Loxapine for Treatment of Patients With Refractory, Chemotherapy-Induced Neuropathic Pain: A Prematurely Terminated Pilot Study Showing Efficacy But Limited Tolerability

Sven Schmiedl, David Peters, Oliver Schmalz, Anke Mielke, Tanja Rossmanith, Shirin Diop, Martina Piefke, Petra Thürmann, Achim Schmidtko, Sven Schmiedl, David Peters, Oliver Schmalz, Anke Mielke, Tanja Rossmanith, Shirin Diop, Martina Piefke, Petra Thürmann, Achim Schmidtko

Abstract

Neuropathic pain is a debilitating and commonly treatment-refractory condition requiring novel therapeutic options. Accumulating preclinical studies indicate that the potassium channel Slack (KNa1.1) contributes to the processing of neuropathic pain, and that Slack activators, when injected into mice, ameliorate pain-related hypersensitivity. However, whether Slack activation might reduce neuropathic pain in humans remains elusive. Here, we evaluated the tolerability and analgesic efficacy of loxapine, a first-generation antipsychotic drug and Slack activator, in neuropathic pain patients. We aimed to treat 12 patients with chronic chemotherapy-induced, treatment-refractory neuropathic pain (pain severity ≥ 4 units on an 11-point numerical rating scale) in a monocentric, open label, proof-of-principle study. Patients received loxapine orally as add-on analgesic in a dose-escalating manner (four treatment episodes for 14 days, daily dose: 20, 30, 40, or 60 mg loxapine) depending on tolerability and analgesic efficacy. Patient-reported outcomes of pain intensity and/or relief were recorded daily. After enrolling four patients, this study was prematurely terminated due to adverse events typically occurring with first-generation antipsychotic drugs that were reported by all patients. In two patients receiving loxapine for at least two treatment episodes, a clinically relevant analgesic effect was found at a daily dose of 20-30 mg of loxapine. Another two patients tolerated loxapine only for a few days. Together, our data further support the hypothesis that Slack activation might be a novel strategy for neuropathic pain therapy. However, loxapine is no valid treatment option for painful polyneuropathy due to profound dopamine and histamine receptor-related side effects. Clinical Trial Registration: www.ClinicalTrials.gov, identifier NCT02820519.

Keywords: Slack channel; analgesia; loxapine; neuropathic pain; tolerability and safety.

Figures

Figure 1
Figure 1
Study flow chart.
Figure 2
Figure 2
Loxapine dose per day and time course of neuropathic pain as determined by an 11-point numerical rating scale (NRS) for subject #1 (A) and #2 (B).

References

    1. Alles S. R. A., Smith P. A. (2018). Etiology and pharmacology of neuropathic pain. Pharmacol. Rev. 70 (2), 315–347. 10.1124/pr.117.014399
    1. Attal N., Lanteri-Minet M., Laurent B., Fermanian J., Bouhassira D. (2011). The specific disease burden of neuropathic pain: Results of a French nationwide survey. Pain 152 (12), 2836–2843. 10.1016/j.pain.2011.09.014
    1. Azermai M., Petrovic M., Engelborghs S., Elseviers M. M., Van der Mussele S., Debruyne H., et al. (2013). The effects of abrupt antipsychotic discontinuation in cognitively impaired older persons: a pilot study. Aging Ment. Health 17 (1), 125–132. 10.1080/13607863.2012.717255
    1. Baron R., Binder A., Wasner G. (2010). Neuropathic pain: diagnosis, pathophysiological mechanisms, and treatment. Lancet Neurol. 9 (8), 807–819. 10.1016/S1474-4422(10)70143-5
    1. Baron R., Treede R. D., Birklein F., Cegla T., Freynhagen R., Heskamp M. L., et al. (2017). Treatment of painful radiculopathies with capsaicin 8% cutaneous patch. Curr. Med. Res. Opin. 33 (8), 1401–1411. 10.1080/03007995.2017.1322569
    1. Biton B., Sethuramanujam S., Picchione K., Bhattacharjee A., Khessibi N., Chesney F., et al. (2011). The antipsychotic drug loxapine is an opener of the Na+-activated potassium channel Slack (slo2.2). J. Pharmacol. Exp. Ther. 340 (3), 706–715. 10.1124/jpet.111.184622
    1. Chakrabarti A., Bagnall A., Chue P., Fenton M., Palaniswamy V., Wong W., et al. (2007). Loxapine for schizophrenia. Cochrane Database Syst. Rev. 2007 (4), CD001943. 10.1002/14651858.CD001943.pub2
    1. Declercq T., Petrovic M., Azermai M., Vander Stichele R., De Sutter A. I., van Driel M. L., et al. (2013). Withdrawal versus continuation of chronic antipsychotic drugs for behavioural and psychological symptoms in older people with dementia. Cochrane Database Syst. Rev. 2013 (3), CD007726. 10.1002/14651858.CD007726.pub2
    1. Doth A. H., Hansson P. T., Jensen M. P., Taylor R. S. (2010). The burden of neuropathic pain: a systematic review and meta-analysis of health utilities. Pain 149 (2), 338–344. 10.1016/j.pain.2010.02.034
    1. Finnerup N. B., Attal N., Haroutounian S., McNicol E., Baron R., Dworkin R. H., et al. (2015). Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet. Neurol 14 (2), 162–173. 10.1016/S1474-4422(14)70251-0
    1. Frampton C. L., Hughes-Webb P. (2011). The measurement of pain. Clin. Oncol. (R Coll. Radiol.) 23 (6), 381–386. 10.1016/j.clon.2011.04.008
    1. Freynhagen R., Baron R., Gockel U., Tolle T. R. (2006. a). painDETECT: a new screening questionnaire to identify neuropathic components in patients with back pain. Curr. Med. Res. Opin. 22 (10), 1911–1920. 10.1185/030079906X132488
    1. Freynhagen R., Busche P., Konrad C., Balkenohl M. (2006. b). Effectiveness and time to onset of pregabalin in patients with neuropathic pain. Schmerz 20285–20288 (4), 290–282. 10.1007/s00482-005-0449-0
    1. Haanpää M., Attal N., Backonja M., Baron R., Bennett M., Bouhassira D., et al. (2011). NeuPSIG guidelines on neuropathic pain assessment. Pain 152 (1), 14–27. 10.1016/j.pain.2010.07.031
    1. Heel R. C., Brogden R. N., Speight T. M., Avery G. S. (1978). Loxapine: a review of its pharmacological properties and therapeutic efficacy as an antipsychotic agent. Drugs 15 (3), 198–217. 10.2165/00003495-197815030-00002
    1. Hermann-Lingen C., Buss U., Snaith R. P. (2011). Hospital Anxiety and Depression Scale - Deutsche Version. Göttingen: Hogrefe.
    1. Jensen N. H., Rodriguiz R. M., Caron M. G., Wetsel W. C., Rothman R. B., Roth B. L. (2008). N-desalkylquetiapine, a potent norepinephrine reuptake inhibitor and partial 5-HT1A agonist, as a putative mediator of quetiapine’s antidepressant activity. Neuropsychopharmacology 33 (10), 2303–2312. 10.1038/sj.npp.1301646
    1. Jensen T. S., Baron R., Haanpaa M., Kalso E., Loeser J. D., Rice A. S., et al. (2011). A new definition of neuropathic pain. Pain 152 (10), 2204–2205. 10.1016/j.pain.2011.06.017
    1. Jimenez X. F., Sundararajan T., Covington E. C. (2018). A systematic review of atypical antipsychotics in chronic pain management: olanzapine demonstrates potential in central sensitization, fibromyalgia, and headache/migraine. Clin. J. Pain 34 (6), 585–591. 10.1097/AJP.0000000000000567
    1. Kaczmarek L. K. (2013). Slack, slick and sodium-activated potassium channels. ISRN Neurosci. 2013, 1–14. 10.1155/2013/354262
    1. Knezevic N. N., Yekkirala A., Yaksh T. L. (2017). Basic/translational development of forthcoming opioid- and nonopioid-targeted pain therapeutics. Anesth. Analg. 125 (5), 1714–1732. 10.1213/ANE.0000000000002442
    1. Lampela P., Paajanen T., Hartikainen S., Huupponen R. (2015). Central anticholinergic adverse effects and their measurement. Drugs Aging 32 (12), 963–974. 10.1007/s40266-015-0321-6
    1. Lu R., Bausch A. E., Kallenborn-Gerhardt W., Stoetzer C., Debruin N., Ruth P., et al. (2015). Slack channels expressed in sensory neurons control neuropathic pain in mice. J. Neurosci. 35 (3), 1125–1135. 10.1523/JNEUROSCI.2423-14.2015
    1. Morfeld M., Kirchberger I., Bullinger M. (2011). SF-36: Fragebogen zum Gesundheitszustand. Göttingen: Hogrefe.
    1. Nightingale S. (2012). The neuropathic pain market. Nat. Rev. Drug Discov. 11 (2), 101–102. 10.1038/nrd3624
    1. Popovic D., Nuss P., Vieta E. (2015). Revisiting loxapine: a systematic review. Ann. Gen. Psychiatry 14, 15. 10.1186/s12991-015-0053-3
    1. Roth B. L., Lopez E., Patel S., Kroeze W. K. (2000). The multiplicity of serotonin receptors: uselessly diverse molecules or an embarrassment of riches? Neuroscientist 6, 252–262. 10.1177/107385840000600408
    1. Saif M. W., Syrigos K., Kaley K., Isufi I. (2010). Role of pregabalin in treatment of oxaliplatin-induced sensory neuropathy. Anticancer Res. 30 (7), 2927–2933.
    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 (13), 1359–1367. 10.1001/jama.2013.2813
    1. Staff N. P., Grisold A., Grisold W., Windebank A. J. (2017). Chemotherapy-induced peripheral neuropathy: a current review. Ann. Neurol. 81 (6), 772–781. 10.1002/ana.24951
    1. Starobova H., Vetter I. (2017). Pathophysiology of chemotherapy-induced peripheral neuropathy. Front. Mol. Neurosci. 10, 174. 10.3389/fnmol.2017.00174
    1. Tsantoulas C., McMahon S. B. (2014). Opening paths to novel analgesics: the role of potassium channels in chronic pain. Trends Neurosci. 37 (3), 146–158. 10.1016/j.tins.2013.12.002
    1. van Hecke O., Torrance N., Smith B. H. (2013). Chronic pain epidemiology and its clinical relevance. Br. J. Anaesth. 111 (1), 13–18. 10.1093/bja/aet123
    1. Vanelle J. M., Olie J. P., Levy-Soussan P. (1994). New antipsychotics in schizophrenia: the french experience. Acta Psychiatr. Scand. Suppl. 380, 59–63. 10.1111/j.1600-0447.1994.tb05834.x
    1. Waxman S. G., Zamponi G. W. (2014). Regulating excitability of peripheral afferents: emerging ion channel targets. Nat. Neurosci. 17 (2), 153–163. 10.1038/nn.3602
    1. Wilder-Smith C. H. (1998). Pain treatment in multimorbid patients, the older population and other high-risk groups. The clinical challenge of reducing toxicity. Drug Saf. 18 (6), 457–472. 10.2165/00002018-199818060-00006
    1. Yekkirala A. S., Roberson D. P., Bean B. P., Woolf C. J. (2017). Breaking barriers to novel analgesic drug development. Nat. Rev. Drug Discov. 16 (8), 545–564. 10.1038/nrd.2017.87

Source: PubMed

3
Prenumerera