37
Views
4
CrossRef citations to date
0
Altmetric
Theme: Pain - Review

Biologics: the next-generation therapeutics for analgesia?

, &
Pages 1653-1658 | Published online: 09 Jan 2014

References

  • Melnikova I. Pain market. Nat. Rev. Drug Discov.9(8), 589–590 (2010).
  • Hohenburg KA, Lyons J, Dalety TL. Pain Management Study 2: Chronic Pain. Decision Resources, MA, USA, 1–179 (2008).
  • Emery P, Zeidler H, Kvien TK et al. Celecoxib versus diclofenac in long-term management of rheumatoid arthritis: randomised double-blind comparison. Lancet354(9196), 2106–2111 (1999).
  • McGettigan P, Henry D. Cardiovascular risk and inhibition of cyclooxygenase: a systematic review of the observational studies of selective and nonselective inhibitors of cyclooxygenase 2. JAMA296(13), 1633–1644 (2006).
  • Dajani EZ, Islam K. Cardiovascular and gastrointestinal toxicity of selective cyclo-oxygenase-2 inhibitors in man. J. Physiol. Pharmacol.59(Suppl. 2), 117–133 (2008).
  • Collins SD, Chessell IP. Emerging therapies for neuropathic pain. Expert Opin. Emerg. Drugs.10(1), 95–108 (2005).
  • Chopade AR, Mulla WA. Novel strategies for the treatment of inflammatory hyperalgesia. Eur. J. Clin. Pharmacol.66, 429–444 (2010).
  • Schuster D, Laggner C, Langer T. Why drugs fail – a study on side effects in new chemical entities. Curr. Pharm. Des.11(27), 3545–3559 (2005).
  • Daugherty AL, Mrsny RJ. Formulation and delivery issues for monoclonal antibody therapeutics. Adv. Drug Del. Rev.58(5–6), 686–706 (2006).
  • Filpula D. Antibody engineering and modification techniques. Biomol. Eng.24(2), 201–215 (2007).
  • Bacher M, Depboylua C, Du Y et al. Peripheral and central biodistribution of (111)in-labeled anti-β-amyloid autoantibodies in a transgenic mouse model of Alzheimer’s disease. Neurosci. Lett.449(3), 240–245 (2009).
  • Schroder UA, Sabel BA. Nanoparticles, a drug carrier system to pass the blood–brain barrier, permit central analgesic effects of i.v. dalargin injections. Brain Res.710, 121–124 (1996).
  • Boado RJ, Zhang Y, Zhang Y, Xia CF, Pardridge WM. Fusion antibody for Alzheimer’s disease with bidirectional transport across the blood–brain barrier and aβ fibril disaggregation. Bioconjugate Chem.18(2), 447–455 (2007).
  • Cunha TM, Verri WA Jr, Silva JS, Poole S, Cunha FQ, Ferreira SH. A cascade of cytokines mediates mechanical inflammatory hypernociception in mice. Proc. Natl Acad. Sci.102(5), 1755–1760 (2005).
  • Woolf CJ, Allchorne A, 
Safieh-Garabedian B, Poole S. Cytokines, nerve growth factor and inflammatory hyperalgesia: the contribution of tumour necrosis factor α. Br. J. Pharmacol.121(3), 417–424 (1997).
  • Schaible HG, von Banchet GS, Boettger MK et al. The role of proinflammatory cytokines in the generation and maintenance of joint pain. Ann. NY Acad. Sci.1193(1), 60–69 (2010).
  • Moreland LW, Baumgartner SW, Schiff MH et al. Treatment of rheumatoid arthritis with a recombinant human tumor necrosis factor receptor (p75)–Fc fusion protein. N. Engl. J. Med.337(3), 141–147 (1997).
  • Smolen JS, Beaulieu A, Rubbert-Roth A et al. Effect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-controlled, randomised trial. Lancet371(9617), 987–997 (2008).
  • Sommer C, Schäfers M, Marziniak M, Toyka KV. Etanercept reduces hyperalgesia in experimental painful neuropathy. J. Peripher. Nerv. Syst.6(2), 67–72 (2001).
  • Maini RN, Breedveld FC, Kalden JR et al. Therapeutic efficacy of multiple intravenous infusions of anti-tumor necrosis factor α monoclonal antibody combined with low-dose weekly methotrexate in rheumatoid arthritis. Arthritis Rheum.41(9), 1552–1563 (1998).
  • Zalevsky J, Secher T, Ezhevsky SA et al. Dominant-negative inhibitors of soluble TNF attenuate experimental arthritis without suppressing innate immunity to infection. J. Immunol.179(3), 1872–1883 (2007).
  • Mukai Y, Nakamura T, Yoshioka Y et al. Fast binding kinetics and conserved 3D structure underlie the antagonistic activity of mutant TNF: useful information for designing artificial proteo-antagonists. J. Biochem.146(2), 167–172 (2009).
  • Chessell IP, Hatcher JP, Bountra C et al. Disruption of the P2X7 purinoceptor gene abolishes chronic inflammatory and neuropathic pain. Pain114(3), 386–396 (2005).
  • DeLeo JA, Colburn RW, Nichols, M, Malhotra A. Interleukin-6-mediated hyperalgesia/allodynia and increased spinal IL-6 expression in a rat mononeuropathy model. J. Interferon Cytokine Res.16(9), 695–700 (1996).
  • Murphy PG, Ramer MS, Borthwick L, Gauldie J, Richardson PM, Bisby MA. Endogenous interleukin-6 contributes to hypersensitivity to cutaneous stimuli and changes in neuropeptides associated with chronic nerve constriction in mice. Eur. J. Neurosci.11(7), 2243–2253 (1999).
  • Oka T, Oka K, Hosoi M, Hori, T. Intracerebroventricular injection of interleukin-6 induces thermal hyperalgesia in rats. Brain Res.692(1–2), 123–128 (1995).
  • Vardanyan M, Melemedjian OK, Price TJ et al. Reversal of pancreatitis-induced pain by an orally available, small molecule interleukin-6 receptor antagonist. Pain151(2), 257–265 (2010).
  • Crowley C, Spencer SD, Nishimura MC et al. Mice lacking nerve growth factor display perinatal loss of sensory and sympathetic neurons yet develop basal forebrain cholinergic neurons. Cell76(6), 1001–1011 (1994).
  • Stucky CL, Koltzenburg M, Schneider M, Engle MG, Albers KM, Davis BM. Overexpression of nerve growth factor in skin selectively affects the survival and functional properties of nociceptors. J. Neurosci.19(19), 8509–8516 (1999).
  • Andreev NY, Dimitrieva N, Koltzenburg M, McMahon SB. Peripheral administration of nerve growth factor in the adult rat produces a thermal hyperalgesia that requires the presence of sympathetic post-ganglionic neurones. Pain63(1), 109–115 (1995).
  • Dyck PJ, Peroutka S, Rask C et al. Intradermal recombinant human nerve growth factor induces pressure allodynia and lowered heatpain threshold in humans. Neurology48(2), 501–505 (1997).
  • McMahon SB, Bennett DL, Priestley JV, Shelton DL. The biological effects of endogenous nerve growth factor on adult sensory neurons revealed by a trkA–IgG fusion molecule. Nat. Med.1(8), 774–780 (1995).
  • Iannone F, De Bari C, Dell’Accio F et al. Increased expression of nerve growth factor (NGF) and high affinity NGF receptor (p140 TrkA) in human osteoarthritic chondrocytes. Rheumatology41(12), 1413–1418 (2002).
  • Friess H, Zhu ZW, di Mola FF et al. Nerve growth factor and its high-affinity receptor in chronic pancreatitis. Ann. Surg.230(5), 615–624 (1999).
  • Wild KD, Bian D, Zhu D et al. Antibodies to nerve growth factor reverse established tactile allodynia in rodent models of neuropathic pain without tolerance. J. Pharmacol. Exp. Ther.322(1), 282–287 (2007).
  • Lane NE, Schnitzer TJ, Birbara CA et al. Tanezumab for the treatment of pain from osteoarthritis of the knee. N. Engl. J. Med.363(16), 1521–1531 (2010).
  • Cattaneo A. Tanezumab, a recombinant humanised mAb against nerve growth factor for the treatment of acute and chronic pain. Curr. Opin. Mol. Ther.12(1), 94–106 (2010).
  • Micera A, Lambiase A, Stampachiacchiere B, Bonini S, Bonini S, Levi-Schaffer F. Nerve growth factor and tissue repair remodeling: trkA(NGFR) and p75(NTR), two receptors one fate. Cytokine Growth Factor Rev.18(3–4), 245–256 (2007).
  • Owolabi JB, Rizkalla G, Tehim A et al. Characterization of anti-allodynic actions of ALE-0540, a novel nerve growth factor receptor antagonist, in the rat. J. Pharmacol. Exp. Ther.289(3), 1271–1276 (1999).
  • Ren K, Torres R. Role of interleukin-1β during pain and inflammation. Brain Res. Rev.60(1), 57–64 (2009).
  • Gabay E, Wolf G, Shavit Y, Yirmiya R, Tal M. Chronic blockade of interleukin-1 (IL-1) prevents and attenuates neuropathic pain behavior and spontaneous ectopic neuronal activity following nerve injury. Eur. J. Pain.15(3), 242–248 (2010).
  • So A, De Smedt T, Revaz S, Tschopp J. A pilot study of IL-1 inhibition by anakinra in acute gout. Arthritis Res. Ther.9(2), R28 (2007).
  • Terkeltaub R, Sundy JS, Schumacher HR et al. The interleukin 1 inhibitor rilonacept in treatment of chronic gouty arthritis: results of a placebo-controlled, monosequence crossover, non-randomised, single-blind pilot study. Ann. Rheum. Dis.68(10), 1613–1617 (2009).
  • So A, Meulemeester MD, Pikhlak A et al. Canalinumab for the treatment of acute flares in difficult-to-treat gouty arthritis: Results of a multicenter, Phase II, dose ranging study. Arthritis Rheum.62(10), 3064–3076 (2010).
  • Lubberts E. IL-17/Th17 targeting: on the road to prevent chronic destructive arthritis. Cytokine41, 84–91 (2008).
  • Kim CF, Moalem-Taylor G. Interleukin-17 contributes to neuroinflammation and neuropathic pain following peripheral nerve injury in mice. J. Pain12(3), 370–383 (2010).
  • Noma N, Khan J, Chen IF et al. Interleukin-17 levels in rat models of nerve damage and neuropathic pain. Neurosci. Lett.493(3), 86–91 (2011).
  • Pinto LG, Cunha TM, Vieira SM et al. IL-17 mediates articular hypernociception in antigen-induced arthritis in mice. Pain148(2), 247–256 (2010).
  • Genovese MC, Van den Bosch F, Roberson SA et al. LY2439821, a humanized anti-interleukin-17 monoclonal antibody, in the treatment of patients with rheumatoid arthritis: a Phase I randomized, double-blind, placebo-controlled, proof-of-concept study. Arthritis Rheum.62(4), 929–939 (2010).
  • Dorn G, Patel S, Wotherspoon G et al. siRNA relieves chronic neuropathic pain. Nucleic Acids Res.32(5), e49 (2004).
  • Tan PH, Yang LC, Shih HC et al. Gene knockdown with intrathecal siRNA of NMDA receptor NR2B subunit reduces formalin-induced nociception in the rat. Gene Ther.12(1), 59–66 (2005).
  • Anesti AM, Coffin RS. Delivery of RNA interference triggers to sensory neurones in vivo using herpes simplex virus. Expert Opin. Biol. Ther.10(1), 89–103 (2010).
  • Buell G, Chessell IP, Michel AD et al. Blockade of human P2X7 receptor function with a monoclonal antibody. Blood92(10), 3521–3528 (1998).
  • Stone LS, Molliver DC. In search of analgesia: emerging roles of GPCRs in pain. Mol. Interv.9(5), 234–251 (2009).
  • Vergunst CE, Gerlag DM, Lopatinskaya L et al. Modulation of CCR2 in rheumatoid arthritis: a double-blind, randomized, placebo-controlled clinical trial. Arthritis Rheum.58(7), 1931–1939 (2008).
  • Mabry R, Snavely M. Therapeutic bispecific antibodies: the selection of stable single chain fragments to overcome engineering obstacles. iDrugs13(8), 543–549 (2010).

Website

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.