Randomised, double-blind, parallel group, placebo-controlled study to evaluate the analgesic efficacy and safety of VVZ-149 injections for postoperative pain following laparoscopic colorectal surgery

Srdjan S Nedeljkovic, Darin J Correll, Xiaodong Bao, Natacha Zamor, Jose L Zeballos, Yi Zhang, Mark J Young, Johanna Ledley, Jessica Sorace, Kristen Eng, Carlyle P Hamsher, Rajivan Maniam, Jonathan W Chin, Becky Tsui, Sunyoung Cho, Doo H Lee, Srdjan S Nedeljkovic, Darin J Correll, Xiaodong Bao, Natacha Zamor, Jose L Zeballos, Yi Zhang, Mark J Young, Johanna Ledley, Jessica Sorace, Kristen Eng, Carlyle P Hamsher, Rajivan Maniam, Jonathan W Chin, Becky Tsui, Sunyoung Cho, Doo H Lee

Abstract

Introduction: In spite of advances in understanding and technology, postoperative pain remains poorly treated for a significant number of patients. In colorectal surgery, the need for developing novel analgesics is especially important. Patients after bowel surgery are assessed for rapid return of bowel function and opioids worsen ileus, nausea and constipation. We describe a prospective, double-blind, parallel group, placebo-controlled randomised controlled trial testing the hypothesis that a novel analgesic drug, VVZ -149, is safe and effective in improving pain compared with providing opioid analgesia alone among adults undergoing laparoscopic colorectal surgery.

Methods and analysis: Based on sample size calculations for primary outcome, we plan to enrol 120 participants. Adult patients without significant medical comorbidities or ongoing opioid use and who are undergoing laparoscopic colorectal surgery will be enrolled. Participants are randomly assigned to receive either VVZ-149 with intravenous (IV) hydromorphone patient-controlled analgesia (PCA) or the control intervention (IV PCA alone) in the postoperative period. The primary outcome is the Sum of Pain Intensity Difference over 8 hours (SPID-8 postdose). Participants receive VVZ-149 for 8 hours postoperatively to the primary study end point, after which they continue to be assessed for up to 24 hours. We measure opioid consumption, record pain intensity and pain relief, and evaluate the number of rescue doses and requests for opioid. To assess safety, we record sedation, nausea and vomiting, respiratory depression, laboratory tests and ECG readings after study drug administration. We evaluate for possible confounders of analgesic response, such as anxiety, depression and catastrophising behaviours. The study will also collect blood sample data and evaluate for pharmacokinetic and pharmacodynamic relationships.

Ethics and dissemination: Ethical approval of the study protocol has been obtained from Institutional Review Boards at the participating institutions. Trial results will be disseminated through scientific conference presentations and by publication in scientific journals.

Trial registration number: NCT02489526; pre-results.

Keywords: CLINICAL PHARMACOLOGY; PAIN MANAGEMENT.

Conflict of interest statement

Competing interests: The following authors confirm that research grant funding has been assigned by the study sponsor, Vivozon, to Brigham and Women's Hospital, Massachusetts General Hospital, and Beth Israel Deaconess Medical Center, which are the employing institutions of authors SSN, DJC, XB, JLZ, YZ, MJY, JL, JS, KE, CPH, RM, JWC and BT. In addition, SSN received consulting fees from Vivozon. SC and DHL are employees of Vivozon Company, the sponsor of this study, and have received funding from the South Korean Ministry of Health and Welfare for R&D support for pharmaceutical development of the product that is described in this manuscript.

Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.

Figures

Figure 1
Figure 1
CONSORT diagram for VVZ-149 study flow. IV, intravenous; NRS, Numerical Pain Rating Scale; PCA, patient-controlled analgesia; PONV, Post-operative Nausea and Vomiting scale; RASS, Richmond Agitation and Sedation Scale.

References

    1. White PF, Kehlet H. Improving postoperative pain management: what are the unresolved issues? Anesthesiology 2010;112:220–5. 10.1097/ALN.0b013e3181c6316e
    1. Kissin I. The development of new analgesics over the past 50 years: a lack of real breakthrough drugs. Anesth Analg 2010;110:780–9. 10.1213/ANE.0b013e3181cde882
    1. Apfelbaum JL, Chen C, Mehta SS et al. . Postoperative pain experience: result from a national survey suggest postoperative pain continues to be undermanaged. Anesth Analg 2003;97:534–40. 10.1213/01.ANE.0000068822.10113.9E
    1. Koppert W, Schmelz M. The impact of opioid-induced hyperalgesia for postoperative pain. Best Pract Res Clin Aneaesthesiol 2007;21:65–83. 10.1016/j.bpa.2006.12.004
    1. Horl WH. Nonsteroidal anti-inflammatory drugs and the kidney. Pharmaceuticals (Basel) 2010;3:2291–321. 10.3390/ph3072291
    1. Ong CKS, Lirk P, Tan CH et al. . An evidence-based update on nonsteroidal anti-inflammatory drugs. Clin Med Ros 2007;5:19–34. 10.3121/cmr.2007.698
    1. Ho KY, Gen TJ, Habib AS. Gabapentin and postoperative pain—a systematic review of randomized controlled trials. Pain 2006;126:91–101. 10.1016/j.pain.2006.06.018
    1. Wongyingsinn M, Baldini G, Stein B et al. . Spinal analgesia for laparoscopic colonic resection using an enhanced recovery after surgery programme: better analgesia, but no benefits on postoperative recovery: a randomized controlled trial. Br J Anaesth 2012;108:850–6. 10.1093/bja/aes028
    1. Barthel FV, Urban AV, Schlosser L et al. . Long-term application of glycine transporter inhibitors acts antineuropathic and modulates spinal N-methyl-D-aspartate receptor subunit NR-1 expression in rats. Anesthesiology 2014;121:160–9. 10.1097/ALN.0000000000000203
    1. Betz H, Gomeza J, Armsen W et al. . Glycine transporters: essential regulators of synaptic transmission. Biochem Soc Trans 2006;34:55–8.
    1. Dohi T, Morita K, Kitayama T et al. . Glycine transporter inhibitors as a novel drug discovery strategy for neuropathic pain. Pharmacol Ther 2009;123:54–79. 10.1016/j.pharmthera.2009.03.018
    1. Werdehausen R, Mittnacht S, Bee LA et al. . The lidocaine metabolite N-ethylglycine has antinociceptive effects in experimental inflammatory and neuropathic pain. Pain 2015;156:1647–59. 10.1097/j.pain.0000000000000206
    1. Yoshikawa S, Oguchi T, Funahashi Y et al. . Glycine Transporter Type 2 (GlyT2) inhibitor ameliorates bladder overactivity and nociceptive behavior in rats. Eur Urol 2012;62:704–12. 10.1016/j.eururo.2012.01.044
    1. Zeilhofer HU. The glycinergic control of spinal pain processing. Cell Mol Life Sci 2005;62:2027–35. 10.1007/s00018-005-5107-2
    1. Beig MI, Baumert M, Walker FR et al. . Blockade of 5-HT2A receptors suppresses hyperthermic but not cardiovascular responses to psychosocial stress in rats. Neuroscience 2009;159:1185–91. 10.1016/j.neuroscience.2009.01.038
    1. Calejesan AA, Ch'ang MHC, Zhuo M. Spinal serotonergic receptors mediate facilitation of a nociceptive reflex by subcutaneous formalin injection into the hindpaw in rats. Brain Res 1998;798:46–54. 10.1016/S0006-8993(98)00394-1
    1. Mannion RJ, Costigan M, Decosterd I et al. . Neurotrophins: peripherally and centrally acting modulators of tactile stimulus-induced inflammatory pain hypersensitivity. Proc Natl Acad Sci USA 1999;96:9385–90. 10.1073/pnas.96.16.9385
    1. Okamoto K, Imbe H, Morikawa Y et al. . 5-HT2A receptor subtype in the peripheral branch of sensory fibers is involved in the potentiation of inflammatory pain in rats. Pain 2002;99:133–43. 10.1016/S0304-3959(02)00070-2
    1. Silveira JW, Dias QM, Bel EAD et al. . Serotonin receptors are involved in the spinal mediation of descending facilitation of surgical incision-induced increase of Fos-like immunoreactivity in rats. Mol Pain 2010;6:17 10.1186/1744-8069-6-17
    1. Zimmerman GR, Lehar J, Keith CT. Multi-target therapeutics: when the whole is greater than the sum of the parts. Drug Discov Today 2007;12:34–42. 10.1016/j.drudis.2006.11.008
    1. Lee D. Phase 1 study to investigate safety, tolerability, and pharmacokinetics of VVZ-149 injection (P1_VVZ149IV). In: Investigational New Drug 1.14.4.1 Investigational Brochure, IND124041; Version 2.1. 22 September, 2014 Q4. New Drug Application (NDA) submitted to the U.S. Food and Drug Administration.
    1. Joshi GP, Bonnet F, Kehlet H, PROSPECT collaboration. Evidence-based postoperative pain management after laparoscopic colorectal surgery. Colorectal Dis 2013;15:146–55. 10.1111/j.1463-1318.2012.03062.x
    1. Katz JN, Chang LC, Sangha O et al. . Can comorbidity be assessed by questionnaire rather than medical record review? Med Care 1996;34:73–84. 10.1097/00005650-199601000-00006
    1. Callahan CM, Unverzagt FW, Hui SL et al. . Six-item screener to identify cognitive impairment among potential subjects for research. Med Care 2002;40:771–81. 10.1097/01.MLR.0000024610.33213.C8
    1. Brown RL, Leonard T, Saunders LA et al. . A two-item conjoint screen for alcohol and other drug problems. J Am Board Fam Pract 2001;14:95–106.
    1. Zigmond AS, Snaith RP. The hospital anxiety and depression scale. Acta Psychiatr Scand 1983;67:361–70. 10.1111/j.1600-0447.1983.tb09716.x
    1. Sullivan MJ, Thorn B, Haythornthwaite JA et al. . Theoretical perspectives on the relation between catastrophizing and pain. Clin J Pain 2001;17:52–64. 10.1097/00002508-200103000-00008
    1. Wallace LM. Surgical patients’ expectations of pain and discomfort: does accuracy of expectations minimize post-surgical pain and distress? Pain 1985;22:363–73. 10.1016/0304-3959(85)90042-9
    1. Jarzyna D, Jungquist CR, Pasero C et al. . American Society for Pain Management Nursing guidelines on monitoring for opioid-induced sedation and respiratory depression. Pain Manag Nurs 2011;12:118–45.e10. 10.1016/j.pmn.2011.06.008
    1. Sessler CN, Gosnell MS, Grap MJ et al. . The Richmond Agitation-Sedation Scale: validity and reliability in adult intensive care unit patients. Am J Respir Crit Care Med 2002;166:1338–44. 10.1164/rccm.2107138
    1. Wengritzky R, Mettho T, Myles PS et al. . Development and validation of a postoperative nausea and vomiting intensity scale. Br J Anaesth 2010;104:158–66.
    1. Kolahi J, Bang H, Park J. Towards a proposal for assessment of blinding success in clinical trials: up-to-date review. Community Dent Oral Epidemiol 2009;37:477–84. 10.1111/j.1600-0528.2009.00494.x
    1. Barden J, Edwards JE, Mason L et al. . Outcomes in acute pain trials: systematic review of what was reported? Pain 2004;109:351–6. 10.1016/j.pain.2004.01.032
    1. Singla N, Hunsinger M, Chang PD et al. . Assay sensitivity of pain intensity versus pain relief in acute pain clinical trials: ACTTION systematic review and meta-analysis. J Pain 2015;16:683–91. 10.1016/j.jpain.2015.03.015
    1. Patel S, Lutz JM, Panchagnula U et al. . Anesthesia and perioperative management of colorectal surgical patients—a clinical review (Part 1). J Anaesthesiol Clin Pharmacol 2012;28: 162–71. 10.4103/0970-9185.94831
    1. Rawlinson A, Kang P, Evans J et al. . A systematic review of enhanced recovery protocols in colorectal surgery. Ann R Coll Surg Engl 2011;93:583–8. 10.1308/147870811X605219

Source: PubMed

3
Tilaa