Duloxetine, a Balanced Serotonin-Norepinephrine Reuptake Inhibitor, Improves Painful Chemotherapy-Induced Peripheral Neuropathy by Inhibiting Activation of p38 MAPK and NF-κB

Jing Meng, Qiuyan Zhang, Chao Yang, Lu Xiao, Zhenzhen Xue, Jing Zhu, Jing Meng, Qiuyan Zhang, Chao Yang, Lu Xiao, Zhenzhen Xue, Jing Zhu

Abstract

Chemotherapy-induced peripheral neuropathy (CIPN) is a severe, toxic side effect that frequently occurs in anticancer treatment and may result in discontinuation of treatment as well as a serious reduction in life quality. The CIPN incidence rate is as high as 85-90%. Unfortunately, there is currently no standard evidence-based CIPN treatment. In several clinical trials, it has been reported that duloxetine can improve CIPN pain induced by oxaliplatin (OXA) and paclitaxel (PTX); thus, The American Society of Clinical Oncology (ASCO) recommends duloxetine as the only potential treatment for CIPN. However, this guidance lacks the support of sufficient evidence. Our study shows that duloxetine markedly reduces neuropathic pain evoked by OXA or PTX. Duloxetine acts by inhibiting the activation of p38 phosphorylation, thus preventing the activation and nuclear translocation of the NF-κB transcription factor, reducing the inflammatory response and inhibiting nerve injury by regulating nerve growth factor (NGF). Furthermore, in this study, it is shown that duloxetine does not affect the antitumor activity of OXA or PTX. This study not only provides biological evidence to support the use of duloxetine as the first standard CIPN drug but will also lead to potential new targets for CIPN drug development.

Keywords: chemotherapy-induced peripheral neewopathy (CIPN); dorsal root ganglia (DRG); duloxetine; eripheral neuropathic pain; oxaliplatin (OXA); paclitaxel (PTX).

Figures

FIGURE 1
FIGURE 1
Effect of duloxetine on axonal injury caused by oxaliplatin or paclitaxel (A) Primary rat DRG neurons were grown and axons were extended for 24 h. Neurons were then treated with OXA (3 μM) or duloxetine (3 μM) for 48 h. Cells were then subjected to immunofluorescence staining, and axon length measurements were performed (∗∗∗p < 0.001 vs. control; ##p < 0.01; ###p < 0.001 vs. oxaliplatin alone). (B) Primary rat DRG neuronal cells were grown and axons were extended for 24 h. Neurons were then treated with PTX (300 nM) or duloxetine (300 nM) for 24 h. Cells were then subjected to immunofluorescence staining, and axon length measurements were performed (∗∗∗p < 0.001 vs. control; ##p < 0.01; ###p < 0.001 vs. paclitaxel alone). The results are expressed as the mean ± SEM (n = 10).
FIGURE 2
FIGURE 2
Effect of duloxetine on DRG neuronal apoptosis through flow cytometry and TUNEL assay. After growing in culture for 24 h, DRG neuronal cells were exposed to OXA (3 μM) with (or without) duloxetine (3 μM) for another 48 h (A,C) or exposed to PTX (300 nM) with (or without) duloxetine (300 nM) for another 24 h (B,D). (A,B):Cells were then double-stained with annexin V-FITC/PI. Annexin V-FITC fluorescence was measured with the FL1 channel, and PI fluorescence was measured with the FL3 channel. Representative pictures are from one of three independent experiments with similar results. (C,D) The cell death of DRG was examined by TUNEL assay. (∗p < 0.05, ∗∗∗p < 0.001 compared with control; #p < 0.05, ##p < 0.01, ###p < 0.001 compared with model). The data are presented as mean ± SEM. Representative pictures are from one of three independent experiments with similar results (×200).
FIGURE 3
FIGURE 3
Effect of duloxetine on the anticancer activity of OXA or PTX in vitro. (A,B) When HT-29 or SUM-159 cancer cells were grown in medium, OXA or PTX reduced their cell viability by 20–80%. (C,D) Various concentrations of duloxetine together with OXA or PTX showed no significant changes in cell viability when compared to treatments with OXA or PTX alone. (∗p < 0.05, ∗∗p < 0.01,∗∗∗p < 0.001 vs. control). All the data are the mean ± SEM for each experiment (n = 4).
FIGURE 4
FIGURE 4
Partial prevention by duloxetine against OXA or PTX-induced peripheral neuropathy in ICR mice. (A) The effect of duloxetine on the heat withdrawal latency in mice treated with vehicle or chemotherapeutic drugs (OXA and PTX) was evaluated. (B) The effect of duloxetine on the cold threshold in mice treated with vehicle or chemotherapeutic drugs (OXA and PTX) was determined. (C) The effect of duloxetine on the mechanical withdrawal threshold in mice treated with vehicle or chemotherapeutic drugs (OXA and PTX) was assessed. (∗p < 0.05, ∗∗p < 0.01,∗∗∗p < 0.001 vs. control; #p < 0.05,##p < 0.01, ###p < 0.001 vs. OXA; Δp < 0.05, ΔΔp < 0.01 vs. PTX). The data are presented as the mean ± SEM (n = 5–8).
FIGURE 5
FIGURE 5
Effect of duloxetine on IENF retraction induced by OXA or PTX. Paw biopsies were obtained from the hind paws of mice when behavior tests were finished. Tissues were fixed and stained with antibodies (PGP9.5) for IENFs. (A) Representative images from six groups are shown. (B) IENF density = number of nerve fibers crossing the basement membrane/length of the basement membrane (mm). (Magnification × 400. ∗p < 0.05,∗∗p < 0.01,∗∗∗p < 0.001). The data are presented as the mean ± SEM (n = 5).
FIGURE 6
FIGURE 6
Effect of duloxetine on phosphorylation of p38 MAPK and ERK1/2 expression in the neuropathic mouse DRG following oxaliplatin treatment. (A) Duloxetine significantly decreased the expression of NF-κB protein in the DRG of OXA-treated mice. (B) Duloxetine significantly decreased the expression of pp38 protein in the DRG of OXA-treated mice. (C) The ratio of pERK1/2 to ERK1/2 expression was not changed significantly following oxaliplatin treatment and duloxetine did not modify p-ERK1/2 expression. (∗∗p < 0.01 vs. control, ##p < 0.01, ###p < 0.001 vs. OXA). The data are presented as the mean ± SEM (n = 4).
FIGURE 7
FIGURE 7
Effect of duloxetine on NF-κB, phosphorylation of p38 MAPK and ERK1/2 expression in the neuropathic mouse DRG following PTX treatment. (A) Duloxetine significantly decreased the expression of NF-κB protein in the DRG of PTX-treated mice. (B) Duloxetine significantly decreased the expression of pp38 protein in the DRG of PTX-treated mice. (C) The ratio of pERK1/2 to ERK1/2 expression was significantly increased after PTX treatment, but duloxetine did not modify the p-ERK1/2 expression induced by PTX. (∗∗p < 0.01 vs. control, ∗∗∗p < 0.001 vs. control, #p < 0.05, ##p < 0.01, ###p < 0.001 vs. PTX). The data are presented as the mean ± SEM (n = 4).
FIGURE 8
FIGURE 8
Effect of oxaliplatin and duloxetine on the expression of nuclear NF-κB in DRG neuronal cells. DRG neuronal cells were treated with (or without) OXA (3 μM) and duloxetine (3 μM) for 48 h and were double-stained with DAPI and NF-κB. The fluorescence intensity was observed using fluorescence microscopy (×200). (∗∗∗p < 0.001 vs. control; ###p < 0.001 vs. OXA).
FIGURE 9
FIGURE 9
Effect of paclitaxel and duloxetine on the expression of nuclear NF-κB in DRG neuronal cells. DRG neuronal cells were treated with (or without) PTX (300 nM) and duloxetine (300 nM) for 24 h and were double-stained with DAPI and NF-κB. The fluorescence intensity was observed using fluorescence microscopy (×200). (∗∗∗p < 0.001 vs. control; ###p < 0.001 vs. PTX).
FIGURE 10
FIGURE 10
Transcript levels of the proinflammatory cytokines IL-1β, IL-6 and TNF-α in the mouse DRG. Total mRNA was extracted from mouse DRG tissues. qPCR was performed and a comparison is shown between different groups after saline or 28 days of drug treatment. (A) IL-1β expression was significantly decreased after OXA treatment, but duloxetine did not affect this decrease. (B) Duloxetine significantly decreased the expression of IL-6 mRNA in the DRG of OXA-treated mice. (C) Duloxetine significantly decreased the expression of TNF-α mRNA in the DRG of OXA-treated mice. (∗∗p < 0.01, ∗∗∗p < 0.001 vs. control; #p < 0.05 vs. OXA, ###p < 0.001 vs. OXA). The data are presented as the means ± SEM (n = 5).
FIGURE 11
FIGURE 11
Transcript levels of the proinflammatory cytokines IL-1β, IL-6 and TNF-α in the mouse DRG. Total mRNA was extracted from mouse DRG tissues. qPCR was performed and a comparison is shown between different groups after saline or 28 days of drug treatment. (A) IL-1β expression was significantly decreased after PTX treatment, but duloxetine did not alter this decrease. (B) Duloxetine significantly decreased the expression of IL-6 mRNA in the DRG of PTX-treated mice. (C) Duloxetine significantly decreased the expression of TNF-α mRNA in the DRG of PTX-treated mice. (∗∗p < 0.01, ∗∗∗p < 0.001 vs. control; ##p < 0.01, ###p < 0.001 vs. PTX). The data are presented as the mean ± SEM (n = 5).

References

    1. Apfel S. C. (2002). Nerve growth factor for the treatment of diabetic neuropathy: what went wrong, what went right, and what does the future hold? Int. Rev. Neurobiol. 50 393–413. 10.1016/S0074-7742(02)50083-0
    1. Apfel S. C., Arezzo J. C., Brownlee M., Federoff H., Kessler J. A. (1994). Nerve growth factor administration protects against experimental diabetic sensory neuropathy. Brain Res. 634 7–12. 10.1016/0006-8993(94)90252-6
    1. Areti A., Komirishetty P., Akuthota M., Malik R. A., Kumar A. (2017). Melatonin prevents mitochondrial dysfunction and promotes neuroprotection by inducing autophagy during oxaliplatin-evoked peripheral neuropathy. J. Pineal Res. 62:12393. 10.1111/jpi.12393
    1. Austin P. J., Moalem-Taylor G. (2010). The neuro-immune balance in neuropathic pain: involvement of inflammatory immune cells, immune-like glial cells and cytokines. J. Neuroimmunl. 229 26–50. 10.1016/j.jneuroim.2010.08.013
    1. Bennett G. J., Doyle T., Salvemini D. (2014). Mitotoxicity in distal symmetrical sensory peripheral neuropathies. Nat. Rev. Neurol. 10 326–336. 10.1038/nrneurol.2014.77
    1. Boyette-Davis J., Dougherty P. M. (2011). Protection against oxaliplatin-induced mechanical hyperalgesia and intraepidermal nerve fiber loss by minocycline. Exp. Neurol. 229 353–357. 10.1016/j.expneurol.2011.02.019
    1. Boyette-Davis J., Xin W., Zhang H., Dougherty P. M. (2011). Intraepidermal nerve fiber loss corresponds to the development of taxol-induced hyperalgesia and can be prevented by treatment with minocycline. Pain 152 308–313. 10.1016/j.pain.2010.10.030
    1. Btesh J., Fischer M. J. M., Stott K., McNaughton P. A. (2013). Mapping the binding site of TRPV1 on AKAP79: implications for inflammatory hyperalgesia. J. Neurosci. 33 9184–9193. 10.1523/JNEUROSCI.4991-12.2013
    1. Cetinkaya-Fisgin A., Joo M. G., Ping X., Thakor N. V., Ozturk C., Hoke A., et al. (2016). Identification of fluocinolone acetonide to prevent paclitaxel-induced peripheral neuropathy. J. Peripher. Nerv. Syst. 21 128–133. 10.1111/jns.12172
    1. Chen H., Wang Q., Shi D., Yao D., Zhang L., Xiong J., et al. (2016). Celecoxib alleviates oxaliplatin-induced hyperalgesia through inhibition of spinal ERK1/2 signaling. J. Toxicol. Pathol. 29 253–259. 10.1293/tox.2016-0032
    1. Chen W., Mi R., Haughey N., Oz M., Hoke A. (2007). Immortalization and characterization of a nociceptive dorsal root ganglion sensory neuronal line. J. Peripher. Nerv. Syst. 12 121–130. 10.1111/j.1529-8027.2007.00131.x
    1. Connor B., Dragunow M. (1998). The role of neuronal growth factors in neurodegenerative disorders of the human brain. Brain Res. Brain Res. Rev. 27 1–39. 10.1016/S0165-0173(98)00004-6
    1. Doyle T., Chen Z., Muscoli C., Bryant L., Esposito E., Cuzzocrea S., et al. (2012). Targeting the overproduction of peroxynitrite for the prevention and reversal of paclitaxel-induced neuropathic pain. J. Neurosci. 32 6149–6160. 10.1523/JNEUROSCI.6343-11.2012
    1. Ewertz M., Qvortrup C., Eckhoff L. (2015). Chemotherapy-induced peripheral neuropathy in patients treated with taxanes and platinum derivatives. Acta Oncol. 54 587–591. 10.3109/0284186X.2014.995775
    1. Hershman D. L., Lacchetti C., Dworkin R. H., Lavoie S. E., Bleeker J., Cavaletti G., et al. (2014). Prevention and management of chemotherapy-induced peripheral neuropathy in survivors of adult cancers: american society of clinical oncology clinical practice guideline. J. Clin. Oncol. 32 1941–1967. 10.1200/JCO.2013.54.0914
    1. Hirayama Y., Ishitani K., Sato Y., Iyama S., Takada K., Murase K., et al. (2015). Effect of duloxetine in Japanese patients with chemotherapy-induced peripheral neuropathy: a pilot randomized trial. Int. J. Clin. Oncol. 20 866–871. 10.1007/s10147-015-0810-y
    1. Hopkins H. L., Duggett N. A., Flatters S. J. (2016). Chemotherapy-induced painful neuropathy: pain-like behaviours in rodent models and their response to commonly used analgesics. Curr. Opin. Support. Palliat. Care 10 119–128. 10.1097/SPC.0000000000000204
    1. Janes K., Esposito E., Doyle T., Cuzzocrea S., Tosh D. K., Jacobson K. A., et al. (2014). A3 adenosine receptor agonist prevents the development of paclitaxel-induced neuropathic pain by modulating spinal glial-restricted redox-dependent signaling pathways. PAIN 155 2560–2567. 10.1016/j.pain.2014.09.016
    1. Kim C., Lee J., Kim W., Li D., Kim Y., Lee K., et al. (2016). The suppressive effects of cinnamomi cortex and its phytocompound coumarin on oxaliplatin-induced neuropathic cold allodynia in rats. Molecules 21:1253. 10.3390/molecules21091253
    1. Ko M., Hu M., Hsieh Y., Lan C., Tseng T. (2014). Peptidergic intraepidermal nerve fibers in the skin contribute to the neuropathic pain in paclitaxel-induced peripheral neuropathy. Neuropeptides 48 109–117. 10.1016/j.npep.2014.02.001
    1. Krukowski K., Nijboer C. H., Huo X., Kavelaars A., Heijnen C. J. (2015). Prevention of chemotherapy-induced peripheral neuropathy by the small-molecule inhibitor pifithrin-mu. Pain 156 2184–2192. 10.1097/j.pain.0000000000000290
    1. Lewin G. R., Lechner S. G., Smith E. S. (2014). Nerve growth factor and nociception: from experimental embryology to new analgesic therapy. Handb. Exp. Pharmacol. 220 251–282. 10.1007/978-3-642-45106-5_10
    1. Li X., Wang J., Wang Z., Dong C., Dong X., Jing Y., et al. (2008). Tumor necrosis factor-α of red nucleus involved in the development of neuropathic allodynia. Brain Res. Bull. 77 233–236. 10.1016/j.brainresbull.2008.08.025
    1. Li Y., Zhang H., Kosturakis A. K., Cassidy R. M., Zhang H., Kennamer-Chapman R. M., et al. (2015). MAPK signaling downstream to TLR4 contributes to paclitaxel-induced peripheral neuropathy. Brain Behav. Immun. 49 255–266. 10.1016/j.bbi.2015.06.003
    1. Makker P. G. S., Duffy S. S., Lees J. G., Perera C. J., Tonkin R. S., Butovsky O., et al. (2017). Characterisation of immune and neuroinflammatory changes associated with chemotherapy-induced peripheral neuropathy. PLoS One 12:170814. 10.1371/journal.pone.0170814
    1. Okuma K., Shiraishi K., Kanai Y., Nakagawa K. (2016). Improvement in quality of life by using duloxetine for chemotherapy-induced peripheral neuropathy (CIPN): a case report. Support. Care Cancer 24 4483–4485. 10.1007/s00520-016-3349-1
    1. Park S. B., Goldstein D., Krishnan A. V., Lin C. S., Friedlander M. L., Cassidy J., et al. (2013). Chemotherapy-induced peripheral neurotoxicity: a critical analysis. CA Cancer J. Clin. 63 419–437. 10.3322/caac.21204
    1. Piccolo J., Kolesar J. M. (2013). Prevention and treatment of chemotherapy-induced peripheral neuropathy. Am. J. Health Syst. Pharm. 71 19–25. 10.2146/ajhp130126
    1. Russe O. Q., Möser C. V., Kynast K. L., King T. S., Stephan H., Geisslinger G., et al. (2013). Activation of the AMP-activated protein kinase reduces inflammatory nociception. J. Pain 14 1330–1340. 10.1016/j.jpain.2013.05.012
    1. Smith E. M., Pang H., Cirrincione C., Fleishman S., Paskett E. D., Ahles T., et al. (2013). Effect of duloxetine on pain, function, and quality of life among patients with chemotherapy-induced painful peripheral neuropathy: a randomized clinical trial. JAMA 309 1359–1367. 10.1001/jama.2013.2813
    1. Smith E. M., Pang H., Ye C., Cirrincione C., Fleishman S., Paskett E. D., et al. (2015). Predictors of duloxetine response in patients with oxaliplatin-induced painful chemotherapy-induced peripheral neuropathy (CIPN): a secondary analysis of randomised controlled trial - CALGB/alliance 170601. Eur. J. Cancer Care 26:12421. 10.1111/ecc.12421
    1. Ta L. E., Espeset L., Podratz J., Windebank A. J. (2006). Neurotoxicity of oxaliplatin and cisplatin for dorsal root ganglion neurons correlates with platinum-DNA binding. Neurotoxicology 27 992–1002. 10.1016/j.neuro.2006.04.010
    1. Taillibert S., Le R. E., Chamberlain M. C. (2016). Chemotherapy-related neurotoxicity. Curr. Neurol. Neurosci. Rep. 16:81. 10.1007/s11910-016-0686-x
    1. Vallejo R., Tilley D. M., Vogel L., Benyamin R. (2010). The role of glia and the immune system in the development and maintenance of neuropathic pain. Pain Pract. 10 167–184. 10.1111/j.1533-2500.2010.00367.x
    1. Wallace V. C. J., Blackbeard J., Pheby T., Segerdahl A. R., Davies M., Hasnie F., et al. (2007). Pharmacological, behavioural and mechanistic analysis of HIV-1 gp120 induced painful neuropathy. Pain 133 47–63. 10.1016/j.pain.2007.02.015
    1. Wang Z., Wang J., Li X., Yuan Y., Fan G. (2008). Interleukin-1 beta of red nucleus involved in the development of allodynia in spared nerve injury rats. Exp. Brain Res. 188 379–384. 10.1007/s00221-008-1365-1
    1. Xiao W. H., Zheng H., Zheng F. Y., Nuydens R., Meert T. F., Bennett G. J. (2011). Mitochondrial abnormality in sensory, but not motor, axons in paclitaxel-evoked painful peripheral neuropathy in the rat. Neuroscience 199 461–469. 10.1016/j.neuroscience.2011.10.010
    1. Yeo J., Yoon S., Kim S., Oh S., Lee J., Beitz A. J., et al. (2016). Clonidine, an alpha-2 adrenoceptor agonist relieves mechanical allodynia in oxaliplatin-induced neuropathic mice; potentiation by spinal p38 MAPK inhibition without motor dysfunction and hypotension. Int. J. Cancer 138 2466–2476. 10.1002/ijc.29980
    1. Zhang H., Boyette-Davis J. A., Kosturakis A. K., Li Y., Yoon S., Walters E. T., et al. (2013). Induction of monocyte chemoattractant protein-1 (MCP-1) and its receptor CCR2 in primary sensory neurons contributes to paclitaxel-induced peripheral neuropathy. J. Pain 14 1031–1044. 10.1016/j.jpain.2013.03.012
    1. Zhang H., Li Y., de Carvalho-Barbosa M., Kavelaars A., Heijnen C. J., Albrecht P. J., et al. (2016). Dorsal root ganglion infiltration by macrophages contributes to paclitaxel chemotherapy-induced peripheral neuropathy. J. Pain 17 775–786. 10.1016/j.jpain.2016.02.011
    1. Zhu J., Chen W., Mi R., Zhou C., Reed N., Höke A. (2013). Ethoxyquin prevents chemotherapy-induced neurotoxicity via Hsp90 modulation. Ann. Neurol. 74 893–904. 10.1002/ana.24004
    1. Zimmermann M. (1983). Ethical guidelines for investigations of experimental pain in conscious animals. Pain 16 109–110. 10.1016/0304-3959(83)90201-4

Source: PubMed

3
Suscribir