Stratifying patients with peripheral neuropathic pain based on sensory profiles: algorithm and sample size recommendations

Jan Vollert, Christoph Maier, Nadine Attal, David L H Bennett, Didier Bouhassira, Elena K Enax-Krumova, Nanna B Finnerup, Rainer Freynhagen, Janne Gierthmühlen, Maija Haanpää, Per Hansson, Philipp Hüllemann, Troels S Jensen, Walter Magerl, Juan D Ramirez, Andrew S C Rice, Sigrid Schuh-Hofer, Märta Segerdahl, Jordi Serra, Pallai R Shillo, Soeren Sindrup, Solomon Tesfaye, Andreas C Themistocleous, Thomas R Tölle, Rolf-Detlef Treede, Ralf Baron, Jan Vollert, Christoph Maier, Nadine Attal, David L H Bennett, Didier Bouhassira, Elena K Enax-Krumova, Nanna B Finnerup, Rainer Freynhagen, Janne Gierthmühlen, Maija Haanpää, Per Hansson, Philipp Hüllemann, Troels S Jensen, Walter Magerl, Juan D Ramirez, Andrew S C Rice, Sigrid Schuh-Hofer, Märta Segerdahl, Jordi Serra, Pallai R Shillo, Soeren Sindrup, Solomon Tesfaye, Andreas C Themistocleous, Thomas R Tölle, Rolf-Detlef Treede, Ralf Baron

Abstract

In a recent cluster analysis, it has been shown that patients with peripheral neuropathic pain can be grouped into 3 sensory phenotypes based on quantitative sensory testing profiles, which are mainly characterized by either sensory loss, intact sensory function and mild thermal hyperalgesia and/or allodynia, or loss of thermal detection and mild mechanical hyperalgesia and/or allodynia. Here, we present an algorithm for allocation of individual patients to these subgroups. The algorithm is nondeterministic-ie, a patient can be sorted to more than one phenotype-and can separate patients with neuropathic pain from healthy subjects (sensitivity: 78%, specificity: 94%). We evaluated the frequency of each phenotype in a population of patients with painful diabetic polyneuropathy (n = 151), painful peripheral nerve injury (n = 335), and postherpetic neuralgia (n = 97) and propose sample sizes of study populations that need to be screened to reach a subpopulation large enough to conduct a phenotype-stratified study. The most common phenotype in diabetic polyneuropathy was sensory loss (83%), followed by mechanical hyperalgesia (75%) and thermal hyperalgesia (34%, note that percentages are overlapping and not additive). In peripheral nerve injury, frequencies were 37%, 59%, and 50%, and in postherpetic neuralgia, frequencies were 31%, 63%, and 46%. For parallel study design, either the estimated effect size of the treatment needs to be high (>0.7) or only phenotypes that are frequent in the clinical entity under study can realistically be performed. For crossover design, populations under 200 patients screened are sufficient for all phenotypes and clinical entities with a minimum estimated treatment effect size of 0.5.

Figures

Figure 1.
Figure 1.
Receiver Operating Characteristic analysis of the discriminatory power of the proposed algorithm to separate between patients with neuropathic pain and healthy subjects. Black line: full sensory testing, gray line: reduced protocol, using only warm detection threshold and mechanical pain sensitivity. The green dotted diagonal line indicates random classification (“coin flipping”). The area marked by dashed lines indicates the optimum ratio of sensitivity and specificity at 64% for probability for being healthy for full phenotyping (reduced phenotyping: 63%).
Figure 2.
Figure 2.
Sensory phenotype probabilities and probability of being healthy for (A) n = 902 patients with neuropathic pain and (B) n = 188 healthy subjects. Gray line: probability for being healthy, blue line: sensory loss, red line: thermal hyperalgesia, yellow line: mechanical hyperalgesia. Subjects on the x-axis are sorted by their individual probability of being healthy. Dotted line: a phenotype with a probability over 64% should be considered relevant in the individual patient. Thirteen healthy subjects (7%) did not reach this criterion, 198 patients (22%) had profiles consistent with being normal.
Figure 3.
Figure 3.
Sensory phenotype frequency and overlap between phenotypes for (A) diabetic polyneuropathy, (B) peripheral nerve injury, and (C) postherpetic neuralgia. Gray: healthy (H), blue: sensory loss (SL), red: thermal hyperalgesia (TH), yellow: mechanical hyperalgesia (MH). First bar (DET): deterministic algorithm (adds to 100%), 3 subsequent bars: probabilistic approach (a patient may be allocated to more than one phenotype, percentages are not additive). Bars are to scale, sizes of the circles in Venn diagrams and their overlaps are illustrative, not to scale.

References

    1. Attal N, de Andrade DC, Adam F, Ranoux D, Teixeira MJ, Galhardoni R, Raicher I, Üçeyler N, Sommer C, Bouhassira D. Safety and efficacy of repeated injections of botulinum toxin A in peripheral neuropathic pain (BOTNEP): a randomised, double-blind, placebo-controlled trial. Lancet Neurol 2016;15:555–65.
    1. Attal N, Rouaud J, Brasseur L, Chauvin M, Bouhassira D. Systemic lidocaine in pain due to peripheral nerve injury and predictors of response. Neurology 2004;62:218–25.
    1. Backonja MM, Attal N, Baron R, Bouhassira D, Drangholt M, Dyck PJ, Edwards RR, Freeman R, Gracely R, Haanpaa MH, Hansson P, Hatem SM, Krumova EK, Jensen TS, Maier C, Mick G, Rice AS, Rolke R, Treede RD, Serra J, Toelle T, Tugnoli V, Walk D, Walalce MS, Ware M, Yarnitsky D, Ziegler D. Value of quantitative sensory testing in neurological and pain disorders: NeuPSIG consensus. PAIN 2013;154:1807–19.
    1. Baron R, Förster M, Binder A. Subgrouping of patients with neuropathic pain according to pain-related sensory abnormalities: a first step to a stratified treatment approach. Lancet Neurol 2012;11:999–1005.
    1. Baron R, Maier C, Attal N, Binder A, Bouhassira D, Cruccu G, Finnerup NB, Haanpaa M, Hansson P, Hullemann P, Jensen TS, Freynhagen R, Kennedy JD, Magerl W, Mainka T, Reimer M, Rice ASC, Segerdahl M, Serra J, Sindrup S, Sommer C, Tolle T, Vollert J, Treede RD. Peripheral Neuropathic Pain: a mechanism-related organizing principle based on sensory profiles. PAIN 2017;158:261–72.
    1. Baumgartner U, Magerl W, Klein T, Hopf HC, Treede RD. Neurogenic hyperalgesia versus painful hypoalgesia: two distinct mechanisms of neuropathic pain. PAIN 2002;96:141–51.
    1. Bouhassira D, Attal N. Translational neuropathic pain research: a clinical perspective. Neuroscience 2016;338:27–35.
    1. DeLong ER, DeLong DM, Clarke-Pearson DL. Comparing the areas under two or more correlated receiver operating characteristic curves: a nonparametric approach. Biometrics 1988;44:837–45.
    1. Demant DT, Lund K, Finnerup NB, Vollert J, Maier C, Segerdahl MS, Jensen TS, Sindrup SH. Pain relief with lidocaine 5% patch in localized peripheral neuropathic pain in relation to pain phenotype: a randomised, double-blind, and placebo-controlled, phenotype panel study. PAIN 2015;156:2234–44.
    1. Demant DT, Lund K, Vollert J, Maier C, Segerdahl M, Finnerup NB, Jensen TS, Sindrup SH. The effect of oxcarbazepine in peripheral neuropathic pain depends on pain phenotype: a randomised, double-blind, placebo-controlled phenotype-stratified study. PAIN 2014;155:2263–73.
    1. Dworkin RH, Edwards RR. Phenotypes and treatment response. PAIN 2017:158;187–89.
    1. Edwards RR, Dworkin RH, Turk DC, Angst MS, Dionne R, Freeman R, Hansson P, Haroutounian S, Arendt-Nielsen L, Attal N, Baron R, Brell J, Bujanover S, Burke LB, Carr D, Chappell AS, Cowan P, Etropolski M, Fillingim RB, Gewandter JS, Katz NP, Kopecky EA, Markman JD, Nomikos G, Porter L, Rappaport BA, Rice ASC, Scavone JM, Scholz J, Simon LS, Smith SM, Tobias J, Tockarshewsky T, Veasley C, Versavel M, Wasan AD, Wen W, Yarnitsky D. Patient phenotyping in clinical trials of chronic pain treatments: IMMPACT recommendations. PAIN 2016;157:1851–71.
    1. Edwards RR, Haythornthwaite JA, Tella P, Max MB, Raja S. Basal heat pain thresholds predict opioid analgesia in patients with postherpetic neuralgia. Anesthesiology 2006;104:1243–8.
    1. European Medicines Agency. EMA/CHMP/970057/2011: guideline on the clinical development of medicinal products intended for the treatment of pain. Available at: . Accessed December 15, 2016.
    1. Faul F, Erdfelder E, Lang AG, Buchner A. G*Power 3: a flexible statistical power analysis program for the social, behavioral, and biomedical sciences. Behav Res Methods 2007;39:175–91.
    1. Fields HL, Rowbotham M, Baron R. Postherpetic neuralgia: irritable nociceptors and deafferentation. Neurobiol Dis 1998;5:209–27.
    1. Finnerup NB, Attal N, Haroutounian S, McNicol E, Baron R, Dworkin RH, Gilron I, Haanpaa M, Hansson P, Jensen TS, Kamerman PR, Lund K, Moore A, Raja SN, Rice ASC, Rowbotham M, Sena E, Siddall P, Smith BH, Wallace M. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol 2015;14:162–73.
    1. Finnerup NB, Haroutounian S, Kamerman P, Baron R, Bennett DLH, Bouhassira D, Cruccu G, Freeman R, Hansson P, Nurmikko T, Raja SN, Rice ASC, Serra J, Smith BH, Treede RD, Jensen TS. Neuropathic pain: an updated grading system for research and clinical practice. PAIN 2016;157:1599–606.
    1. Fleiss JL. Statistical methods for rates and proportions. New York: Wiley, 1973.
    1. Fruhstorfer H. Thermal sensibility changes during ischemic nerve block. PAIN 1984;20:355–61.
    1. Gierthmuhlen J, Enax-Krumova EK, Attal N, Bouhassira D, Cruccu G, Finnerup NB, Haanpaa M, Hansson P, Jensen TS, Freynhagen R, Kennedy JD, Mainka T, Rice ASC, Segerdahl M, Sindrup SH, Serra J, Tolle T, Treede R-D, Baron R, Maier C. Who is healthy? Aspects to consider when including healthy volunteers in QST–based studies-a consensus statement by the EUROPAIN and NEUROPAIN consortia. PAIN 2015;156:2203–11.
    1. Gustorff B, Sycha T, Lieba-Samal D, Rolke R, Treede RD, Magerl W. The pattern and time course of somatosensory changes in the human UVB sunburn model reveal the presence of peripheral and central sensitization. PAIN 2013;154:586–97.
    1. Katz J, Finnerup NB, Dworkin RH. Clinical trial outcome in neuropathic pain: relationship to study characteristics. Neurology 2008;70:263–72.
    1. Katz NP, Mou J, Paillard FC, Turnbull B, Trudeau J, Stoker M. Predictors of response in patients with postherpetic neuralgia and HIV-associated neuropathy treated with the 8% capsaicin patch (Qutenza). Clin J Pain 2015;31:859–66.
    1. Klein T, Magerl W, Rolke R, Treede RD. Human surrogate models of neuropathic pain. PAIN 2005;115:227–33.
    1. Lang S, Klein T, Magerl W, Treede RD. Modality-specific sensory changes in humans after the induction of long-term potentiation (LTP) in cutaneous nociceptive pathways. PAIN 2007;128:254–63.
    1. Lotsch J, Oertel BG, Ultsch A. Human models of pain for the prediction of clinical analgesia. PAIN 2014;155:2014–21.
    1. Magerl W, Krumova EK, Baron R, Tolle T, Treede RD, Maier C. Reference data for quantitative sensory testing (QST): refined stratification for age and a novel method for statistical comparison of group data. PAIN 2010;151:598–605.
    1. Maier C, Baron R, Tolle TR, Binder A, Birbaumer N, Birklein F, Gierthmuhlen J, Flor H, Geber C, Huge V, Krumova EK, Landwehrmeyer GB, Magerl W, Maihofner C, Richter H, Rolke R, Scherens A, Schwarz A, Sommer C, Tronnier V, Uceyler N, Valet M, Wasner G, Treede RD. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): somatosensory abnormalities in 1236 patients with different neuropathic pain syndromes. PAIN 2010;150:439–50.
    1. Mainka T, Malewicz NM, Baron R, Enax-Krumova EK, Treede RD, Maier C. Presence of hyperalgesia predicts analgesic efficacy of topically applied capsaicin 8% in patients with peripheral neuropathic pain. Eur J Pain 2016;20:116–29.
    1. Pfau DB, Krumova EK, Treede RD, Baron R, Toelle T, Birklein F, Eich W, Geber C, Gerhardt A, Weiss T, Magerl W, Maier C. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): reference data for the trunk and application in patients with chronic postherpetic neuralgia. PAIN 2014;155:1002–15.
    1. Reimer M, Hullemann P, Hukauf M, Keller T, Binder A, Gierthmuhlen J, Baron R. Prediction of response to tapentadol in chronic low back pain. Eur J Pain 2017;21:322–33.
    1. Rolke R, Baron R, Maier C, Tolle TR, Treede RD, Beyer A, Binder A, Birbaumer N, Birklein F, Botefur IC, Braune S, Flor H, Huge V, Klug R, Landwehrmeyer GB, Magerl W, Maihofner C, Rolko C, Schaub C, Scherens A, Sprenger T, Valet M, Wasserka B. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): standardized protocol and reference values. PAIN 2006;123:231–43.
    1. Rolke R, Magerl W, Campbell KA, Schalber C, Caspari S, Birklein F, Treede RD. Quantitative sensory testing: a comprehensive protocol for clinical trials. Eur J Pain 2006;10:77–88.
    1. Simpson DM, Schifitto G, Clifford DB, Murphy TK, Durso-de Cruz E, Glue P, Whalen E, Emir B, Scott GN, Freeman R. Pregabalin for painful HIV neuropathy: a randomized, double-blind, placebo-controlled trial. Neurology 2010;74:413–20.
    1. Themistocleous AC, Ramirez JD, Shillo PR, Lees JG, Selvarajah D, Orengo C, Tesfaye S, Rice ASC, Bennett DLH. The Pain in Neuropathy Study (PiNS): a cross-sectional observational study determining the somatosensory phenotype of painful and painless diabetic neuropathy. PAIN 2016;157:1132–45.
    1. Treede RD, Jensen TS, Campbell JN, Cruccu G, Dostrovsky JO, Griffin JW, Hansson P, Hughes R, Nurmikko T, Serra J. Neuropathic pain: redefinition and a grading system for clinical and research purposes. Neurology 2008;70:1630–5.
    1. Truini A, Padua L, Biasiotta A, Caliandro P, Pazzaglia C, Galeotti F, Inghilleri M, Cruccu G. Differential involvement of A-delta and A-beta fibres in neuropathic pain related to carpal tunnel syndrome. PAIN 2009;145:105–9.
    1. Vollert J, Attal N, Baron R, Freynhagen R, Haanpaa M, Hansson P, Jensen TS, Rice ASC, Segerdahl M, Serra J, Sindrup SH, Tolle TR, Treede RD, Maier C. Quantitative sensory testing using DFNS protocol in Europe: an evaluation of heterogeneity across multiple centers in patients with peripheral neuropathic pain and healthy subjects. PAIN 2016;157:750–8.
    1. Vollert J, Mainka T, Baron R, Enax-Krumova EK, Hullemann P, Maier C, Pfau DB, Tolle T, Treede R-D. Quality assurance for Quantitative Sensory Testing laboratories: development and validation of an automated evaluation tool for the analysis of declared healthy samples. PAIN 2015;156:2423–30.
    1. von Hehn CA, Baron R, Woolf CJ. Deconstructing the neuropathic pain phenotype to reveal neural mechanisms. Neuron 2012;73:638–52.
    1. Wasner G, Kleinert A, Binder A, Schattschneider J, Baron R. Postherpetic neuralgia: topical lidocaine is effective in nociceptor-deprived skin. J Neurol 2005;252:677–86.
    1. Westermann A, Krumova EK, Pennekamp W, Horch C, Baron R, Maier C. Different underlying pain mechanisms despite identical pain characteristics: a case report of a patient with spinal cord injury. PAIN 2012;153:1537–40.
    1. Yarnitsky D, Ochoa JL. Differential effect of compression-ischaemia block on warm sensation and heat-induced pain. Brain 1991;114:907–13.
    1. Youden WJ. Index for rating diagnostic tests. Cancer 1950;3:32–5.
    1. Zweig MH, Campbell G. Receiver-operating characteristic (ROC) plots: a fundamental evaluation tool in clinical medicine. Clin Chem 1993;39:561–77.

Source: PubMed

3
Suscribir