Reproducibility of the heat/capsaicin skin sensitization model in healthy volunteers

Laura F Cavallone, Karen Frey, Michael C Montana, Jeremy Joyal, Karen J Regina, Karin L Petersen, Robert W Gereau 4th, Laura F Cavallone, Karen Frey, Michael C Montana, Jeremy Joyal, Karen J Regina, Karin L Petersen, Robert W Gereau 4th

Abstract

Introduction: Heat/capsaicin skin sensitization is a well-characterized human experimental model to induce hyperalgesia and allodynia. Using this model, gabapentin, among other drugs, was shown to significantly reduce cutaneous hyperalgesia compared to placebo. Since the larger thermal probes used in the original studies to produce heat sensitization are now commercially unavailable, we decided to assess whether previous findings could be replicated with a currently available smaller probe (heated area 9 cm(2) versus 12.5-15.7 cm(2)).

Study design and methods: After Institutional Review Board approval, 15 adult healthy volunteers participated in two study sessions, scheduled 1 week apart (Part A). In both sessions, subjects were exposed to the heat/capsaicin cutaneous sensitization model. Areas of hypersensitivity to brush stroke and von Frey (VF) filament stimulation were measured at baseline and after rekindling of skin sensitization. Another group of 15 volunteers was exposed to an identical schedule and set of sensitization procedures, but, in each session, received either gabapentin or placebo (Part B).

Results: Unlike previous reports, a similar reduction of areas of hyperalgesia was observed in all groups/sessions. Fading of areas of hyperalgesia over time was observed in Part A. In Part B, there was no difference in area reduction after gabapentin compared to placebo.

Conclusion: When using smaller thermal probes than originally proposed, modifications of other parameters of sensitization and/or rekindling process may be needed to allow the heat/capsaicin sensitization protocol to be used as initially intended. Standardization and validation of experimental pain models is critical to the advancement of translational pain research.

Keywords: central sensitization; experimental pain model; hyperalgesia; peripheral sensitization.

Figures

Figure 1
Figure 1
Timeline of training and study sessions. The procedures performed in each session are illustrated. (A) screening/training day and (B) study days. Notes: In the control group there was no drug administration, and additional sets of the same measurements described below (M) were taken after RK2 and RK3. In the gabapentin/placebo group, on session 1 and 2, subjects were randomly assigned to receive gabapentin or placebo. M refers to a set of measurements (mapping of area of hyperalgesia/allodynia, heat pain detection threshold, pain during thermal stimulation). Abbreviations: HC, heat/capsaicin sensitization; RK, rekindling.
Figure 2
Figure 2
Enrollment and allocation of participants in the study. Note: *Reasons for exclusions on screening day: 5= HPDT >47°C; 9= unable to achieve sensitization (area); 1= admitted history of drug abuse; 1= showed lesions on forearm at physical examination; 1= body mass index >33. Abbreviation: HPDT, heat pain detection threshold.
Figure 3
Figure 3
Areas of secondary hyperalgesia and allodynia. Notes: Percentage change of area size after treatment in the placebo/gabapentin group and at the same time-point, after the fourth rekindling, in the control group (M2) compared to baseline (M1); data are presented as medians, 1st, and 3rd quartiles; P-values refer to comparison of percentage changes in VF and brush areas between sessions (control group) and between treatments (placebo/gabapentin group). A P-value less than 0.05 was considered statistically significant. Abbreviations: RK, rekindling; VF, von Frey.
Figure 4
Figure 4
HPDTs in the treatment group. Notes: HPDT changes at the first and second set of measurements (M1 and M2, respectively) are compared to presensitization baseline (M0) in the gabapentin and the placebo sessions; data are presented as medians, 1st, and 3rd quartiles; significantly different couples of measurements are indicated: #P=0.03; *P=0.002. (A P-value less than 0.05 was considered statistically significant). In the placebo group HPDTs remained significantly different than baseline at each time point after induction of heat/capsaicin sensitization. Abbreviations: HPDT, heat pain detection thresholds; RK, rekindling.
Figure 5
Figure 5
Brush and von Frey areas: spontaneous fading despite rekindling procedure in control group. Notes: Spontaneous fading of Brush areas (A and B) and von Frey areas (C and D) despite rekindling procedures is shown in the control group. Data are presented as medians, 1st, and 3rd quartiles. Initial von Frey areas in session 2 are significantly smaller than session 1: *P<0.02 (A P-value less than 0.05 was considered statistically significant). Abbreviation: RK, rekindling.
Figure 6
Figure 6
Mean VAS scores for each minute during the 5-minute RK preceding the sets of measurements M1 (RK1) and M2 (RK4). Notes: Mean VAS scores for each minute during the 5-minute RK preceding the sets of measurements M1 (RK1) and M2 (RK4). Data are presented as means and standard deviations. (A) Session 1 and 2 of the control group are compared; (B) Placebo day and gabapentin day of the treatment group are compared. In both groups and both study sessions, painfulness at each minute of the rekindling procedures decreased significantly between the first and fourth rekindling, despite treatment (or no treatment). Part A: both in session 1 and session 2, VAS scores during RK4 were lower than scores during RK1 (session1: P<0.0025 and session 2 P<0.0009). Part B: both on placebo and gabapentin day RK4 (post-drug), VAS scores were significantly lower than RK1 scores (P<0.00005 and P<0.00001, respectively). A P-value less than 0.05 was considered statistically significant. Abbreviations: RK, rekindling procedure; VAS, visual analog scale.

References

    1. Dirks J, Petersen KL, Dahl JB. The heat/capsaicin sensitization model: a methodologic study. J Pain. 2003;4(3):122–128.
    1. Frymoyer AR, Rowbotham MC, Petersen KL. Placebo-controlled comparison of a morphine/dextromethorphan combination with morphine on experimental pain and hyperalgesia in healthy volunteers. J Pain. 2007;8(1):19–25.
    1. Staud R. Evidence for shared pain mechanisms in osteoarthritis, low back pain, and fibromyalgia. Curr Rheumatol Rep. 2011;13(6):513–520.
    1. Steyaert A, De Kock M. Chronic postsurgical pain. Curr Opin Anaesthesiol. 2012;25(5):584–588.
    1. Staud R. Is it all central sensitization? Role of peripheral tissue nociception in chronic musculoskeletal pain. Curr Rheumatol Rep. 2010;12(6):448–454.
    1. Staud R. Brain imaging in fibromyalgia syndrome. Clin Exp Rheumatol. 2011;29(6 Suppl 69):S109–S117.
    1. Staud R. Peripheral pain mechanisms in chronic widespread pain. Best Pract Res Clin Rheumatol. 2011;25(2):155–164.
    1. Graeber MB, Christie MJ. Multiple mechanisms of microglia: a gatekeeper’s contribution to pain states. Exp Neurol. 2012;234(2):255–261.
    1. Gustorff B, Anzenhofer S, Sycha T, Lehr S, Kress HG. The sunburn pain model: the stability of primary and secondary hyperalgesia over 10 hours in a crossover setting. Anesth Analg. 2004;98(1):173–177. table of contents.
    1. Petersen KL, Rowbotham MC. A new human experimental pain model: the heat/capsaicin sensitization model. Neuroreport. 1999;10(7):1511–1516.
    1. Dirks J, Petersen KL, Rowbotham MC, Dahl JB. Effect of systemic adenosine on pain and secondary hyperalgesia associated with the heat/capsaicin sensitization model in healthy volunteers. Reg Anesth Pain Med. 2001;26(5):414–419.
    1. Dirks J, Fabricius P, Petersen KL, Rowbotham MC, Dahl JB. The effect of systemic lidocaine on pain and secondary hyperalgesia associated with the heat/capsaicin sensitization model in healthy volunteers. Anesth Analg. 2000;91(4):967–972.
    1. Petersen KL, Maloney A, Hoke F, Dahl JB, Rowbotham MC. A randomized study of the effect of oral lamotrigine and hydromorphone on pain and hyperalgesia following heat/capsaicin sensitization. J Pain. 2003 Sep;4(7):400–406.
    1. Abrams DI, Jay CA, Shade SB, et al. Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial. Neurology. 2007;68(7):515–521.
    1. Petersen KL, Jones B, Segredo V, Dahl JB, Rowbotham MC. Effect of remifentanil on pain and secondary hyperalgesia associated with the heat – capsaicin sensitization model in healthy volunteers. Anesthesiology. 2001;94(1):15–20.
    1. Dirks J, Petersen KL, Rowbotham MC, Dahl JB. Gabapentin suppresses cutaneous hyperalgesia following heat-capsaicin sensitization. Anesthesiology. 2002;97(1):102–107.
    1. Hughes A, Macleod A, Growcott J, Thomas I. Assessment of the reproducibility of intradermal administration of capsaicin as a model for inducing human pain. Pain. 2002;99(1–2):323–331.
    1. Staahl C, Olesen AE, Andresen T, Arendt-Nielsen L, Drewes AM. Assessing efficacy of non-opioid analgesics in experimental pain models in healthy volunteers: an updated review. Br J Clin Pharmacol. 2009;68(3):322–341.
    1. Oertel BG, Lotsch J. Clinical pharmacology of analgesics assessed with human experimental pain models: bridging basic and clinical research. Br J Pharmacol. 2013;168(3):534–553.
    1. Yucel A, Miyazawa A, Andersen OK, Arendt-Nielsen L. The effect of heat conditioning of the primary area before and after induction of hyperalgesia by topical/intradermal capsaicin or by controlled heat injury. Somatosens Mot Res. 2001;18(4):295–302.
    1. Pedersen JL, Kehlet H. Hyperalgesia in a human model of acute inflammatory pain: a methodological study. Pain. 1998;74(2–3):139–151.
    1. Wang H, Bolognese J, Calder N, et al. Effect of morphine and pregabalin compared with diphenhydramine hydrochloride and placebo on hyperalgesia and allodynia induced by intradermal capsaicin in healthy male subjects. J Pain. 2008;9(12):1088–1095.
    1. Modir JG, Wallace MS. Human Experimental Pain Models 3: Heat/CapsaicinSensitization and Intradermal Capsaicin Models. In: Szallasi A, editor. Analgesia: Methods and Protocols, Methods in Molecular Biology. Vol. 617. New York: Humana Press; 2010. pp. 169–174.
    1. Petersen K, Schmelz M. Human Pain Models: Virtues and Limitations; Paper presented at: IASP Refresher Course presented at the 12th World Congress on Pain; August 17–22, 2008; Glasgow, Scotland, UK.
    1. LaMotte RH, Lundberg LE, Torebjörk HE. Pain, hyperalgesia and activity in nociceptive C units in humans after intradermal injection of capsaicin. J Physiol. 1992;448:749–764.
    1. Koltzenburg M, Torebjörk HE, Wahren LK. Nociceptor modulated central sensitization causes mechanical hyperalgesia in acute chemogenic and chronic neuropathic pain. Brain. 1994;117(Pt 3):579–591.
    1. Hood DD, Curry R, Eisenach JC. Intravenous remifentanil produces withdrawal hyperalgesia in volunteers with capsaicin-induced hyperalgesia. Anesth Analg. 2003;97(3):810–815.
    1. Iannetti GD, Zambreanu L, Wise RG, et al. Pharmacological modulation of pain-related brain activity during normal and central sensitization states in humans. Proc Natl Acad Sci U S A. 2005;102(50):18195–18200.
    1. Campbell CM, Bounds SC, Simango MB, et al. Self-reported sleep duration associated with distraction analgesia, hyperemia, and secondary hyperalgesia in the heat-capsaicin nociceptive model. Eur J Pain. 2011;15(6):561–567.
    1. Eisenach JC, Curry R, Tong C, Houle TT, Yaksh TL. Effects of intrathecal ketorolac on human experimental pain. Anesthesiology. 2010;112(5):1216–1224.
    1. LaMotte RH, Shain CN, Simone DA, Tsai EF. Neurogenic hyperalgesia: psychophysical studies of underlying mechanisms. J Neurophysiol. 1991;66(1):190–211.
    1. Moiniche S, Dahl JB, Kehlet H. Time course of primary and secondary hyperalgesia after heat injury to the skin. Br J Anaesth. 1993;71(2):201–205.
    1. Pedersen JL, Kehlet H. Secondary hyperalgesia to heat stimuli after burn injury in man. Pain. 1998;76(3):377–384.
    1. Cervero F, Gilbert R, Hammond RG, Tanner J. Development of secondary hyperalgesia following non-painful thermal stimulation of the skin: a psychophysical study in man. Pain. 1993;54(2):181–189.
    1. Andersen OK, Jensen LM, Brennum J, Arendt-Nielsen L. Evidence for central summation of C and A delta nociceptive activity in man. Pain. 1994;59(2):273–280.
    1. Gustorff B, Hauer D, Thaler J, Seis A, Draxler J. Antihyperalgesic efficacy of 5% lidocaine medicated plaster in capsaicin and sunburn pain models – two randomized, double-blinded, placebo-controlled crossover trials in healthy volunteers. Expert Opin Pharmacother. 2011;12(18):2781–2790.
    1. Klein T, Magerl W, Hanschmann A, Althaus M, Treede RD. Antihyperalgesic and analgesic properties of the N-methyl-D-aspartate (NMDA) receptor antagonist neramexane in a human surrogate model of neurogenic hyperalgesia. Eur J Pain. 2008;12(1):17–29.
    1. O’Neill J, Brock C, Olesen AE, Andresen T, Nilsson M, Dickenson AH. Unravelling the mystery of capsaicin: a tool to understand and treat pain. Pharmacol Rev. 2012;64(4):939–971.
    1. Torebjörk HE, Lundberg LE, LaMotte RH. Central changes in processing of mechanoreceptive input in capsaicin-induced secondary hyperalgesia in humans. J Physiol. 1992;448:765–780.
    1. Jensen MT, Petersen KL. Gender differences in pain and secondary hyperalgesia after heat/capsaicin sensitization in healthy volunteers. J Pain. 2006;7(3):211–217.
    1. Wallace MS, Schulteis G. Effect of chronic oral gabapentin on capsaicin-induced pain and hyperalgesia: a double-blind, placebo-controlled, crossover study. Clin J Pain. 2008;24(6):544–549.
    1. Gottrup H, Juhl G, Kristensen AD, et al. Chronic oral gabapentin reduces elements of central sensitization in human experimental hyperalgesia. Anesthesiology. 2004;101(6):1400–1408.

Source: PubMed

3
구독하다