290
Views
2
CrossRef citations to date
0
Altmetric
Reviews

Targeted biological therapies for pain

&
Pages 1315-1326 | Published online: 01 Jun 2011

Bibliography

  • Arnoff GM, Dupuy DN. Evaluation and management of back pain: preventing disability. J Back Musculoskel Rehabil 1997;9:109-24
  • Stewart WF, Ricci JA, Chee EC, Lost productive time and cost due to common pain conditions in the US workforce. JAMA 2003;290:2443-54
  • Verhaak PF, Kerssens JJ, Dekker J, Prevalence of chronic benign pain disorder among adults: a review of the literature. Pain 1998;77:231-9
  • Fink D, Mata M, Glorioso JC. Cell and gene therapy in the treatment of pain. Adv Drug Deliv Rev 2003;55:1055-64
  • Glorioso JC, Mata M, Fink DJ. Gene therapy for chronic pain. Curr Opin Mol Ther 2003;5:483-8
  • Pohl M, Braz J. Gene therapy of pain: emerging strategies and future directions. Eur J Pharmacol 2001;429:39-48
  • Pohl M, Meunier A, Hamon M, Gene therapy of chronic pain. Curr Gene Ther 2003;3:223-38
  • Ballantyne JC, Mao J. Opioid therapy for chronic pain. N Engl J Med 2003;349:1943-53
  • Wilson SP, Yeomans DC. Virally mediated delivery of enkephalin and other neuropeptide transgenes in experimental pain models. Ann NY Acad Sci 2002;971:515-21
  • Wu CL, Garry MG, Zollo RA, Gene therapy for the management of pain: part II: molecular targets. Anesthesiol 2001;95:216-40
  • Garrity-Moses ME, Liu JK, Boulis NM. Molecular biology and gene therapy in the treatment of chronic pain. Neurosurg Clin N Am 2003;14:419-35
  • Mata M, Glorioso JC, Fink DJ. Gene therapy: novel treatments for polyneuropathy and chronic pain. Curr Neurol Neurosci Rep 2004;4:1-2
  • Wilson SP, Yeomans DC, Bender MA, Antihyperalgesic effects of infection with a preproenkephalin-encoding herpes virus. Proc Natl Acad Sci USA 1999;96:3211-16
  • Wilson SP, Yeomans DC. Genetic therapy for pain management. Curr Rev Pain 2000;4:445-50
  • Dubin AE, Patapoutian A. Nociceptors: the sensors of the pain pathway. J Clin Invest 2010;120:3760-72
  • Wolfe D, Mata M, Fink DJ. A human trial of HSV-mediated gene transfer for the treatment of chronic pain. Gene Ther 2009;16:455-60
  • Hao S, Mata M, Fink DJ. Viral vector-based gene transfer for treatment of chronic pain. Int Anesthesiol Clin 2007;45:59-71
  • Hao S, Mata M, Goins W, Transgene-mediated enkephalin release enhances the effect of morphine and evades tolerance to produce a sustained antiallodynic effect in neuropathic pain. Pain 2003;102:135-42
  • Buchser E, Goddard M, Heyd B, Immunoisolated xenogenic chromaffin cell therapy for chronic pain. Initial clinical experience. Anesthesiol 1996;85:1005-12
  • Decosterd I, Buchser E, Gilliard N, Intrathecal implants of bovine chromaffin cells alleviate mechanical allodynia in a rat model of neuropathic pain. Pain 1998;76:159-66
  • Sagen J, Kemmler JE, Wang H. Adrenal medullary transplants increase spinal cord cerebrospinal fluid catecholamine levels and reduce pain sensitivity. J Neurochem 1991;56:623-7
  • Winn SR, Emerich DF. Managing chronic pain with encapsulated cell implants releasing catecholamines and endogenous opiods. Front Biosci 2005;10:367-78
  • Kim YM, Kwak KH, Lim JO, Reduction of allodynia by intrathecal transplantation of microencapsulated porcine chromaffin cells. Artif Organs 2009;33:240-9
  • Herzberg U, Hama A, Sagen J. Spinal subarachnoid adrenal medullary transplants reduce hind paw swelling and peripheral nerve transport following formalin injection in rats. Brain Res 2008;1198:85-92
  • Jozan S, Aziza J, Chatelin S, Human fetal chromaffin cells: a potential tool for cell pain therapy. Exp Neurol 2007;205:525-35
  • Zhou H, Aziza J, Sol JC, Cell therapy of pain: Characterization of human fetal chromaffin cells at early adrenal medulla development. Exp Neurol 2006;198:370-81
  • Eaton MJ, Plunkett JA, Martinez MA, Transplants of neuronal cells bioengineered to synthesize GABA alleviate chronic neuropathic pain. Cell Transplant 1999;8:87-101
  • Cejas PJ, Martinez M, Karmally S, Lumbar transplant of neurons genetically modified to secrete brain-derived neurotrophic factor attenuates allodynia and hyperalgesia after sciatic nerve constriction. Pain 2000;86:195-210
  • Sugaya I, Qu T, Sugaya K, Genetically engineered human mesenchymal stem cells produce met-enkephalin at augmented higher levels in vitro. Cell Transplant 2006;15:225-30
  • Lazorthes Y, Sagen J, Sallerin B, Human chromaffin cell graft into the CSF for cancer pain management: a prospective Phase II clinical study. Pain 2000;87:19-32
  • Bes JC, Tkaczuk J, Czech KA, One-year chromaffin cell allograft survival in cancer patients with chronic pain: morphological and functional evidence. Cell Transplant 1998;7:227-38
  • Hao S, Mata M, Wolfe D, HSV-mediated gene transfer of the glial cell-derived neurotrophic factor provides an antiallodynic effect on neuropathic pain. Mol Ther 2003;8:367-75
  • Liu J, Wolfe D, Hao S, Peripherally delivered glutamic acid decarboxylase gene therapy for spinal cord injury pain. Mol Ther 2004;10:57-66
  • Siniscalco D, de Novellis V, Rossi F, Neuropathic pain: is the end of suffering starting in the gene therapy? Curr Drug Targets 2005;6:75-80
  • Xu Y, Gu Y, Wu P, Efficiencies of transgene expression in nociceptive neurons through different routes of delivery of adeno-associated viral vectors. Hum Gene Ther 2003;14:897-906
  • Finegold AA, Mannes AJ, Iadarola MJ. A paracrine paradigm for in vivo gene therapy in the central nervous system: treatment of chronic pain. Hum Gene Ther 1999;10:1251-7
  • Wu CL, Garry MG, Zollo RA, Gene therapy for the management of pain: Part I: Methods and strategies. Anesthesiol 2001;94:1119-32
  • Beutler AS, Reinhardt M. AAV for pain: steps towards clinical translation. Gene Ther 2009;16:461-9
  • Yeomans DC, Wilson SP. Herpes virus-based recombinant herpes vectors: gene therapy for pain and molecular tool for pain science. Gene Ther 2009;16:502-8
  • Glorioso JC, Fink DJ. Herpes vector-mediated gene transfer in the treatment of chronic pain. Mol Ther 2009;17:13-18
  • Meunier A, Latremoliere A, Mauborgne A, Attenuation of pain-related behavior in a rat model of trigeminal neuropathic pain by viral-driven enkephalin overproduction in trigeminal ganglion neurons. Mol Ther 2005;11:608-16
  • Storek B, Reinhardt M, Wang C, Sensory neuron targeting by self-complementary AAV8 via lumbar puncture for chronic pain. Proc Natl Acad Sci USA 2008;105:1055-60
  • Machelska H, Schroff M, Oswald D, Peripheral non-viral MIDGE vector-driven delivery of beta-endorphin in inflammatory pain. Mol Pain 2009;5:72
  • Wolfe D, Wechuck J, Krisky D, A clinical trial of gene therapy for chronic pain. Pain Med 2009;10:1325-30
  • Liu W, Liu Z, Liu L, A novel human foamy virus mediated gene transfer of GAD67 reduces neuropathic pain following spinal cord injury. Neurosci Lett 2008;432:13-18
  • Kim J, Kim SJ, Lee H, Effective neuropathic pain relief through sciatic nerve administration of GAD65-expressing rAAV2. Biochem Biophys Res Com 2009;388:73-8
  • Vit JP, Ohara PT, Sundberg C, Adenovector GAD65 gene delivery into the rat trigeminal ganglion produces orofacial analgesia. Mol Pain 2009;5:42
  • Jasmin L, Rabkin SD, Granato A, Analgesia and hyperalgesia from GABA-mediated modulation of the cerebral cortex. Nature 2003;424:316-20
  • Lima MC, Fregni F. Motor cortex stimulation for chronic pain: Systematic review and meta-analysis of the literature. Neurology 2008;70:2329-37
  • Eaton MJ, Blits B, Ruitenberg MJ, Amelioration of chronic neuropathic pain after partial nerve injury by adeno-associated viral (AAV) vector-mediated overexpression of BDNF in the rat spinal cord. Gene Ther 2002;9:1387-95
  • Boucher TJ, Okuse K, Bennett DL, Potent analgesic effects of GDNF in neuropathic pain states. Science 2000;290:124-7
  • Hao S, Mata M, Wolfe D, HSV-mediated gene transfer of the glial cell-derived neurotrophic factor provides an antiallodynic effect on neuropathic pain. Mol Ther 2003;8:367-75
  • Sloane EM, Soderquist RG, Maier SF, Long-term control of neuropathic pain in a non-viral gene therapy paradigm. Gene Ther 2009;16:470-5
  • Milligan ED, Langer SJ, Sloane EM, Controlling pathological pain by adenovirally driven spinal production of the anti-inflammatory cytokine, interleukin-10. Eur J Neurosci 2005;21:2136-48
  • Milligan ED, Sloane EM, Langer SJ, Controlling neuropathic pain by adenoassociated virus driven production of the anti-inflammatory cytokine, interleukin-10. Mol Pain 2005;1:9
  • Song P, Zhao ZQ, Liu XY. Expression of IL-2 receptor in dorsal root ganglion neurons and peripheral antinociception. Neuroreport 2000;11:1433-6
  • Zhao ZQ, Yang HQ, Liu XY. Inhibitory action of immunoregulator interleukin-2 on pain-sensitive spinal neurons in the cat. Chin J Neurosci 1994;1:37-42
  • Yao MZ, Gu JF, Wang JH, Adenovirus-mediated interleukin-2 gene therapy of nociception. Gene Ther 2003;10:1392-9
  • Hao S, Mata M, Glorioso JC, HSV-mediated expression of interleukin-4 in dorsal root ganglion neurons reduces neuropathic pain. Mol Pain 2006;2:6
  • Mata M, Hao S, Fink DJ. Gene therapy directed at the neuroimmune component of chronic pain with particular attention to the role of TNFalpha. Neurosci Lett 2008;437:209-13
  • Homma Y, Brull SJ, Zhang JM. A comparison of chronic pain behavior following local application of tumor necrosis factor alpha to the normal and mechanically compressed lumbar ganglia in the rat. Pain 2002;95:239-46
  • Peng XM, Zhou ZG, Glorioso JC, Tumor necrosis factor-alpha contributes to below-level neuropathic pain after spinal cord injury. Ann Neurol 2006;59:843-51
  • Wiley RG, Lappi DA. Targeted toxins in pain. Adv Drug Deliv Rev 2003;55:1043-54
  • Wiley RG, Kline IV RH. Neuronal lesioning with axonally transported toxins. J Neurosci Methods 2000;103:73-82
  • Wiley RG, Kline RH 4th, Vierck CJ Jr. Anti-nociceptive effects of selectively destroying substance P receptor-expressing dorsal horn neurons using [Sar9,Met(O2)11]-substance P-saporin: behavioral and anatomical analyses. Neurosci 2007;146:1333-45
  • Foster KA. The analgesic potential of clostridial neurotoxin derivatives. Expert Opin Investig Drugs 2004;13:1437-43
  • Mense S. Neurobiological basis for the use of botulinum toxin in pain therapy. J Neurol 2004;251(Suppl 1):I1-7
  • Dib-Hajj SD, Black JA, Waxman SG. Voltage-gated sodium channels: therapeutic targets for pain. Pain Med 2009;10:1260-9
  • Lai J, Porreca F, Hunter JC, Gold MS. Voltage-gated sodium channels and hyperalgesia. Annu Rev Pharmacol Toxicol 2004;44:371-97
  • Becker S, Terlau H. Toxins from cone snails: properties, applications and biotechnological production. Appl Microbiol Biotechnol. 2008;79:1-9
  • French RJ, Terlau H. Sodium channel toxins - receptor targeting and therapeutic potential. Curr Med Chem 2004;11:3053-64
  • Olivera BM, Cruz LJ, Desantos V, Neuronal calcium-channel antagonists - discrimination between calcium-channel subtypes using omega-conotoxin from Conus magus venom. Biochemistry 1987;26:2086-90
  • Miljanich GP. Ziconotide: neuronal calcium channel blocker for treating severe chronic pain. Curr Med Chem 2004;11:3029-40
  • Staats PS, Yearwood T, Charapata SG, Intrathecal ziconotide in the treatment of refractory pain in patients with cancer or AIDS: a randomized controlled trial. JAMA 2004;291:63-70
  • Smith HS, Deer TR. Safety and efficacy of intrathecal ziconotide in the management of severe chronic pain. Ther Clin Risk Manag 2009;5:521-34
  • Wallace MS, Rauck R, Fisher R, Intrathecal ziconotide for severe chronic pain: safety and tolerability results of an open-label, long-term trial. Anesth Analg 2008;106:628-37
  • Livett BG, Sandall DW, Keays D, Therapeutic applications of conotoxins that target the neuronal nicotinic acetylcholine receptor. Toxicon 2006;48:810-29
  • Sandall DW, Satkunanathan N, Keays DA, A novel alpha-conotoxin identified by gene sequencing is active in suppressing the vascular response to selective stimulation of sensory nerves in vivo. Biochemistry 2003;42:6904-11
  • Satkunanathan N, Livett B, Gayler K, Alpha-conotoxin Vc1.1 alleviates neuropathic pain and accelerates functional recovery of injured neurons. Brain Res 2005;1059:149-58
  • Olivera BM. Conus peptides: biodiversity-based discovery and exogenomics. J Biol Chem 2006;281:31173-7
  • Stone LS, Vulchanova L. The pain of antisense: in vivo application of antisense oligonucleotides for functional genomics in pain and analgesia. Adv Drug Deliv Rev 2003;55:1081-112
  • Kim SJ, Lee WI, Lee YS, Effective relief of neuropathic pain by adeno-associated virus-mediated expression of a small hairpin RNA against GTP cyclohydrolase 1. Mol Pain 2009;5:67
  • Sarret P, Dore-Savard L, Beaudet N. Direct application of siRNA for in vivo pain research. Methods Mol Biol 2010;623:383-95
  • Tan PH, Yang LC, Ji RR. Therapeutic potential of RNA interference in pain medicine. Open Pain J 2009;2:57-63
  • Tan PH, Yu SW, Lin VC, RNA interference-mediated gene silence of the NR1 subunit of the NMDA receptor by subcutaneous injection of vector-encoding short hairpin RNA reduces formalin-induced nociception in the rat. Pain 2011;152:573-81
  • Tan PH, Yang LC, Shih HC, Gene knockdown with intrathecal siRNA of NMDA receptor NR2B subunit reduces formalin-induced nociception in the rat. Gene Ther 2005;12:59-66
  • Garraway SM, Xu Q, Inturrisi CE. Design and evaluation of small interfering RNAs that target expression of the N-Methyl-D-aspartate receptor NR1 subunit gene in the spinal cord dorsal horn. J Pharmacol Exp Ther 2007;322:982-8
  • Castanotto D, Rossi JJ. The promises and pitfalls of RNA interference-based therapeutics. Nature 2009;457:426-33
  • Lee S, Kim Y, Back SK, Analgesic effect of highly reversible omega-conotoxin FVIA on N type Ca2+ channels. Mol Pain 2010;6:97
  • Gasior M, White NA, Rogawski MA. Prolonged attenuation of amygdala-kindled seizure measures in rats by convection-enhanced delivery of the N-type calcium channel antagonists omega-conotoxin GVIA and omega-conotoxin MVIIA. J Pharmacol Exp Ther 2007;323:458-68
  • Kress HG, Simpson KH, Marchettini P, Intrathecal therapy: what has changed with the introduction of ziconotide. Pain Pract 2009;9:338-47
  • Wallace MS, Rauck RL, Deer T. Ziconotide combination intrathecal therapy: rationale and evidence. Clin J Pain 2010;26:635-44
  • Rainov NG, Ren H. Gene therapy for human malignant brain tumors. Cancer J 2003;9:180-8

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.