766
Views
20
CrossRef citations to date
0
Altmetric
Drug Evaluation

Tanezumab in the treatment of chronic musculoskeletal conditions

&
Pages 245-254 | Received 07 Sep 2016, Accepted 09 Dec 2016, Published online: 25 Dec 2016

References

  • United States Bone and Joint Initiative: The Burden of Musculoskeletal Diseases in the United States (BMUS), Third Edition. Rosemont, IL. Available at http://www.boneandjointburden.org. Accessed Aug 2016. 2014.
  • Kelsey JL, White AA, Sci M. Epidemiology and impact of low-back pain. Spine (Phila Pa 1976). 1980;5:133–142.
  • Loveless MS, Fry AL. Pharmacologic therapies in musculoskeletal conditions. Med Clin America. 2016;100:869–890. DOI:10.1016/j.mcna.2016.03.015
  • McAlindon TE, Bannuru RR, Sullivan MC, et al. OARSI guidelines for the non-surgical management of knee osteoarthritis. Osteoarthritis and Cartilage. 2014;22:363–388. DOI:10.1016/j.joca.2014.01.003
  • Candido KD, Chiweshe J, Anantamongkol U, et al. Can chronic pain patients be adequately treated using generic pain medications to the exclusion of brand-name ones? Am J Ther. 2016;23:e489–e497. DOI:10.1097/MJT.0000000000000098
  • Malfait AM, Miller RJ. Emerging targets for the management of osteoarthritis pain. Curr Osteoporos Rep. 2016;14(6):260–268.
  • Hefti FF, Rosenthal A, Walicke PA, et al. Novel class of pain drugs based on antagonism of NGF. Trends Pharmacol Sci. 2006;27:85–91. DOI:10.1016/j.tips.2005.12.001
  • McMahon SB. NGF as a mediator of inflammatory pain. Philosophical Trans Royal Soc B: Biol Sci. 1996;351:431–440. DOI:10.1098/rstb.1996.0039
  • Raychaudhuri SK, Raychaudhuri SP. NGF and its receptor system: a new dimension in the pathogenesis of psoriasis and psoriatic arthritis. Ann N Y Acad Sci. 2009;1173:470–477. DOI:10.1111/j.1749-6632.2009.04652.x
  • Fiore M, Chaldakov GN, Aloe L. Nerve growth factor as a signaling molecule for nerve cells and also for the neuroendocrine-immune systems. Rev Neurosci. 2009;20:133–145.
  • Cohen S. Origins of growth factors: NGF and EGF. Ann N Y Acad Sci. 2004;1038:98–102. DOI:10.1196/annals.1315.017
  • Tuszynski MH. Nerve growth factor gene delivery: animal models to clinical trials. Dev Neurobiol. 2007;67:1204–1215. DOI:10.1002/dneu.20510
  • Halliday DA, Zettler C, Rush RA, et al. Elevated nerve growth factor levels in the synovial fluid of patients with inflammatory joint disease. Neurochem Res. 1998;23:919–922.
  • Svensson P, Cairns BE, Wang K, et al. Injection of nerve growth factor into human masseter muscle evokes long-lasting mechanical allodynia and hyperalgesia. Pain. 2003;104:241–247.
  • Bergin MJG, Hirata R, Mista C, et al. Movement evoked pain and mechanical hyperalgesia after intramuscular injection of nerve growth factor: a model of sustained elbow pain. Pain Medicine (United States). 2015;16:2180–2191. DOI:10.1111/pme.12824
  • Indo Y, Tsuruta M, Hayashida Y, et al. Mutations in the TRKA/NGF receptor gene in patients with congenital insensitivity to pain with anhidrosis. Nat Genet. 1996;13:485–488. DOI:10.1038/ng0896-485
  • Lad SP, Peterson DA, Bradshaw RA, et al. Individual and combined effects of TrkA and p75NTR nerve growth factor receptors: a role for the high affinity receptor site. J Biol Chem. 2003;278:24808–24817. DOI:10.1074/jbc.M212270200
  • Holtzman DM, Kilbridge J, Li Y, et al. TrkA expression in the CNS: evidence for the existence of several novel NGF-responsive CNS neurons. J Neurosci. 1995;15:1567–1576.
  • Evans L, Andrew D, Robinson P, et al. Increased cutaneous NGF and CGRP-labelled trkA-positive intra-epidermal nerve fibres in rat diabetic skin. Neurosci Lett. 2012;506:59–63. DOI:10.1016/j.neulet.2011.10.049
  • Bonnington JK, McNaughton PA. Signalling pathways involved in the sensitisation of mouse nociceptive neurones by nerve growth factor. J Physiol. 2003;551:433–446. DOI:10.1113/jphysiol.2003.039990
  • Zhang X, Huang J, McNaughton PA. NGF rapidly increases membrane expression of TRPV1 heat-gated ion channels. Embo J. 2005;24:4211–4223. DOI:10.1038/sj.emboj.7600893
  • Zhu W, Oxford GS. Phosphoinositide-3-kinase and mitogen activated protein kinase signaling pathways mediate acute NGF sensitization of TRPV1. Mol Cell Neurosci. 2007;34:689–700. DOI:10.1016/j.mcn.2007.01.005
  • Kerr BJ, Souslova V, McMahon SB, et al. A role for the TTX-resistant sodium channel Nav 1.8 in NGF-induced hyperalgesia, but not neuropathic pain. Neuroreport. 2001;12:3077–3080.
  • Xu L, Nwosu LN, Burston JJ, et al. The anti-NGF antibody muMab 911 both prevents and reverses pain behaviour and subchondral osteoclast numbers in a rat model of osteoarthritis pain. Osteoarthritis and Cartilage. 2016;24:1587–1595. DOI:10.1016/j.joca.2016.05.015
  • Mantyh P, Tive L, Shelton D. Tanezumab, a humanized anti-nerve growth factor antibody for the treatment of pain. J Pain 2009;10. DOI:10.1016/j.jpain.2009.03.011
  • Jonsson EN, Xie R, Marshall SF, et al. Population pharmacokinetics of tanezumab in phase 3 clinical trials for osteoarthritis pain. Br J Clin Pharmacol. 2016;81:688–699. DOI:10.1111/bcp.12850
  • Arends R, Lalovic B, Marshall S, et al. Population pharmacokinetics of an anti-NGF humanized antibody, tanezumab, in patients with osteoarthritis: support to adoption of fixed-dose regimen. National Biotechnology Conference 2009.
  • Zorbas M, Hurst S, Shelton D, et al. A multiple-dose toxicity study of tanezumab in cynomolgus monkeys. Regul Toxicol Pharmacol. 2011;59:334–342. DOI:10.1016/j.yrtph.2010.11.005
  • Xie R, Arends R, Olson S, et al. Preliminary pharmacokinetic/pharmacodynamic (PK/PD) analysis for the effect of tanezumab on overall daily pain score data in adults with moderate-to-severe pain due to osteoarthritis of the knee. Presented At: The 2009 Annual Meeting of the Population Approach Group in Europe (PAGE) 2009.
  • Mariani G, Strober W. Immunoglobulin metabolism. Fc Receptors and the Action of Antibodies. In H. Metzger (ed).1990;94–177.
  • Bramson C, Herrmann DN, Carey W, et al. Exploring the role of tanezumab as a novel treatment for the relief of neuropathic pain. Pain Medicine (United States). 2015;16:1163–1176. DOI:10.1111/pme.12677
  • Evans RJ, Moldwin RM, Cossons N, et al. Proof of concept trial of tanezumab for the treatment of symptoms associated with interstitial cystitis. J Urol. 2011;185:1716–1721. DOI:10.1016/j.juro.2010.12.088
  • Nickel JC, Atkinson G, Krieger J. Tanezumab therapy for chronic prostatitis/chronic pelvic pain syndrome (cp/cpps): preliminary assessment of efficacy and safety in a randomized controlled trial. J Urol. 2011;185:e573–e574. DOI:10.1016/j.juro.2011.02.1344
  • Sevcik MA, Ghilardi JR, Peters CM, et al. Anti-NGF therapy profoundly reduces bone cancer pain and the accompanying increase in markers of peripheral and central sensitization. Pain. 2005;115:128–141. DOI:10.1016/j.pain.2005.02.022
  • Lane NE, Schnitzer TJ, Birbara CA, et al. Tanezumab for the treatment of pain from osteoarthritis of the knee. New England J Med. 2010;363:1521–1531. DOI:10.1056/NEJMoa0901510 .
  • Schnitzer TJ, Lane NE, Birbara C, et al. Long-term open-label study of tanezumab for moderate to severe osteoarthritic knee pain. Osteoarthritis and Cartilage. 2011;19:639–646. DOI:10.1016/j.joca.2011.01.009 .
  • Nagashima H, Suzuki M, Araki S, et al. Preliminary assessment of the safety and efficacy of tanezumab in Japanese patients with moderate to severe osteoarthritis of the knee: a randomized, double-blind, dose-escalation, placebo-controlled study. Osteoarthritis and Cartilage. 2011;19:1405–1412. DOI:10.1016/j.joca.2011.09.006 .
  • Brown MT, Murphy FT, Radin DM, et al. Tanezumab reduces osteoarthritic hip pain: results of a randomized, double-blind, placebo-controlled phase III trial. Arthritis Rheum. 2013;65:1795–1803. DOI:10.1002/art.37950 .
  • Brown MT, Murphy FT, Radin DM, et al. Tanezumab reduces osteoarthritic knee pain: results of a randomized, double-blind, placebo-controlled phase III trial. J Pain. 2012;13:790–798. DOI:10.1016/j.jpain.2012.05.006 .
  • Tive L, Dabezies EJ, Fountaine RJ. Long-term safety and efficacy of subcutaneous tanezumab in patients with knee or hip osteoarthritis (NCT00994890) [abstract]. Arthritis Rheum. 2013;65:S911–S912. DOI:10.1002/art.37830
  • Ekman EF, Gimbel JS, Bello AE, et al. Efficacy and safety of intravenous tanezumab for the symptomatic treatment of osteoarthritis: 2 randomized controlled trials versus naproxen. J Rheumatol. 2014;41:2249–2259. DOI:10.3899/jrheum.131294
  • Spierings ELH, Fidelholtz J, Wolfram G, et al. A phase III placebo- and oxycodone-controlled study of tanezumab in adults with osteoarthritis pain of the hip or knee. Pain. 2013;154:1603–1612. DOI:10.1016/j.pain.2013.04.035 .
  • Schnitzer TJ, Ekman EF, Spierings ELH, et al. Efficacy and safety of tanezumab monotherapy or combined with non-steroidal anti-inflammatory drugs in the treatment of knee or hip osteoarthritis pain. Ann Rheum Dis. 2015;74:1202–1211. DOI:10.1136/annrheumdis-2013-204905 .
  • Balanescu AR, Feist E, Wolfram G, et al. Efficacy and safety of tanezumab added on to diclofenac sustained release in patients with knee or hip osteoarthritis: a double-blind, placebo-controlled, parallel-group, multicentre phase III randomised clinical trial. Ann Rheum Dis. 2014;73:1665–1672.
  • Katz N, Borenstein DG, Birbara C, et al. Efficacy and safety of tanezumab in the treatment of chronic low back pain. Pain. 2011;152:2248–2258. DOI:10.1016/j.pain.2011.05.003
  • Kivitz AJ, Gimbel JS, Bramson C, et al. Efficacy and safety of tanezumab versus naproxen in the treatment of chronic low back pain. Pain. 2013;154:1009–1021. DOI:10.1016/j.pain.2013.03.006 .
  • Gimbel JS, Kivitz AJ, Bramson C, et al. Long-term safety and effectiveness of tanezumab as treatment for chronic low back pain. Pain. 2014;155:1793–1801. DOI:10.1016/j.pain.2014.06.004
  • Hochberg MC. Serious joint-related adverse events in randomized controlled trials of anti-nerve growth factor monoclonal antibodies. Osteoarthritis and Cartilage. 2015;23:S18–S21. DOI:10.1016/j.joca.2014.10.005
  • Hochberg MC, Tive LA, Abramson SB, et al. When is osteonecrosis not osteonecrosis? Adjudication of reported serious adverse joint events in the tanezumab clinical development program. Arthritis Rheumatol. 2016;68:382–391. DOI:10.1002/art.39492
  • Walsh N Tanezumab Eases OA Pain, Safety Still and Issue. Available at: http://www.medpagetoday.com/rheumatology/arthritis/44822. Medpage Today 2014.
  • Huskisson EC, Berry H, Gishen P, et al. Effects of antiinflammatory drugs on the progression of osteoarthritis of the knee. J Rheumatol. 1995;22:1941–1946.
  • Reijman M, Bierma-Zeinstra SMA, Pols HAP, et al. Is there an association between the use of different types of nonsteroidal antiinflammatory drugs and radiologic progression of osteoarthritis? The Rotterdam Study. Arthritis Rheum. 2005;52:3137–3142. DOI:10.1002/art.21357
  • Hurwitz DE, Ryals AR, Block JA, et al. Knee pain and joint loading in subjects with osteoarthritis of the knee. J Orthopaedic Res. 2000;18:572–579. DOI:10.1002/jor.1100180409
  • Tomlinson RZL, Riddle R, Clemens T Sensory nerve signals mediate skeletal adaptation to mechanical loads. Americal Society of Bone and Mineral Research Annual Meeting 2016; Abstract 1042.
  • Boursinos LA, Karachalios T, Poultsides L, et al. Do steroids, conventional non-steroidal anti-inflammatory drugs and selective COX-2 inhibitors adversely affect fracture healing? J Musculoskelet Neuronal Interactions. 2009;9:44–52.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.