4,736
Views
26
CrossRef citations to date
0
Altmetric
Review

Capsaicin 8% dermal patch in clinical practice: an expert opinion

, , , , , , ORCID Icon, & show all
Pages 1377-1387 | Received 16 Jan 2020, Accepted 20 Apr 2020, Published online: 08 Jun 2020

ABSTRACT

Introduction

Neuropathic pain (NP) is caused by a lesion or disease of the somatosensory system, which can severely impact patients’ quality of life. The current-approved treatments for NP comprise of both centrally acting agents and topical drugs, including capsaicin 8% dermal patches, which is approved for the treatment of peripheral NP.

Areas covered

The authors summarize literature data regarding capsaicin use in patients who suffer from NP and discuss the clinical applications of this topical approach.

Expert opinion

Overall, the capsaicin 8% dermal patch is as effective in reducing pain intensity as other centrally active agents (i.e. pregabalin). Some studies have also reported fewer systemic side effects, a faster onset of action and superior treatment satisfaction compared with systemic agents. In our opinion, capsaicin 8% dermal patches also present additional advantages, such as a good systemic tolerability, the scarcity of adverse events, the possibility to combine it with other agents, and a good cost-effective profile. It is important to note that, as the mechanism of action of capsaicin 8% is the ‘defunctionalization’ of small afferent fibers through interaction with TRPV1 receptors, the peripheral expression of this receptor on nociceptor fibers, is crucial to predict patient’s response to treatment.

1. Introduction

Neuropathic pain (NP) is caused by a lesion or disease of the somatosensory system [Citation1]. Depending on the site of the lesion or the underlying disease, this type of pain can be classified as either central or peripheral [Citation1,Citation2]. The most relevant causes of peripheral NP (PNP) are painful diabetic peripheral neuropathy (PDPN), postherpetic neuralgia (PHN), HIV-associated neuropathy (HIV-AN) and chemotherapy-induced peripheral neuropathy, as well as trauma or surgical procedures [Citation2]. In addition to pain, patients with PNP experience several symptoms that include, burning, tingling, numbness, allodynia and hyperalgesia, which collectively have a severe impact on quality of life [Citation3]. NP is often poorly responsive to available treatments [Citation2].

Transient receptor potential vanilloid-1 (TRPV1) acts as a thermal nociceptor; however, it plays an important role in detecting a number of painful stimuli, which include heat, acids and irritant chemicals [Citation1]. Capsaicin, which is the main active ingredient in hot chili peppers, is a potent and highly selective TRPV1 agonist [Citation1,Citation4,Citation5]. This molecule activates TRPV1-expressing nociceptors on the skin, causing the onset of pain and erythema [Citation6]. Following this action, topical capsaicin attenuates cutaneous hypersensitivity and reduces pain through a process usually described as ‘defunctionalization’ of nociceptor fibers [Citation5]. Defunctionalization is the cellular consequence of calcium influx triggered by capsaicin-activated TRPV1. High levels of intracellular calcium, its associated enzymatic, cytoskeletal and osmotic changes, and the disruption of mitochondrial respiration all lead to an impaired local nociceptor function for extended periods. This explains why the effects of capsaicin last well beyond its application and TRPV1 stimulation.

Capsaicin creams of low concentrations (0.025–0.075%) have shown moderate efficacy in the topical treatment of PNP [Citation1]. Nevertheless, these creams require several applications per day, and the initial burning sensation is often poorly tolerated. Qutenza®, which is a high-concentration (8%) capsaicin dermal patch, has been developed with the aim of providing long-lasting pain relief resulting from a single application. The capsaicin 8% dermal patch is approved in the EU, either alone or in combination with other medicinal products (for pain), for the treatment of PNP in adults [Citation6]. Overall, patients using the capsaicin dermal patch reported limited adverse events. The general incidence of serious adverse events that resulted from controlled trials was 6% compared to 4% with the control patch [Citation7]. The most common adverse reactions were application site reactions, such as dryness, erythema, edema, pain, papules and pruritus. Nevertheless, those reactions were mild to moderate in severity, resolved spontaneously within 7 days and did not preclude the completion of the treatment (99% of the patients completed ≥90% of the treatment) [Citation8].

Possible disadvantages of the use of capsaicin dermal patch are the fact that its application occurs under the supervision of a healthcare professional and that the treatment may be repeated after 3 months, in case of reappearance of the pain [Citation3,Citation4]. Despite the discomfort, the treatment is well tolerated by the patients.

However, real-life experiences, including well-grounded Expert Opinions, appear necessary for a further elucidation of the role of the high-concentration capsaicin dermal patch in clinical practice, including proper selection of patients most suitable to this therapy [Citation1]. This article reviews available evidence on the pharmacological treatment of PNP, focusing on the use of the capsaicin 8% dermal patch: it presents clinical data on this drug, and discusses the role of high-dose capsaicin in clinical practice, according to the experience of a group of Experts.

2. Localized neuropathic pain: basic principles and treatment options

2.1. Molecular basis and pathophysiology

Localized NP is defined as ‘a type of peripheral neuropathic pain characterized by a circumscribed and consistent area of maximum pain’ [Citation9Citation13]. All types of fibers can be damaged in this condition, but Aδ fibers and, particularly, C fibers have the greatest relevance [Citation14,Citation15]. Indeed, since multiple Aδ and C fiber populations converge onto different spinal neuronal populations, addressing pain mechanisms at the level of peripheral C fibers can result in high levels of analgesic control by attenuation of the drivers of central changes.

Voltage-gated sodium channels (NaV channels) are of paramount importance for nociception, since they generate and propagate action potentials [Citation16,Citation17]. Moreover, HCN channels have recently been associated with inflammatory, neuropathic and postoperative pain [Citation18]. According to the above, targeting peripheral channels, and in particular sodium channels, prevents painful stimuli from reaching the CNS, although individual response may vary [Citation19,Citation20]. Sodium channels may not therefore be the only target to address.

For instance, the thermo-transient receptor potentials (TRPs), which provide information about thermal changes in the environment, are expressed in small primary sensory nerve terminals [Citation21,Citation22]. To date, six thermo-TRPs have been characterized: TRPV1, TRPV2, TRPV3, TRPV4, TRP melastatin 8 and TRP ankyrin (TRPA) 1. TRPV1 and TRPV2 are activated by painful levels of heat; TRPV3 and TRPV4 respond to non-painful warmth; TRP melastatin 8 is activated by non-painful cold temperatures; and TRP ankyrin (TRPA) 1 reacts to painful cold. The different thermal thresholds are controlled by extracellular mediators, which are released by tissue damage or inflammation (e.g., bradykinin, prostaglandins and growth factors).

Blockers of TRPs have been extensively researched, since they may be used as novel analgesics, but they are associated with unacceptable side effects. On the other hand, targeting inflammatory mediators to control the modulation of thermo-TRPs may be a different strategy to develop novel analgesics.

Central sensitization does have an essential role in the process of pain – that is, the exaggerated pain felt after nerve injury and inflammation that can be induced in healthy volunteers by acute intense peripheral stimulation and is observed in many pain conditions [Citation23,Citation24]. Central sensitization is also associated with windup and long-term potentiation and can be recruited by activation of TRPV1 using capsaicin [Citation25].

2.2. Treatment

International guidelines and recommendations from the European Federation of Neurological Societies and the Special Interest Group on Neuropathic Pain (NeuPSIG) of the International Association for the Study of Pain recommend oral medicines, such as tricyclic antidepressants, anticonvulsants (including gabapentin and pregabalin), and selective serotonin and noradrenaline reuptake inhibitors as first-line options [Citation5,Citation26,Citation27]. However, these oral therapies provide satisfactory pain relief only in a minority 30–40% of patients. They are also associated with undesirable systemic side effects. As a consequence, many individuals with neuropathic pain still suffer from persistent pain and poor quality of life despite their use [Citation9,Citation28Citation30].

A pharmacologic treatment algorithm for localized NP has been suggested: primary care physicians and non-pain specialists should consider the use of topical analgesic agents as the first-line treatment [Citation31]. Overall, topical agents targeting the peripheral nervous system can be effective at providing rapid, targeted pain relief of PNP without the side effects, which are sometimes associated with systemic, oral therapies. Topical agents may be especially useful when there are concerns about systemic side effects or compliance, and in frail/elderly patients [Citation9]. Indeed, localized activity and low systemic absorption can prevent issues associated with oral or intravenous routes, such as gastric disturbances, CNS side-effects and variable serum concentrations, thus resulting in a low risk of drug–drug interactions [Citation9].

Two topical treatments are currently licensed by the European Medicines Agency (EMA) for peripheral NP: lidocaine 5%-medicated plaster for PHN only and the capsaicin 179 mg 8% cutaneous patch (capsaicin 8% patch) for all types of peripheral neuropathic pain.

3. Capsaicin: pharmacodynamic and pharmacokinetic properties

Inactivation of NaV channels and pharmacological desensitization of TRPV1 receptors may contribute to an immediate reduction in neuronal excitability and responsiveness [Citation5]. Capsaicin is an agonist at the TRPV1 ligand-gated cation channel, which is highly expressed in nociceptive nerve fibers (mainly C and Aδ fibers) [Citation1]. Multiple mechanisms are involved in capsaicin-induced so-called ‘defunctionalization’ of the sensory nerve fibers [Citation5].

Furthermore, topical exposure to capsaicin causes sensations of heat, burning, stinging or itching, since activation of TRPV1 results in sensory neuronal depolarization, and can induce local sensitization to receptor activation by heat, acidosis and endogenous agonists. However, high concentrations of capsaicin or repeated applications can lead to a persistent local effect on cutaneous nociceptors, constituted by reduced spontaneous activity and a loss of responsiveness to a wide range of sensory stimuli [Citation5]. At concentrations higher than those required to activate TRPV1, capsaicin can also cause mitochondria dysfunction by directly inhibiting electron chain transport. At the same time, the peripheral fibers in the affected zone pull back from their cutaneous innervation territories rendering them less likely to be activated by peripheral stimuli. Thus, the peripheral fibers with their nociceptors and associated ion channels are no longer amenable to activation by stimuli applied to the treated area, and central sensitization is thereby attenuated. This retraction is due to several effects that include temporary loss of membrane potential, inability to transport neurotrophic factors and reversible retraction of epidermal and dermal nociceptive fiber terminals, which all block the transmission of nociceptive stimuli for a prolonged period [Citation5,Citation32]. This condition is temporary and reversible, and terminals are usually reconstituted within 3 months after the administration of capsaicin. Remarkably, due to the high selectivity of capsaicin for the TRPV1 receptor and the selective expression of TRPV1 in nociceptive sensory nerves, other skin sensory nerve endings may remain intact and functional, with no loss of tactile and vibratory sensations [Citation33].

Topical capsaicin acts locally, and pain relief is not facilitated by transdermal systemic delivery. Indeed, due to its insolubility in water, capsaicin is not readily absorbed into the microvasculature [Citation5,Citation6]. Over a 60-min application of the capsaicin 8% dermal patch, ≈1% of capsaicin is estimated to be absorbed into the epidermal and dermal layers of the skin [Citation6]. In patients with PHN, HIV-AN or PDPN, systemic exposure to capsaicin appeared to be low and transient following a single 60- or 90-min application of the capsaicin 8% dermal patch [Citation34]. Capsaicin is rapidly metabolized in vitro by liver enzymes to form three major metabolites – that is, 16-hydroxycapsaicin, 17-hydroxycapsaicin and 16,17-dehydrocapsaicin. These metabolites were not detected in the plasma of patients with peripheral neuropathy treated with topical capsaicin and are not pharmacologically active on TRPV1 receptors [Citation1].

4. Therapeutic efficacy of the Capsaicin 8% dermal patch

The therapeutic efficacy of the high-concentration 8% capsaicin dermal patch for the treatment of PNP has been assessed in patients with PDPN and in nondiabetic patients with PNP of various etiologies, including PHN and HIV-AN, both in large, randomized clinical trials ( and ) [Citation35Citation47], and in ‘field-practice’ experiences (-) [Citation8,Citation48Citation67].

Table 1. Randomized short therapy (8–12 weeks) clinical trials on the efficacy of capsaicin 8% dermal patch.

Table 2. Randomized long therapy (48–52 weeks) clinical trials on the efficacy of capsaicin 8% dermal patch.

Table 3. Field practice experiences on the efficacy of capsaicin 8% dermal patch for short time therapy (2, 4 and 8 weeks).

Table 4. Field practice experiences on the efficacy of capsaicin 8% dermal patch for medium time treatment (12 weeks).

Table 5. Field practice experiences on the efficacy of capsaicin 8% dermal patch for long time treatment (6 months, 1 year).

Table 6. Risk of adverse events for the different oral agents in comparison to placebo, data taken from [Citation68].

A detailed description of those trials is provided in an excellent recent review by Blair et al., and goes beyond the scopes of the present article [Citation1]. We focus here on the studies comparing capsaicin with pregabalin, a centrally acting agent widely used for neuropathic pain.

In the open-label, randomized, multicenter, non-inferiority ELEVATE trial, 282 patients with PNP received capsaicin 8%, while 277 were assigned to pregabalin [Citation36]. The primary endpoint was a ≥ 30% mean decrease in Numeric Pain Rating Scale (NPRS) score from baseline to week 8. Secondary endpoints were time-to-onset of pain relief and treatment satisfaction. Overall, the capsaicin 8% patch was non-inferior to pregabalin in the achievement of a ≥ 30% mean decrease in NPRS score at week 8 (55.7% vs 54.5%, respectively; odds ratio: 1.03 [95% CI: 0.72–1.50]). The median time-to-onset of pain relief was shorter for capsaicin 8% patch compared with pregabalin (7.5 vs 36.0 days; hazard ratio: 1.68 [95% CI: 1.35–2.08]; p < 0.0001). In addition, treatment satisfaction was greater with the capsaicin 8% patch compared with pregabalin. Capsaicin was also associated with a more favorable tolerability profile compared with pregabalin (incidence of systemic drug reactions with capsaicin: 0–1.1%; with pregabalin: 2.5–18.4%). All adverse events were of mild-to-moderate severity; treatment discontinuation only occurred with pregabalin (n = 24). The authors concluded that the capsaicin 8% patch provides non-inferior pain relief compared with an optimized dose of pregabalin in PNP, and showed a faster onset of action, fewer systemic side effects and greater treatment satisfaction. In a subsequent analysis of the same trial, the capsaicin 8% dermal patch was superior to pregabalin in reducing the intensity and area of dynamic mechanical allodynia (DMA), a common clinical manifestation of PNP and a consequence of central sensitization [Citation11]. Indeed, the change in DMA intensity from baseline to study end was greater with the capsaicin 8% patch, as compared with pregabalin [−0.63 (95% CI: −1.04 to −0.23; p = 0.002)]. Similarly, the capsaicin 8% patch was superior over pregabalin in the reduction of DMA area [−39.5 cm2 (95% CI: −69.1 to −10.0; p = 0.009)]. A greater number of patients experienced a complete resolution of allodynia with the capsaicin 8% patch treatment compared with pregabalin (24.1% vs 12.3%; p = 0.001).

A network meta-analysis of 25 randomized controlled trials showed that the capsaicin 8% dermal patch was just as effective as oral, centrally acting agents (i.e. pregabalin, duloxetine and gabapentin) in patients with PDPN; however, it demonstrated benefits of better tolerability due to a lack of systemic effects [Citation68]. For the endpoint of ≥30% pain reduction, the capsaicin 8% patch was significantly more effective than placebo (OR: 2.28 [95% CI: 1.19–4.03]), showed a clear advantage over pregabalin (OR: 1.83 [95% CI: 0.91–3.34]) and gabapentin (OR: 1.66 [95% CI: 0.74–3.23]), and exhibited a similar efficacy compared with duloxetine (OR: 0.99 [95% CI: 0.5–1.79]). Compared with placebo, oral agents were correlated with a significant elevation in the risk of somnolence, dizziness, fatigue and discontinuation due to adverse events ().

Lastly, to date, published cost–effectiveness analyses of patients who suffer from PNP suggest that the capsaicin 8% dermal patch is cost-effective compared with oral agents, including tricyclic antidepressants, duloxetine, gabapentin and pregabalin (i.e. 59,919 USD vs tricyclic antidepressants; 43,908 USD vs duloxetine; 42008 USD vs gabapentin; 40241 USD versus pregabalin) [Citation69].

5. Expert opinion

The activity of capsaicin is entirely dependent on the amount of TRPV1 on damaged fibers or fibers involved in pain generation. The expression of TRPV1 might be different among patients with the same form of localized PNP and an accurate clinical evaluation of their presence is a prerequisite for a fully successful therapy. Indeed, ‘defunctionalization’ of small afferent fibers localized in the area of skin application is the key pharmacodynamic property of capsaicin 8% patch. This is a long-lasting phenomenon that allows the drug to produce analgesic effects well beyond its removal. This aspect along with the fact that this is a topical therapy with no systemic adverse drug reactions, make capsaicin 8% patch an ideal approach to promote patient adherence to therapy.

It must be pointed out that the efficacy of capsaicin 8% patch requires access to its target and so will be dependent on the peripheral expression of TRPV1 receptors in a pain patient. Therefore, testing of thermal sensibility (hot/cold) should be performed before application. Pain at application is a good predictive factor, since it can be attributed to the presence of TRPV1 receptors in the application zone.

In any case, a reduction in the painful area is observed after the first application, while reduction of pain intensity is observed after 2–8 weeks of the application [Citation1]. The efficacy of capsaicin 8% is greatest if the pain generator is superficial in the affected tissue (approximately 1–1.5 cm), such as in the case of PHN, DPN, painful post-surgical scars (e.g., thoracic/abdominal surgery, orthopedic surgery, hernia removal) or ischemic pain. Advantages of capsaicin 8% patch treatment include its excellent efficacy – which is associated with a reduction of painful area – and systemic tolerability, which make it a suitable therapy in elderly patients, and ease of use. It is particularly suitable for patients with burning pain and allodynia due to peripheral and consequent central sensitization. Other potentially suitable patients include those with contraindication to systemic treatment (e.g., patients on working duties and long-driving requirements), oncological patients – given the lack of interactions with oncological therapies – and patients with chemotherapy-induced neuropathy.

The ability of capsaicin 8% patch in reducing the intensity and area of pain in patients with localized NP whilst avoiding central and systemic side-effects is a great advantage over oral agents. Moreover, given the high patients’ preference for capsaicin and because of the unique pharmacodynamics properties of this drug, capsaicin 8% patch is also suitable for combination therapy, in which synergistic effects can be obtained only by combining drugs with different and complimentary mechanisms of action. Moreover, it can be given as a short course of treatment, with the possibility to repeat as needed. Proper evaluation of the area to be treated (measurement, testing of skin integrity, assessment of thermal sensitivity), as well as proper training of healthcare professionals involved is essential.

In conclusion, topical capsaicin 8% patch has the potential to represent a first-choice treatment for localized PNP, given its peculiar pharmacodynamic properties and its good efficacy/safety ratio.

Article highlights

  • Peripheral neuropathic pain (PNP) is a chronic condition arising from damaged fibers of the somatosensory system and involves the transient receptor potential vanilloid-1 (TRPV-1).

  • Oral drugs are satisfactory to only a portion of patients.

  • Topical treatment with capsaicin is efficient in attenuating pain and is characterized by few side effects.

  • Capsaicin acts through pharmacological defunctionalization of the TRPV-1 receptor.

  • Capsaicin is administered by a single application of a dermal patch; its efficacy has been tested both in clinical trials and in ‘field-practice’ experiences and it is cost-effective compared to other oral agents.

  • Topical capsaicin dermal patch may represent a suitable choice for treatment for localized PNP.

This box summarizes key points contained in the article.

Declaration of interest

AH Dickenson has received speaker fees from Grunenthal, Teva and Allergan. E Polati has received fees as both a speaker and/or consultant from Grunenthal, Alfasigma and Pfizer. D Fornasari has received fees as speaker and/or consultant from: Abiogen, Alfasigma, Bayer, Grunenthal, Lundbeck, Sandoz, SPA and Zambon. P Zini is an employee of Grunenthal. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer Disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Acknowledgments

Editorial assistance was provided by Luca Giacomelli, PhD, Barbara Bartolini, PhD, and Aashni Shah (Polistudium SRL, Milan, Italy), and was supported by Grunethal.

Additional information

Funding

Editorial assistance was utilized in this manuscript and was funded by Grunenthal.

References

  • Blair HA. Capsaicin 8% dermal patch: a review in peripheral neuropathic pain. Drugs. 2018;78(14):1489–1500.
  • Zilliox LA. Neuropathic pain. Continuum. 2017;23(2):512–532.
  • Anand P, Bley K. Topical capsaicin for pain management: therapeutic potential and mechanisms of action of the new high-concentration capsaicin 8% patch. Br J Anaesth. 2011;107(4):490–502.
  • European Medicines Agency. Summary of product characteristics: qutenza 179 mg cutaneous patch. 2017. cited 2018 Sep 10. Available from: http://www.ema.europa.eu.
  • Colloca L, Ludman T, Bouhassira D, et al. Neuropathic pain. Nat Rev Dis Primers. 2017;3:17002.
  • O’Neill J, Brock C, Olesen AE, et al. Unravelling the mystery of capsaicin: a tool to understand and treat pain. Pharmacol Rev. 2012;64(4):939–971.
  • European Medicines Agency. CHMP assessment report for Qutenza. 2009. Available from: cited 2018 Sep 10 http://www.ema.europa.eu.
  • Peppin JF, Majors K, Webster LR, et al. Tolerability of NGX- 4010, a capsaicin 8% patch for peripheral neuropathic pain. J Pain Res. 2011;4:385–392.
  • Sommer C, Cruccu G. Topical treatment of peripheral neuropathic pain: applying the evidence. J Pain Symptom Manage. 2017;53(3):614–629.
  • Mick G, Baron R, Finnerup NB, et al. What is localized neuropathic pain? A first proposal to characterize and define a widely used term. Pain Manag. 2012;2(1):71–77.
  • Cruccu G, Truini A. A review of neuropathic pain: from guidelines to clinical practice. Pain Ther. 2017;6(Suppl 1):35–42.
  • Pickering G, Martin E, Tiberghien F, et al. Localized neuropathic pain: an expert consensus on local treatments. Drug Des Devel Ther. 2017;11:2709–2718.
  • Price TJ, Gold MS. From mechanism to cure: renewing the goal to eliminate the disease of pain. Pain Med. 2018;19(8):1525–1549.
  • Usoskin D, Furlan A, Islam S, et al. Unbiased classification of sensory neuron types by large-scale single-cell RNA sequencing. Nat Neurosci. 2015;18(1):145–153.
  • Häring M, Zeisel A, Hochgerner H, et al. Neuronal atlas of the dorsal horn defines its architecture and links sensory input to transcriptional cell types. Nat Neurosci. 2018;21(6):869–880.
  • Nau C, Leipold E Voltage-gated sodium channels and pain. cited 20 June 2019. Available at: https://www.degruyter.com/view/j/nf.2017.23.issue-3/nf-2017-A017/nf-2017-A017.xml.
  • Dib-Hajj SD, Waxman SG. Diversity of composition and function of sodium channels in peripheral sensory neurons. Pain. 2015;156(12):2406–2407.
  • Hu T, Liu N, Lv M, et al. Lidocaine inhibits HCN currents in rat spinal substantia gelatinosa neurons. Anesth Analg. 2016;122(4):1048–1059.
  • Dickenson AH, Patel R. Sense and sensibility-logical approaches to profiling in animal models. Pain. 2018;159(7):1426–1428.
  • Demant DT, Lund K, Vollert J, et al. The effect of oxcarbazepine in peripheral neuropathic pain depends on pain phenotype: a randomised, double-blind, placebo-controlled phenotype-stratified study. Pain. 2014;155(11):2263–2273.
  • Vay L, Gu C, McNaughton PA. The thermo-TRP ion channel family: properties and therapeutic implications. Br J Pharmacol. 2012;165(4):787–801.
  • Kremeyer B, Lopera F, Cox JJ, et al. A gain-of-function mutation in TRPA1 causes familial episodic pain syndrome. Neuron. 2010;66(5):671–680.
  • Todd AJ. Neuronal circuitry for pain processing in the dorsal horn. Nat Rev Neurosci. 2010;11(12):823–836.
  • Gracely RH, Lynch SA, Bennett GJ. Painful neuropathy: altered central processing maintained dynamically by peripheral input. Pain. 1992;51(2):175–194.
  • Baron R, Hans G, Dickenson AH. Peripheral input and its importance for central sensitization. Ann Neurol. 2013;74(5):630–636.
  • Attal N, Cruccu G, Baron R, et al. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. Eur J Neurol. 2010;17:1113e1123.
  • Finnerup NB, Attal N, Haroutounian S, et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol. 2015;14:162e173.
  • Gilron I. Treatment of neuropathic pain: antiepileptic and antidepressant drugs educational objectives. In: Sommer CL, Raja SN, editors. Pain 2014: refresher courses, 15th world congress on pain. Washington, DC, USA: IASP Press; 2014. p. 225e237.
  • Freynhagen R, Serpell M, Emir B, et al. A comprehensive drug safety evaluation of pregabalin in peripheral neuropathic pain. Pain Pract. 2015;15:47e57.
  • Torrance N, Ferguson JA, Afolabi E, et al. Neuropathic pain in the community: more under-treated than refractory? Pain. 2013;154:690e699.
  • Allegri M, Baron R, Hans G, et al. A pharmacological treatment algorithm for localized neuropathic pain. Curr Med Res Opin. 2016;32:377–384.
  • Martini C, Yassen A, Olofsen E, et al. Pharmacodynamic analysis of the analgesic effect of capsaicin 8% patch (Qutenza™) in diabetic neuropathic pain patients: detection of distinct response groups. J Pain Res. 2012;5:51–59.
  • Bley KR. TRPV1 agonist approaches for pain management. In: Gomtsyan A, Faltynek CR, editors. Vanilloid receptor TRPV1 in drug discovery: targeting pain and other pathological disorders. New York: Wiley; 2010. p. 325–347.
  • Babbar S, Marier J-F, Mouksassi M-S, et al. Pharmacokinetic analysis of capsaicin after topical administration of a high-concentration capsaicin patch to patients with peripheral neuropathic pain. Ther Drug Monit. 2009;31(4):502–510.
  • Cruccu G, Nurmikko TJ, Ernault E, et al. Superiority of capsaicin 8% patch versus oral pregabalin on dynamic mechanical allodynia in patients with peripheral neuropathic pain. Eur J Pain. 2018;22(4):700–706.
  • Haanpaa M, Cruccu G, Nurmikko TJ, et al., Capsaicin 8% patch versus oral pregabalin in patients with peripheral neuropathic pain. Eur J Pain. 20(2): 316–328. 2016. .
  • Simpson DM, Robinson-Papp J, Van J, et al. Capsaicin 8% patch in painful diabetic peripheral neuropathy: a randomized, doubleblind, placebo-controlled study. J Pain. 2017;18(1):42–53.
  • Brown S, Simpson DM, Moyle G, et al. NGX-4010, a capsaicin 8% patch, for the treatment of painful HIV-associated distal sensory polyneuropathy: integrated analysis of two phase III, randomized, controlled trials. AIDS Res Ther. 2013;10(1):5.
  • Clifford DB, Simpson DM, Brown S, et al. A randomized, double-blind, controlled study of NGX-4010, a capsaicin 8% dermal patch, for the treatment of painful HIV-associated distal sensory polyneuropathy. J Acquir Immune Defic Syndr. 2012;59(2):126–133.
  • Irving GA, Backonja MM, Dunteman E, et al. A multicenter, randomized, double-blind, controlled study of NGX-4010, a highconcentration capsaicin patch, for the treatment of postherpetic neuralgia. Pain Med. 2011;12:99–109.
  • Simpson DM, Brown S, Tobias J. Controlled trial of high-concentration capsaicin patch for treatment of painful HIV neuropathy. Neurology. 2008;70(24):2305–2313.
  • Backonja M, Wallace MS, Blonsky ER, et al. NGX-4010, a highconcentration capsaicin patch, for the treatment of postherpetic neuralgia: a randomised, double-blind study. Lancet Neurol. 2008;7(12):1106–1112.
  • Vinik AI, Perrot S, Vinik EJ, et al. Capsaicin 8% patch repeat treatment plus standard of care (SOC) versus SOC alone in painful diabetic peripheral neuropathy: a randomised, 52-week, open-label, safety study. BMC Neurol. 2016;16(251):1–14.
  • Perrot S, Ortega E, Vinik EJ, et al. Efficacy, quality of life and treatment satisfaction with capsaicin 8% patch versus standard of care in painful diabetic peripheral neuropathy [abstract no. 1067]. Diabetologia. 2015;58(Suppl.1):S514.
  • Simpson DM, Brown S, Tobias JK, et al., NGX-4010 C107 Study Group. NGX-4010, a capsaicin 8% dermal patch, for the treatment of painful HIV-associated distal sensory polyneuropathy: results of a 52-week open-label study. Clin J Pain. 2014;30(2):134–142.
  • Backonja MM, Malan TP, Vanhove GF. NGX-4010, a highconcentration capsaicin patch, for the treatment of postherpetic neuralgia: a randomized, double-blind, controlled study with an open-label extension. Pain Med. 2010;11:600–608.
  • Simpson DM, Gazda S, Brown S, et al. Long-term safety of NGX-4010, a high-concentration capsaicin patch, in patients with peripheral neuropathic pain. J Pain Symptom Manage. 2010;39(6):1053–1064.
  • Churruca I, Miro P, Suñer R, et al. Capsaicin (8%) patch in urological patients with neuropathic pain due to scar [abstract]. Presented at: 9th World Congress of the World Institute of Pain. 2018 Aug 24.
  • Tenreiro Pinto J, Pereira FC, Loureiro MC, et al. Efficacy analysis of capsaicin 8% patch in neuropathic peripheral pain treatment. Pharmacology. 2018;101(5–6):290–297.
  • Churruca I. Capsaicin 8% patch in urological patients with neuropathic pain [abstract no. WIP16-0048]. Pain Pract. 2016;16(Suppl.1):48.
  • Privitera R, Birch R, Sinisi M, et al. Capsaicin 8% patch treatment for amputation stump and phantom limb pain: a clinical and functional MRI study. J Pain Res. 2017;10:1623–1634.
  • Bardo-Brouard P, Luizard C, Valeyrie-Allanore L, et al. High-concentration topical capsaicin in the management of refractory neuropathic pain in patients with neurofibromatosis type 1: a case series. Curr Med Res Opin. 2018;34(5):887–891.
  • Mankowski C, Poole CD, Ernault E, et al. Effectiveness of the capsaicin 8% patch in the management of peripheral neuropathic pain in European clinical practice: the ASCEND study. BMC Neurol. 2017;17(80):1–11.
  • Levesque A, Riant T, Labat -J-J, et al. Use of high-concentration capsaicin patch for the treatment of pelvic pain: observational study of 60 inpatients. Pain Physician. 2017;20(1):E161–7.
  • Perrot S, Lantéri-Minet M. Patients’ global impression of change in the management of peripheral neuropathic pain: clinical relevance and correlations in daily practice. Epub 2019 Mar 18 Eur J Pain. 2019;236:1117–1128.
  • Hansson P, Jensen TS, Kvarstein G, et al. Pain-relieving effectiveness, quality of life and tolerability of repeated capsaicin 8% patch treatment of peripheral neuropathic pain in Scandinavian clinical practice. Eur J Pain. 2018;22(5):941–950.
  • Gustorff B, Poole C, Kloimstein H, et al. Treatment of neuropathic pain with the capsaicin 8% patch: quantitative sensory testing (QST) in a prospective observational study identifies potential predictors of response to capsaicin 8% patch treatment. Scand J Pain. 2017;4(3):138–145.
  • Filipczak-Bryniarska I, Krzyzewski RM, Kucharz J, et al. High-dose 8% capsaicin patch in treatment of chemotherapy-induced peripheral neuropathy: single-center experience. Med Oncol. 2017;34(162):1–5.
  • Aitken E, McColl G, Kingsmore D. The role of Qutenza® (topical capsaicin 8%) in treating neuropathic pain from critical ischemia in patients with end-stage renal disease: an observational cohort study. Pain Med. 2017;18(2):330–340.
  • Zis P, Bernali N, Argira E, et al. Effectiveness and impact of capsaicin 8% patch on quality of life in patients with lumbosacral pain: an open-label study. Pain Physician. 2016;19(7):E1049–53.
  • Maihofner CG, Heskamp ML. Treatment of peripheral neuropathic pain by topical capsaicin: impact of pre-existing pain in the QUEPP-study. Eur J Pain. 2014;18(5):671–679.
  • Maihofner C, Heskamp ML. Prospective, non-interventional study on the tolerability and analgesic effectiveness over 12 weeks after a single application of capsaicin 8% cutaneous patch in 1044 patients with peripheral neuropathic pain: first results of the QUEPP study. Curr Med Res Opin. 2013;29(6):673–683.
  • Lantéri-Minet M, Perrot S. QAPSA: post-marketing surveillance of capsaicin 8% patch for long-term use in patients with peripheral neuropathic pain in France. Curr Med Res Opin. 2019;35(3):417–426.
  • Galvez R, Navez ML, Moyle G, et al. Capsaicin 8% patch repeat treatment in nondiabetic peripheral neuropathic pain: a 52-week, open-label, single-arm, safety study. Clin J Pain. 2017;33(10):921–931.
  • Rorbaek M, Ventzel L, Gottrup H. Treatment with topical capsaicin: experience from a pain clinic [abstract no. F34]. Scand J Pain. 2017;3(3):197.
  • Marec CLE, Berard J, Queneuille I, et al. Improvement of chemotherapy induced neuropathy (CIN) in cancer patients using capsaicin 8% patch [abstract no. e14031]. J Clin Oncol. 2016;34(15Suppl):e14031–e14031.
  • Reeves K, Tilak D, Putt O. Is an 8% capsicum patch (Qutenza) an alternative to traditional neuropathic pain management following breast surgery [abstract no. 138460]? Reg Anesth Pain Med. 2016;41(5Suppl.):e116.
  • van Nooten F, Treur M, Pantiri K, et al., Capsaicin 8% patch versus oral neuropathic pain medications for the treatment of painful diabetic peripheral neuropathy: a systematic literature review and network meta-analysis. Clin Ther. 39(4): 787–803. 2017. .
  • Mankowski C, Patel S, Trueman D, et al. Cost-effectiveness of capsaicin 8% patch compared with pregabalin for the treatment of patients with peripheral neuropathic pain in Scotland. PLoS One. 2016;11(3):e0150973.