated with high excluded from the review showed positive effects for frequency TENS, which produces a comfortable pares­ TENS. Schulz et al 70 have indicated that inappropriate thesia. Most of the studies involved applying TENS for a blinding and lack of randomization can overestimate 118 TENS review treatment effects by 17% and 40%, respectively. This trois and active TENS treatments for the primary out­ crucial finding should be borne in mind during the inter­ come measures used, leading the authors to suggest that pretation of published trials on TENS. Carroll et al re­ TENS has no significant effect on iabor pain. This nega­ ported that only 2 of the studies gave details of the ran­ tive finding is in contrast with the high levei of consumer domization method, and the methods described were satisfaction associated with the use of TENS in labor re­ inadequate in both. They concluded that TENS was not ported in several studies including Kaplan et al 39and van effective for postoperative pain based on the 17 studies der Spank et al.86 that they reviewed. The authors of the systematic reviews described above have used the gold standard of clinical research, the RCT, Labor Pain as the inclusion criterion for their individual reviews. The application of TENS for labor pain involves posi­ However! it is obvious that many of the trials reviewed tioning 2 pairs of electrodes over the spinal nerve roots had considerable methodologic problems that led to a of nO-L1 and 52-54. The nociceptive information from low score on the criteria list used. These problems in­ the uterus, perineal structures, and cervix enters the spi­ cluded insufficient reporting of treatment techniques, nal cord at these levels. Both continuous and burst TENS inadequate treatment time, lack of standardized out­ are used; continuous high frequency pulses are delivered come measures, and inadequate blinding and random­ during contractions and low frequency bursts are used ization techniques. The Consolidated Standards of Re­ between contractions. A "boost" control is used to porting Trials (CONSORT) statement is a very usefui switch between continuous and burst outputs. Carroll et al'" included 10 RCTs in their systematic review. There checklist and diagram that was developed by an interna­ was no consistency in the pain outcome measures used, tional group of clinical trialists, statisticians, epidemiolo­ which varied from a VAS, 3 or 4 point , and gists, and biomedical editors. The aim of this statement 53 requirement for other analgesic interventions. Among was to help authors to improve reporting of RCTs. The the methodologic problems highlighted by the review­ CONSORT should ideally be considered in the planning ers was the fact that 4 of the studies included in the stage of future RCTs on TENS because it would provide review did not use any form of blinding. In addition, of researchers with a valuable tool for improving not just the 7 studies that used placebo TENS, only 1 described the quality of reporting but also the methodologic qual­ the blinding procedure in sufficient detail to indicate ity oHhe trial. It is only through high quality, adequately that it may have been adequate. None of the 10 studies powered RCTs that the true clinical efficacy of TENS can reported any significant differences between the con- be established.

References 9. Carroll D, Moore RA, Mcquay HJ, Fairman F, Tramer M, Leijon G: Transcutaneous electrical nerve stimulation (TENS) 1. Guide to physical therapy practice, second edition. Phys for chronic pain. Chochrane Database Syst Rev, 2001 Ther 81:9-744,2001 10. Carroll D, Moore RA, Tramer MR, McQuay HJ: Transcuta­ 2. Abelson K, Langley GB, Vlieg M, Wigley RD: Transcuta­ neous electrical nerve stimulation does not relieve labor neous electrical nerve stimulation in rheumatoid arthritis. N pain: updated systematic review. Contemporary Issues in Z Med J 96:156-158, 1983 Obstetrics and Gynecology September:195-205, 1997

3. Ali J, Yaffe C5, 5essle BJ: The effect of transcutaneous 11. Carroll D, Tramer M, McQuay HJ, Nye 8, Moore A: Ran­ electric nerve stimulation on postoperative pain and pulmo­ domisation is important in studies with pain outcomes: Sys­ nary function. Surgery 89:507-512, 1981 tematic review of transcutaneous electrical nerve stimula­ tion in acute postoperative pain. Br J Anesth 77:798-803, 4. Electrotherapy Standards Committee: Electrotherapeu­ 1996 tic terminology in physical therapy (report). Section on clin­ ical electrophysiology and American Physical Therapy Asso~ 12. Chabal C, Fishbain DA. Weaver M, Heine LW: Long-term ciation, Alexandria, VA, 2001 transcutaneous electrical nerve stimulation (TENS) use: Im~ 5. Bennett GJ, Xie YK: A peripheralmononeuropathy in rat pact on medication utilization and physical therapy costs. that produces disorders of pain sensation like those seen in Clin J Pain 14:66-73, 1998 man. Pain 33:87-107,1988 13. Chandran P, Sluka KA: Development of opioid tolerance 6. Berman 8M, Bausell RB: The use of non~pharmacological with repeated TENS administration. Pain 102:195-201, 2003 therapies by pain specialists. Pain 85:313-315, 2000 14. Cheing G, Hui-Chan CW: Repeated applications of trans­ 7. Brosseau L. Milne S, Robinson V, Marchand S, Shea B, cutaneous electrical nerve stimulation (TENS) produce cu­ Wells G, Tugwell P: The effectiveness of the transcutaneous mulative effects on chronic clinical pain but not acute exper­ electrical nerve stimulation (TENS) for the treatment of imental pain in chronic low back pain patients. Abstracts of chronic low back pain: A meta-analysis. Spine 27:596-603, the 8th World Congress of the International Association for 2002 the Study of Pain. IASP Press, Seattle, WA, 1996, P 85 8. Campbell IN, Taub A: Local analgesia from percutaneous 15. Cheing GLY, Hui-Chan CWY, Chan KM: Does four weeks electrical stimulation. Arch Neurol 28:347-350, 1973 of TENS and/or isometric exercise produce cumulative reduc~ CLINIC!'.!. IlEVI[W/5Iuka ef al 119 tion of osteoarthritic knee pain. Clin Rehabil 16:749-760, transcutaneous electrical nerve stimulation on limb blood 2002 flow in healthy humans. Phys Ther 74:361-367, 1994

16. Crombie IK, McQuay HJ: The systematic review: A good 33. Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, guide rather than a guarantee. Pain 76:1 w 2, 1998 Gavaghan DJ, McQuay HJ: Assessing the quality of reports of randomized clinical trials: Is blinding necessary? Control (lin 17. de Vet H, de Bie RA, Van der Heijden GJMG, Verhagen Trials 17:1-12,1996 AP, Sijpkes P, Knipschild PG: Systematic reviews on the basis of methodological criteria. Physiother 83:49-58, 1997 34. Jadad A: Randomised Controlled Trials, a User's Guide. [ondon, UK, BMJ Books, 1998 18. Dubner R, Ren K: Assessing transient and persistent pain in cmimals, in Wall PO, Melz8ck R (eds): Textbook of Pain, 35. Janko M, Trontelj JV: Transcutaneous electrical nerve chap 14. New York, NY, 1999, pp 359-369 stimulation a microneurographic and perceptual study. Pain 9:219-230, 1980 19. Earl-Slater A: The Handbook of Clinical Trials and Other Research. Radcliffe Medical Press, axon, UK, 2002 36. Johnson Ml, Ashton CH, Thompson JW: An in-depth study of longwterm users of transcutaneous electrical nerve 20. Fairbanks CA, Nguyen HO, Grocholski BM, Wilcox GL: stimulation (TENS): Implications for clinical use otTENS. Pain Moxonidine, a selective imidazoline w alpha2 -adrenergic re­ 44:221-229,1991 ceptor agonist, produces spinal synergistic antihyperalgesia with morphine in nerve-injured mice. Anesthesiology 93: 37. Kalra A. Urban MO, Sluka KA: Blockade of opioid recep­ 765-773, 2000 tors in rostral ventral medulla prevents antihyperalgesia produced by transcutaneous electrical nerve stimulation 21. Fairbanks CA, Posthumus IJ, Kitto KF, Stone [S, Wilcox (TENS). J Pharmacal Exp Ther 298:257-263, 2001 GL: Moxonidine, a selective imidazoline/alpha(2} adrenergic receptor agonist, synergizes with morphine and deltorphin 38. Kane K, Taub A: A history of local electrical analgesia. II to inhibit ~induced behavior in mice. Pain 84: Pa in 1: 125-138, 1975 13-20,2000 39. Kaplan B, Rabinerson D, Lurie 5, Bar J, Krieser UR, Neri A: 22. Feldman RS, Meyer JS, Quenzer LF: Principles of Neuro­ Transcutaneous electrical nerve stimulation (TENS) for adju­ psychopharmacology. Sunderland, MA, Sinauer Associates vant pain-relief during labor and delivery. Int J Gynaecol Inc, Publishers, 1997 Obstet 60:251-255,1998

23. Fields HL, Basbaum AI: Central nervous system mecha~ 40. Kim SH, Chung JM: An experimental-model for periph­ nisms of pain modulation, in Wall PO, Melzack R (eds): Text­ eral neuropathy produced by segmental spina! nerve Hga~ book of Pain, chap 12. New York, NY, 1999, pp 243-257 tion in the rat. Pain 50:355-363, 1992 24. Garrison OW, Foreman RD: Decreased actiVity of sponta~ 41. King EW, Sluka KA: The effect of varying frequency and neous and noxiously evoked dorsal horn cells during trans­ intensity of transcutaneous electrical nerve stimulation on cutaneous electrical nerve stimulation (TENS). Pain 58:309­ secondary mechanical hyperalgesia in an animal model of 315,1994 inflammation. J Pain 2:128-133, 2001 stimula~ 25. Garrison DW, Foreman RD: Effects of prolonged transcu­ 42. Kumar VN, Redford JB: Transcutaneous nerve taneous electrical nerve stimulation (TENS) and variation of tion in rheumatoid arthritis. Arch Phys Med Rehabil 63:595­ stimulation variables on dorsal horn cell activity. Eur J Phys 596, 1982 Med Rehabil 6:87-94. 1997 43. Lamotte RH, Shain CN, Simone DA, Tsai EFP: Neurogenic 26. Garry MG, Hargreaves KM: Enhanced release of immu­ hyperalgesia: Psychophysical studies of underlying mecha­ nisms. J Neurophysiol 66:190-211, 1991 noreactive CGRP and substance-P from spinal dorsal horn slices occurs during carrageenan inflammation. Brain Res 44. Lee KH, Chung JM, Willis WD: Inhibition of primate spi­ 582: 139-142, 1992 nothalamictract cells byTENS. J Neurosurg 62:276-287, 1985 27. Ghoname EA, Craig WF, White PF, Ahmed HE, Hamza 45. Leem JW, Park ES, Paik KS: Electrophysiological evidence MA, Gajraj NM, Vakharia AS, Nohr D: The effect of stimulus for the antinociceptive effect of transcutaneous electrical frequency on the analgesic response to percutaneous elec~ nerve stimulation on mechanically evoked responsiveness of trical nerve stimulation in patients with chronic low back dorsal horn neurons in neuropathic rats. Neurosci Lett 192: pain. Anesth Analg 88:841-846,1999 197-200,1995 28. Gopalkrishnan P, Sluka KA: Effect of varying frequency, 46. Ma YT, Sluka KA: Reduction in inflammation-induced intensity and pulse duration otTENS on primary hyperalge­ sensitization of dorsal horn neurons by transcutaneous elec­ sia in inflamed rats. Arch Phys Med Rehabil81 :984-990,2000 trical nerve stimulation in anesthetized rats. Exp Brain Res 137:94-102,2001 29. Han JS, Chen XH, Sun 5L, Xu XJ, Yuan Y, Yan SC, Hao JX, Terenius .L: Effect of low and high frequency TENS on met­ 47. Mannheimer C, Carlsson C-A: The analgesic effect of enkephalin-arg-phe and dynorphin A immunoreactivity in transcutaneous electrical nerve stimulation (TNS) in patients human lumbar CSF. Pain 47:295-298,1991 with rheumatoid arthritis: A comparative study of different pulse patterns. Pain 6:329-334,1979 30. Han J5, Chen XH, Sun SL, Xu XJ, Yuan Y, Yan SC, Hao JX, Terenius L; Effect of low and high frequency TENS 011 met~ 48. Mannheimer C, Lund S, Carlsson C-A: The effect of trans­ enkephalin-arg-phe and dynorphine A immunoreactivity in cutaneous electrical nerve stimulation (TNS) on joint pain in human lumbar CSF. Pain 47:295-298, 1991 patients with rheumatoid arthritis. Scand J Rheumatol 7:13~ 16, 1978 31. Hughes GS, Lichstein PR, Whitlock D, Harker C: Response of plasma beta~endorphins to transcutaneous electrical 49. Marchand S, Charest J, Li J, Chernard JR, Lavignolle B, nerve stimulation in healthy subjects. Phys Ther 64:1062­ Laurencelie L: Is TENS purely a placebo effect? A controlled 1066, 1984 study on chronic low back pain. Pain 54:99-106,1993 32. Indergand HJ, Morgan BJ: Effects of high frequency 50. Marchand S, Li J, Charest J: Effects of caffeine on anal-

.. _.._------_...-.~----_.._----- 120 TENS review gesia from transcutaneous electrical nerve stimulation. 68. 5ato A, Sato A, Schmidt RF: Impact of Somatosensory N Engl J Med 333:325-326, 1995 Input on Autonomic Functions, Heidelberg, Germany, Springer, 1997 51. Melzack R, Wall PD: Pain mechanisms: A new theory. Science 150:971-978, 1965 69. Schaible HG, Grubb SD: Afferent and spinal mechanisms of joint pain. Pain 55:5-54, 1993 52. Milne S, Welch V, Brosseau L, Saglnur M, Shea B, Tugwell P, Wells G: Transcutaneous electrical nerve stimulation 70. Schulz KF, Chalmers I, Hayes RJ, Altman DG: Empirical (TENS) for chronic low back pain. Chochrane Data 8ase Syst evidence o'f bias: Dimensions of methodological quality as~ Rev, 2001 sodated with estimates of treatment effects in controlled trials. JAMA 273:408-412, 1995 53. Moher D, Schulz KF, Altman DG: The CONSORT state­ ment: Revised recommendations for improving the quality 71. Sherry JE, Oehrlein KM, Hegge KS, Morgan BJ: Effect of of reports of parallel-group randomized trials. Ann Intern burst~mode transcutaneous electrical nerve stimulation on Med 134:657-662, 2001 peripheral vascular resistence. Phys Ther 81: 1183-1t91, 2001 54. Nom T5, Choi Y, Yeon D5, Leem JW, Paik KS: Differential 72. Shimizu T, Koja T, Fujisaki T, Fukuda T: Effects of methy­ antinociceptive eHect of transcutaneous electrical stimula­ sergide and naloxone on analgesia induced by the periph~ tion on pain behavior sensitive or insensitive to phentol~ erat electric stimulation in mice. Brain Res 208:463-467, 1981 amine in neuropathic rats. Neurosci Lett 301:17-20,2001 73, Sjolund BH: Peripheral nerve stimulation suppression of 55. Neighbors LE, Clelland J, Jackson JR, 8ergman J, Orr J: (-fiber evoked flexion reflex in rats. Part 1: Parameters of Transcutaneous electrical nerve stimulation for pain relief in continuous stimulation. J Neurosurg 63:612-616,1985 primary dysmenorrhea. Clin J Pain 3:17-22, 1987 74. Sjo!und BH: Peripheral nerve stimulation suppression of 56. Nolan MF: Selected problems in the use of transcutane­ C~fiber evoked flexion reflex in rats. Part 2. Parameters of ous electrical nerve stimulation for pain control: An ap­ low rat train stimulation of skin and muscle afferent nerves, praisal with proposed solutions, A special communication. J Neurosurg 68:279-283, 1988 Phys Ther 68: 1694-1698, 1988 75. SjoJund BH, Eriksson MBE: The influence of naloxone on 57. Osiri M, Welch V, Brosseau L, Shea B, McGowan J, TugM analgesia produced by peripheral conditioning stimulation, well P, Wells G: Transcutaneous electrical nerve stimulation Srain Res 173:295-301, 1979 for knee osteoarthritis. Chochrane Data Base Syst Rev, 2001 76, Sluka KA: Systemic morphine in combination with TENS 58. Parpura V, Basarsky TA, Liu F, Jeftinija K, Jeftinija S, produces an increased analgesia in rats with acute inflam~ Haydon PG; GlutamateMmediated astrocyte-neuron signal­ mation. J Pain 1:204-211,2000 ling. Nature 369:744-747, 1994 77. Sluka KA, Bailey K, Bogush J, Olson R, Ricketts A: Treat­ 59. Price 0, Pandyan AD: Electrical stimulation for prevent­ ment with either high or low frequency TENS reduces the ing and treating post-stroke shoulder pain, Chochrane Da w secondary hyperalgesia observed after injection of kaolin tabase Syst Rev, 2000 and carrageenan into the knee Joint, Pain 77:97M102, 1998

60. Proctor ML, Smith CA, Farquhar CM, Stones RW: Trans­ 78. Sluka KA, Chandran P: Systemic administration of cutaneous electrical nerve stimulation and acupuncture for clonidine in combination wtih TENS produces an increased primary dysmenorrhoea. Chochrane Database Syst Rev, antihyperalgesia in rats. Pain 100:65-76, 2002 2002 79. Sluka KA, Deacon M, Stibal A, Strissel S, Terpstra A: Spinal 61. Ren K, Williams GM, Hylden JLK, Ruda MA, Dubner R: blockade of opioid receptors prevents the analgesia pro­ The intrathecal administration of excitatory amino-acid reM duced by TENS in arthritic rats. J Pharmacol Exp Ther 289: ceptar antagonists selectively attenuated carrageenan-in­ 840-846, 1999 duced behavioral hyperalgesia in rats. Eur J Pharmacol 219: 235-243, 1992 80. Sluka KA, Judge MA, McColley MM, Reveiz PM, Taylor BM: Low frequency TENS is less effective than high "fre­ 62, Robinson AJ: Transcutaneous electrical nerve stimula­ quency TENS at reducing inflammation induced hyperalge­ tion for the control of pain in musculoskeletal disorders, sia in morphine tolerant rats. Eur J Pain 4:185-193,2000 J Orthop Sports Phys Ther 24:208-227, 1996 81. Sluka KA, Westlund KN: Behavioral and immunohistoM 63. Robinson AJ, 5nyder-Mackier L: Clinical Electrophysiol­ chemical changes in an experimental arthritis model in rats. ogy: Electrotherapy and Electrophysiological Testing (2nd Pain 55:367-377, 1993 edition), Baltimore, MD, Williams and Wilkins, 1995 82. Smith CR, Lewith GT, Machin D: TNS and osteoarthritis: 64. Rokugo T, Takeuchi T, Ito H: A histochemical study of Preliminary study to establish a controlled method of assessM substance P in the rat spinal cord: Effect of transcutaneous ing transcutaneous electrical nerve stimulation as a treat­ electrical nerve stimulation. J Nippon Med Sch 69:428-433, ment for pain caused by osteoarthritis, Physiother 69:266­ 2002 268, 1983 65. Rosenberg M, Curtis L, Bourke DL: Transcutaneous elec­ 83. Solomon RA, Viernstein MC, Long DM: Reduction of trical nerve stimulation for the relief of postoperative pain, postoperative pain and narcotic use by transcutaneous elec­ Pain 5:129-133,1978 trical nerve stimulation. Surgery 87:142-146,1980 66. Salar G, Job I, Mingrino S, Sosio A, Trabucchi M: Effect of 84. Somers DL, Clemente FR: High-frequency transcutaneous transcutaneous electrotherapy on (SF ~Mendorphin content electrical nerve stimulation alters thermal but not mechan­ in patients without pain problems. Pain 10:169-172, 1981 ical al10dynia following chronic constriction injury of the rat sciatic nerve. Arch Phys Med RehabiI79:1370-1376, 1998 67. Salter MW, Henry JL: Evidence that adenosine mediates the depression of spinal dorsal horn neurons induced by 85. Spielholz NI, Cameron M, Robinson AJ, Snyder-Mackler L, peripheral vibration in the cat, Neuroscience 22:631 M 650, Underwood F, Wicken A: A review of research articles relat­ 1987 ing to the efficacy of electrical stimulation to alleviate pain

.~~----~-----~--- CLINICAL I1EVIEW/Sluka et al 121

and strengthen muscle: Are insurance companies justified in 92. Wang B, Tang J, White PF, Naruse R, Sioninsky A, Kariger denying reimbursement for these purposes? Section on Clin~ R, Gold J, Wender RH: Effect ofthe intensity of transcutane­ ical Electrophysiology of the American Physical Therapy As­ ous acupoint electrical stimulation on the postoperative an­ sociation, 2001. Available at: www.aptasce.com. Accessed algesic requirement. Anesth Analg 85:406-413,1997 September 12, 2002 93. Warfield CA, Skein JM, Frank HA: Tile effect of transcu­ 86. van der Spank n, Cambler DC, De Paepe HM, Danneels taneous electrical nerve stimulation on pain after thoracot­ LA, Witvrouw EE, Beerens L: Pain relief in labour by transcu­ omy. Ann Thorae Surg 39:462-465, 1985 taneous electrical nerve stimulation (TENS). Arch Gyneco[ Obstet 264: 131-136, 2000 94. Wikstrom SO, Svedman P, Svensson H, Tanweer AS: Ef­ 87. Verdouw BC, Zuurmond WWA, De Lange JJ, Metz GCH, fect of transcutaneous nerVe stimulation on microcircula­ Wagernans MFM: Long~term USe and effectiveness of trans~ tion in intact skin and blister wounds in healthy volunteers. cutaneous electrical nerve stimulation in treatment of Scand J Plast Reco"str Surg Hand Surg 33:195-201, 1999 chronic pain patients. The Pain Clinic 8:341-346,1995 95. Willis WD, Coggeshall RE: Sensory Mechanisms of the 88. Verhagen AP, de Bie RA, Lenssen AF, de Vet HCW, Kessels Spinal Cord (2nd edition) . New York, NY, Plenum, 1991 AGH, Boers M, van den Brandt PA: Quality assessment of trials: A comparison of three criteria lists. Phys Ther Rev 96. Woolf 0, Barrett GD, Mitchell D, Myers RA: Naloxone­ 5:49-58, 2000 reversible peripheral in intact and spinal rats. Eur J PharmacoI45:311-314, 1977 89. Verhagen AP, de Vet HC, de Bie RA, Kessels AG, Boers M, Bouter LM, Knipschild PG: The Delphi list: A criteria list for 97. Woolf CJ, Mitchell D, Barrett GD: Antinociceptive effect quality assessment of randomized clinical trials for conduct­ of peripheral segmental electrical stimulation in the rat. ing systematic reviews developed by Delphi consensus. J(!in Pain 8:237-252, 1980 EpidemioI51:1235-1241,1998 90. Wall PD, 5weet WHo Temporary abolition of pain in man. 98. Zizic TM, Hoffman KC, Holt PA, Hungerford DS, Odell JR, Science 155:108-109, 1967 Jacobs MA, Lewis CG, Deal CL, Caldwell JR, Cholewcznski JG, Free SM: The treatment of osteoarthritis of the knee with 91. Walsh D: TENS: Clinical Applications and Related Theory. pulsed electrical stimulation. J Rheumatol 22:1757-1761, Edinburgh, UK, Churchill Livingstone, 1996 1995