Skip to main content
Log in

Targeting Nerve Growth Factor in Pain

What is the Therapeutic Potential?

  • Mechanisms and Targets
  • Published:
BioDrugs Aims and scope Submit manuscript

Abstract

Chronic pain presents a huge economic and social burden, with existing treatments largely unable to satisfy medical needs. Recently, studies have shown that nerve growth factor (NGF) is a major mediator of inflammatory and neuropathic pain, providing a new therapeutic target. Although originally discovered as a trophic factor for sympathetic and sensory neurons during development, it now appears that in adults, levels of NGF are elevated in many acute and chronic pain conditions. Furthermore, preclinical animal models of inflammatory and neuropathic pain also show increased NGF levels, while the sequestration of NGF alleviates the associated hyperalgesia. The molecular mechanisms involved are being elucidated.

This review briefly examines pain signaling pathways and describes currently available analgesics. It then investigates the approaches taken in targeting NGF-mediated pain. Current options being explored include the development of humanized monoclonal antibodies to NGF or its tyrosine kinase receptor TrkA (also known as neurotrophic tyrosine kinase receptor, type 1 [NTRK1]), and the sequestration of NGF using TrkA domain 5 (TrkAd5), a soluble receptor protein that binds NGF with picomolar affinity. Administration of either antibodies or TrkAd5 has been shown to be effective in a number of preclinical models of pain, including cystitis, osteoarthritis, UV irradiation (sunburn), and skeletal bone pain due to fracture or cancer. Other possible future therapies examined in this review include small-molecule TrkA antagonists, which target either the extracellular NGF binding domain of TrkA or its intracellular tyrosine kinase domain.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Fricker J. Pain in Europe: a report [online]. Available from URL: http://www.paineurope.com/index.php?q=en/book_page/the_pain_in_europe_report [Accessed 2008 Jul 23]

  2. Pezet S, McMahon SB. Neurotrophins: mediators and modulators of pain. Annu Rev Neurosci 2006; 29: 507–38

    Article  PubMed  CAS  Google Scholar 

  3. Pingle SC, Matta JA, Ahern GP. Capsaicin receptor: TRPV1 a promiscuous TRP channel. Handb Exp Pharmacol 2007; (179): 155–71

    Article  PubMed  CAS  Google Scholar 

  4. Lacroix-Fralish ML, Ledoux JB, Mogil JS. The pain genes database: an interactive web browser of pain-related transgenic knockout studies. Pain 2007; 131: 3–4

    Article  PubMed  Google Scholar 

  5. Souslova V, Cesare P, Ding Y, et al. Warm-coding deficits and aberrant inflammatory pain in mice lacking P2X3 receptors. Nature 2000; 407: 1015–7

    Article  PubMed  CAS  Google Scholar 

  6. Sluka KA, Price MP, Breese NM, et al. Chronic hyperalgesia induced by repeated acid injections in muscle is abolished by the loss of ASIC3, but not ASIC1. Pain 2003; 106: 229–39

    Article  PubMed  CAS  Google Scholar 

  7. Rupniak NM, Boyce S, Webb JK, et al. Effects of the bradykinin B1 receptor antagonist des-Arg9[Leu8]bradykinin and genetic disruption of the B2 receptor on nociception in rats and mice. Pain 1997; 71: 89–97

    Article  PubMed  CAS  Google Scholar 

  8. Pesquero JB, Araujo RC, Heppenstall PA, et al. Hypoalgesia and altered inflammatory responses in mice lacking kinin B1 receptors. Proc Natl Acad sci U S A 2000; 97: 8140–5

    Article  PubMed  CAS  Google Scholar 

  9. Bolcskei K, Helyes Z, Szabo A, et al. Investigation of the role of TRPV1 receptors in acute and chronic nociceptive processes using gene-deficient mice. Pain 2005; 117: 368–76

    Article  PubMed  CAS  Google Scholar 

  10. Akopian AN, Souslova V, England S, et al. The tetrodotoxin-resistant sodium channel SNS has a specialized function in pain pathways. Nat Neurosci 1999; 2: 541–8

    Article  PubMed  CAS  Google Scholar 

  11. Hinz B, Cheremina O, Brune K. Acetaminophen (paracetamol) is a selective cyclooxygenase-2 inhibitor in man. FASEB J 2008; 22: 383–90

    Article  PubMed  CAS  Google Scholar 

  12. Ottani A, Leone S, Sandrini M, et al. The analgesic activity of paracetamol is prevented by the blockade of cannabinoid CB1 receptors. Eur J Pharmacol 2006; 531: 280–1

    Article  PubMed  CAS  Google Scholar 

  13. Zeilhofer HU. Prostanoids in nociception and pain. Biochem Pharmacol 2007; 73: 165–74

    Article  PubMed  CAS  Google Scholar 

  14. Flower RJ, Blackwell GJ. Anti-inflammatory steroids induce biosynthesis of a phospholipase A2 inhibitor which prevents prostaglandin generation. Nature 1979; 278: 456–9

    Article  PubMed  CAS  Google Scholar 

  15. Atici S, Cinel I, Cinel L, et al. Liver and kidney toxicity in chronic use of opioids: an experimental long term treatment model. J Biosci 2005; 30: 245–52

    Article  PubMed  CAS  Google Scholar 

  16. Thomas JR, von Gunten CF. Pain in terminally ill patients: guidelines for pharmacological management. CNS Drugs 2003; 17: 621–31

    Article  PubMed  CAS  Google Scholar 

  17. Schrijvers D. Pain control in cancer: recent findings and trends. Ann Oncol 2007; 18Suppl. 9: ix37–42

    Article  PubMed  Google Scholar 

  18. McCleane G. Pharmacological management of neuropathic pain. CNS Drugs 2003; 17: 1031–43

    Article  PubMed  CAS  Google Scholar 

  19. Rowbotham MC, Davies PS, Verkempinck C, et al. Lidocaine patch: double-blind controlled study of a new treatment method for post-herpetic neuralgia. Pain 1996; 65: 39–44

    Article  PubMed  CAS  Google Scholar 

  20. Kalso E, Edwards JE, Moore RA, et al. Opioids in chronic non-cancer pain: systematic review of efficacy and safety. Pain 2004; 112: 372–80

    Article  PubMed  CAS  Google Scholar 

  21. Levi-Montalcini R, Cohen S. In vitro and in vivo effects of a nerve growth-stimulating agent isolated from snake venom. Proc Natl Acad sci U S A 1956; 42: 695–9

    Article  PubMed  CAS  Google Scholar 

  22. Barde YA, Edgar D, Thoenen H. Purification of a new neurotrophic factor from mammalian brain. EMBO J 1982; 1: 549–53

    PubMed  CAS  Google Scholar 

  23. Ernfors P, Ibanez CF, Ebendal T, et al. Molecular cloning and neurotrophic activities of a protein with structural similarities to nerve growth factor: developmental and topographical expression in the brain. Proc Natl Acad sci USA 1990; 87: 5454–8

    Article  PubMed  CAS  Google Scholar 

  24. Berkemeier LR, Winslow JW, Kaplan DR, et al. Neurotrophin-5: a novel neurotrophic factor that activates trk and trkB. Neuron 1991; 7: 857–66

    Article  PubMed  CAS  Google Scholar 

  25. Allen SJ, Dawbarn D. Clinical relevance of the neurotrophins and their receptors. Clin sci (Lond) 2006; 110: 175–91

    Article  CAS  Google Scholar 

  26. Dawbarn D, Allen SJ. Neurotrophins and neurodegeneration. Neuropathol Appl Neurobiol 2003; 29: 211–30

    Article  PubMed  CAS  Google Scholar 

  27. Rodriguez-Tebar A, Dechant G, Barde YA. Binding of brain-derived neurotrophic factor to the nerve growth factor receptor. Neuron 1990; 4: 487–92

    Article  PubMed  CAS  Google Scholar 

  28. Martin-Zanca D, Oskam R, Mitra G, et al. Molecular and biochemical characterization of the human trk proto-oncogene. Mol Cell Biol 1989; 9: 24–33

    PubMed  CAS  Google Scholar 

  29. Klein R, Parada LF, Coulier F, et al. TrkB, a novel tyrosine protein kinase receptor expressed during mouse neural development. EMBO J 1989; 8: 3701–9

    PubMed  CAS  Google Scholar 

  30. Lamballe F, Klein R, Barbacid M. TrkC, a new member of the trk family of tyrosine protein kinases, is a receptor for neurotrophin-3. Cell 1991; 66: 967–79

    Article  PubMed  CAS  Google Scholar 

  31. He XL, Garcia KC. Structure of nerve growth factor complexed with the shared neurotrophin receptor p75. Science 2004; 304: 870–5

    Article  PubMed  CAS  Google Scholar 

  32. Aurikko JP, Ruotolo BT, Grossmann JG, et al. Characterization of symmetric complexes of nerve growth factor and the ectodomain of the pan-neurotrophin receptor, p75NTR. J Biol Chem 2005; 280: 33453–60

    Article  PubMed  CAS  Google Scholar 

  33. Schneider R, Schweiger M. A novel modular mosaic of cell adhesion motifs in the extracellular domains of the neurogenic trk and trkB tyrosine kinase receptors. Oncogene 1991; 6: 1807–11

    PubMed  CAS  Google Scholar 

  34. Perez P, Coll PM, Hempstead BL, et al. NGF binding to the trk tyrosine kinase receptor requires the extracellular immunoglobulin-like domains. Mol Cell Neurosci 1995; 6: 97–105

    Article  PubMed  CAS  Google Scholar 

  35. Wiesmann C, Ultsch MH, Bass SH, et al. Crystal structure of nerve growth factor in complex with the ligand-binding domain of the TrkA receptor. Nature 1999; 401: 184–8

    Article  PubMed  CAS  Google Scholar 

  36. Ultsch MH, Wiesmann C, Simmons LC, et al. Crystal structures of the neurotro-phin-binding domain of TrkA, TrkB and TrkC. J Mol Biol 1999; 290: 149–59

    Article  PubMed  CAS  Google Scholar 

  37. Robertson AG, Banfield MJ, Allen SJ, et al. Identification and structure of the nerve growth factor binding site on TrkA. Biochem Biophys Res Commun 2001; 282: 131–41

    Article  PubMed  CAS  Google Scholar 

  38. Kaplan DR, Miller FD. Neurotrophin signal transduction in the nervous system. Curr Opin Neurobiol 2000; 10: 381–91

    Article  PubMed  CAS  Google Scholar 

  39. Huang EJ, Reichardt LF. Trk receptors: roles in neuronal signal transduction. Annu Rev Biochem 2003; 72: 609–42

    Article  PubMed  CAS  Google Scholar 

  40. 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. Cell 1994; 76: 1001–11

    Article  PubMed  CAS  Google Scholar 

  41. Smeyne RJ, Klein R, Schnapp A, et al. Severe sensory and sympathetic neuropathies in mice carrying a disrupted Trk/NGF receptor gene. Nature 1994; 368: 246–9

    Article  PubMed  CAS  Google Scholar 

  42. Davis BM, Lewin GR, Mendell LM, et al. Altered expression of nerve growth factor in the skin of transgenic mice leads to changes in response to mechanical stimuli. Neuroscience 1993; 56: 789–92

    Article  PubMed  CAS  Google Scholar 

  43. Stucky CL, Koltzenburg M, Schneider M, et al. Overexpression of nerve growth factor in skin selectively affects the survival and functional properties of nociceptors. J Neurosci 1999; 19: 8509–16

    PubMed  CAS  Google Scholar 

  44. 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: 485–8

    Article  PubMed  CAS  Google Scholar 

  45. Mardy S, Miura Y, Endo F, et al. Congenital insensitivity to pain with anhidrosis: novel mutations in the TRKA (NTRK1) gene encoding a high-affinity receptor for nerve growth factor. Am J Hum Genet 1999; 64: 1570–9

    Article  PubMed  CAS  Google Scholar 

  46. Huehne K, Zweier C, Raab K, et al. Novel missense, insertion and deletion mutations in the neurotrophic tyrosine kinase receptor type 1 gene (NTRK1) associated with congenital insensitivity to pain with anhidrosis. Neuromuscul Disord 2008 Feb; 18(2): 159–66

    Article  PubMed  Google Scholar 

  47. Einarsdottir E, Carlsson A, Minde J, et al. A mutation in the nerve growth factor beta gene (NGFB) causes loss of pain perception. Hum Mol Genet 2004; 13: 799–805

    Article  PubMed  CAS  Google Scholar 

  48. Lowe EM, Anand P, Terenghi G, et al. Increased nerve growth factor levels in the urinary bladder of women with idiopathic sensory urgency and interstitial cystitis. Br J Urol 1997; 79: 572–7

    Article  PubMed  CAS  Google Scholar 

  49. Aloe L, Tuveri MA, Carcassi U, et al. Nerve growth factor in the synovial fluid of patients with chronic arthritis. Arthritis Rheum 1992; 35: 351–5

    Article  PubMed  CAS  Google Scholar 

  50. 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: 919–22

    Article  PubMed  CAS  Google Scholar 

  51. Friess H, Zhu ZW, di Mola FF, et al. Nerve growth factor and its high-affinity receptor in chronic pancreatitis. Ann Surg 1999; 230: 615–24

    Article  PubMed  CAS  Google Scholar 

  52. Lewin GR, Ritter AM, Mendell LM. Nerve growth factor-induced hyperalgesia in the neonatal and adult rat. J Neurosci 1993; 13: 2136–48

    PubMed  CAS  Google Scholar 

  53. Andreev NY, Dimitrieva N, Koltzenburg M, et al. Peripheral administration of nerve growth factor in the adult rat produces a thermal hyperalgesia that requires the presence of sympathetic post-ganglionic neurones. Pain 1995; 63: 109–15

    Article  PubMed  CAS  Google Scholar 

  54. Petty BG, Cornblath DR, Adornato BT, et al. The effect of systemically administered recombinant human nerve growth factor in healthy human subjects. Ann Neurol 1994; 36: 244–6

    Article  PubMed  CAS  Google Scholar 

  55. Dyck PJ, Peroutka S, Rask C, et al. Intradermal recombinant human nerve growth factor induces pressure allodynia and lowered heat-pain threshold in humans. Neurology 1997; 48: 501–5

    Article  PubMed  CAS  Google Scholar 

  56. Apfel SC. Nerve growth factor for the treatment of diabetic neuropathy: what went wrong, what went right, and what does the future hold? Int Rev Neurobiol 2002; 50: 393–413

    Article  PubMed  CAS  Google Scholar 

  57. 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: 241–7

    Article  PubMed  CAS  Google Scholar 

  58. Leon A, Buriani A, Dal Toso R, et al. Mast cells synthesize, store, and release nerve growth factor. Proc Natl Acad sci U S A 1994; 91: 3739–43

    Article  PubMed  CAS  Google Scholar 

  59. Bergmann I, Reiter R, Toyka KV, et al. Nerve growth factor evokes hyperalgesia in mice lacking the low-affinity neurotrophin receptor p75. Neurosci Lett 1998; 255: 87–90

    Article  PubMed  CAS  Google Scholar 

  60. 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

    Article  PubMed  CAS  Google Scholar 

  61. Averill S, McMahon SB, Clary DO, et al. Immunocytochemical localization of trkA receptors in chemically identified subgroups of adult rat sensory neurons. Eur J Neurosci 1995; 7: 1484–94

    Article  PubMed  CAS  Google Scholar 

  62. Priestley JV, Michael GJ, Averill S, et al. Regulation of nociceptive neurons by nerve growth factor and glial cell line derived neurotrophic factor. Can J Physiol Pharmacol 2002; 80: 495–505

    Article  PubMed  CAS  Google Scholar 

  63. Delcroix JD, Valletta JS, Wu C, et al. NGF signaling in sensory neurons: evidence that early endosomes carry NGF retrograde signals. Neuron 2003; 39: 69–84

    Article  PubMed  CAS  Google Scholar 

  64. Shu X, Mendell LM. Nerve growth factor acutely sensitizes the response of adult rat sensory neurons to capsaicin. Neurosci Lett 1999; 274: 159–62

    Article  PubMed  CAS  Google Scholar 

  65. 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

    Article  PubMed  CAS  Google Scholar 

  66. Chuang HH, Prescott ED, Kong H, et al. Bradykinin and nerve growth factor release the capsaicin receptor from PtdIns(4,5)P2-mediated inhibition. Nature 2001; 411: 957–62

    Article  PubMed  CAS  Google Scholar 

  67. Bonnington JK, McNaughton PA. Signalling pathways involved in the sensitisation of mouse nociceptive neurones by nerve growth factor. J Physiol 2003; 551: 433–46

    Article  PubMed  CAS  Google Scholar 

  68. Cesare P, Dekker LV, Sardini A, et al. Specific involvement of PKC-epsilon in sensitization of the neuronal response to painful heat. Neuron 1999; 23: 617–24

    Article  PubMed  CAS  Google Scholar 

  69. Premkumar LS, Ahern GP. Induction of vanilloid receptor channel activity by protein kinase C. Nature 2000; 408: 985–90

    Article  PubMed  CAS  Google Scholar 

  70. Numazaki M, Tominaga T, Toyooka H, et al. Direct phosphorylation of capsaicin receptor VR1 by protein kinase Cepsilon and identification of two target serine residues. J Biol Chem 2002; 277: 13375–8

    Article  PubMed  CAS  Google Scholar 

  71. Zhuang ZY, Xu H, Clapham DE, et al. Phosphatidylinositol 3-kinase activates ERK in primary sensory neurons and mediates inflammatory heat hyperalgesia through TRPV1 sensitization. J Neurosci 2004; 24: 8300–9

    Article  PubMed  CAS  Google Scholar 

  72. Ji RR, Samad TA, Jin SX, et al. p38 MAPK activation by NGF in primary sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia. Neuron 2002; 36: 57–68

    Article  PubMed  CAS  Google Scholar 

  73. Zhang X, Huang J, McNaughton PA. NGF rapidly increases membrane expression of TRPV1 heat-gated ion channels. EMBO J 2005; 24: 4211–23

    Article  PubMed  CAS  Google Scholar 

  74. Stein AT, Ufret-Vincenty CA, Hua L, et al. Phosphoinositide 3-kinase binds to TRPV1 and mediates NGF-stimulated TRPV1 trafficking to the plasma membrane. J Gen Physiol 2006; 128: 509–22

    Article  PubMed  CAS  Google Scholar 

  75. Winston J, Toma H, Shenoy M, et al. Nerve growth factor regulates VR-1 mRNA levels in cultures of adult dorsal root ganglion neurons. Pain 2001; 89: 181–6

    Article  PubMed  CAS  Google Scholar 

  76. Leader B, Baca QJ, Golan DE. Protein therapeutics: a summary and pharmacological classification. Nat Rev Drug Discov 2008; 7: 21–39

    Article  PubMed  CAS  Google Scholar 

  77. Porter CJ, Edwards GA, Charman SA. Lymphatic transport of proteins after s.c. injection: implications of animal model selection. Adv Drug Deliv Rev 2001; 50: 157–71

    Article  PubMed  CAS  Google Scholar 

  78. Kompella UB, Lee VH. Pharmacokinetics of peptide and protein drugs. In: Lee V HL, editor. Peptide and protein drug delivery. New York: Marcel Dekker, 1991: 391–484

    Google Scholar 

  79. Toon S. The relevance of pharmacokinetics in the development of biotechnology products. Eur J Drug Metab Pharmacokinet 1996; 21: 93–103

    Article  PubMed  CAS  Google Scholar 

  80. McMahon SB, Bennett DL, Priestley JV, et al. The biological effects of endogenous nerve growth factor on adult sensory neurons revealed by a trkA-IgG fusion molecule. Nat Med 1995; 1: 774–80

    Article  PubMed  CAS  Google Scholar 

  81. Koltzenburg M, Bennett DL, Shelton DL, et al. Neutralization of endogenous NGF prevents the sensitization of nociceptors supplying inflamed skin. Eur J Neurosci 1999; 11: 1698–704

    Article  PubMed  CAS  Google Scholar 

  82. Ro LS, Chen ST, Tang LM, et al. Effect of NGF and anti-NGF on neuropathic pain in rats following chronic constriction injury of the sciatic nerve. Pain 1999; 79: 265–74

    Article  PubMed  CAS  Google Scholar 

  83. Gwak YS, Nam TS, Paik KS, et al. Attenuation of mechanical hyperalgesia following spinal cord injury by administration of antibodies to nerve growth factor in the rat. Neurosci Lett 2003; 336: 117–20

    Article  PubMed  CAS  Google Scholar 

  84. Halvorson KG, Kubota K, Sevcik MA, et al. A blocking antibody to nerve growth factor attenuates skeletal pain induced by prostate tumor cells growing in bone. Cancer Res 2005; 65: 9426–35

    Article  PubMed  CAS  Google Scholar 

  85. Koewler NJ, Freeman KT, Buus RJ, et al. Effects of a monoclonal antibody raised against nerve growth factor on skeletal pain and bone healing after fracture of the C57BL/6J mouse femur. J Bone Miner Res 2007; 22: 1732–42

    Article  PubMed  CAS  Google Scholar 

  86. Jimenez-Andrade JM, Martin CD, Koewler NJ, et al. Nerve growth factor sequestering therapy attenuates non-malignant skeletal pain following fracture. Pain 2007; 133: 183–96

    Article  PubMed  CAS  Google Scholar 

  87. Hongo JS, Laramee GR, Urfer R, et al. Antibody binding regions on human nerve growth factor identified by homolog- and alanine-scanning mutagenesis. Hybridoma 2000; 19: 215–27

    Article  PubMed  CAS  Google Scholar 

  88. 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 2007; 322: 282–7

    Article  PubMed  CAS  Google Scholar 

  89. Amgen®. AMG 403 [online]. Available from URL: http://www.amgen.com/science/pipe_AMG403.html [Accessed 2008 12 Aug]

  90. Ruberti F, Bradbury A, Cattaneo A. Cloning and expression of an anti-nerve growth factor (NGF) antibody for studies using the neuroantibody approach. Cell Mol Neurobiol 1993; 13: 559–68

    Article  PubMed  CAS  Google Scholar 

  91. Capsoni S, Ugolini G, Comparini A, et al. Alzheimer-like neurodegeneration in aged antinerve growth factor transgenic mice. Proc Natl Acad sci U S A 2000; 97: 6826–31

    Article  PubMed  CAS  Google Scholar 

  92. Marinelli S, Ugolini G, Cattaneo A, et al. Analgesic effects of anti-NGF and anti-TrkA antibodies on neuropathic pain in mice and rapid functional recovery. Expanding Vistas in Neuropathic Pain; 2005 Aug 17–20; Uluru

  93. Ugolini G, Marinelli S, Covaceuszach S, et al. Effects of anti-NGF and anti-TrkA antibodies on inflammatory pain in mice. FENS Forum Abstracts 2004; 2: A121.22

    Google Scholar 

  94. Holden PH, Asopa V, Robertson AG, et al. Immunoglobulin-like domains define the nerve growth factor binding site of the TrkA receptor. Nat Biotechnol 1997; 15: 668–72

    Article  PubMed  CAS  Google Scholar 

  95. Banfield MJ, Naylor RL, Robertson AG, et al. Specificity in Trk receptor:neurotrophin interactions: the crystal structure of TrkB-d5 in complex with neurotro-phin-4/5. Structure 2001; 9: 1191–9

    Article  PubMed  CAS  Google Scholar 

  96. Wehrman T, He X, Raab B, et al. Structural and mechanistic insights into nerve growth factor interactions with the TrkA and p75 receptors. Neuron 2007; 53: 25–38

    Article  PubMed  CAS  Google Scholar 

  97. Watson JJ, Fahey MS, van den WE, et al. TrkAd5: a novel therapeutic agent for treatment of inflammatory pain and asthma. J Pharmacol Exp Ther 2006; 316: 1122–9

    Article  PubMed  CAS  Google Scholar 

  98. Djouhri L, Dawbarn D, Robertson A, et al. Time course and nerve growth factor dependence of inflammation-induced alterations in electrophysiological membrane properties in nociceptive primary afferent neurons. J Neurosci 2001; 21: 8722–33

    PubMed  CAS  Google Scholar 

  99. Pasricha J, Winston J, Wang W, et al. NGF induces sensitisation of the vanilloid receptor (TRPV1) response in sensory neurons in acute pancreatitits by recruiting silent nociceptors. Gastroenterology 2005; 128Suppl. 2: 588

    Google Scholar 

  100. Hu VY, Zvara P, Dattilio A, et al. Decrease in bladder overactivity with REN1820 in rats with cyclophosphamide induced cystitis. J Urol 2005; 173: 1016–21

    Article  PubMed  Google Scholar 

  101. Bishop T, Hewson DW, Yip PK, et al. Characterisation of ultraviolet-B-induced inflammation as a model of hyperalgesia in the rat. Pain 2007; 131: 70–82

    Article  PubMed  CAS  Google Scholar 

  102. Sutherland S. Peptibodies: the new cool technology. Drug Discov Today 2004; 9: 683

    Article  PubMed  Google Scholar 

  103. Ugolini G, Marinelli S, Covaceuszach S, et al. The function neutralizing anti-TrkA antibody MNAC13 reduces inflammatory and neuropathic pain. Proc Natl Acad sci U S A 2007; 104: 2985–90

    Article  PubMed  CAS  Google Scholar 

  104. Cattaneo A, Capsoni S, Margotti E, et al. Functional blockade of tyrosine kinase A in the rat basal forebrain by a novel antagonistic anti-receptor monoclonal antibody. J Neurosci 1999; 19: 9687–97

    PubMed  CAS  Google Scholar 

  105. Covaceuszach S, Cattaneo A, Lamba D. Neutralization of NGF-TrkA receptor interaction by the novel antagonistic anti-TrkA monoclonal antibody MNAC13: a structural insight. Proteins 2005; 58: 717–27

    Article  PubMed  CAS  Google Scholar 

  106. LeSauteur L, Wei L, Gibbs BF, et al. Small peptide mimics of nerve growth factor bind TrkA receptors and affect biological responses. J Biol Chem 1995; 270: 6564–9

    Article  PubMed  CAS  Google Scholar 

  107. Owolabi JB, Rizkalla G, Tehim A, et al. Characterization of antiallodynic actions of ALE-0540, a novel nerve growth factor receptor antagonist, in the rat. J Pharmacol Exp Ther 1999; 289: 1271–6

    PubMed  CAS  Google Scholar 

  108. Colquhoun A, Lawrance GM, Shamovsky IL, et al. Differential activity of the nerve growth factor (NGF) antagonist PD90780 [7-(benzolylamino)-4,9-dihydro-4-methyl-9-oxo-pyrazolo[5,1 -b]quinazoline-2-carboxylic acid] suggests altered NGF-p75NTR interactions in the presence of TrkA. J Pharmacol Exp Ther 2004; 310: 505–11

    Article  PubMed  CAS  Google Scholar 

  109. PainCeptor. NGF: background [online]. Available from URL: http://www.painceptor.com/page.asp?IntNodeID=15183 [Accessed 2008 12 Aug]

  110. Dawbarn D, Fahey M, Watson J, et al. NGF receptor TrkAd5: therapeutic agent and drug design target. Biochem Soc Trans 2006; 34: 587–90

    Article  PubMed  CAS  Google Scholar 

  111. Winston JH, Toma H, Shenoy M, et al. Acute pancreatitis results in referred mechanical hypersensitivity and neuropeptide up-regulation that can be suppressed by the protein kinase inhibitor k252a. J Pain 2003; 4: 329–37

    Article  PubMed  CAS  Google Scholar 

  112. LippaB, Morris J, Corbett M, et al. Discovery of novel isothiazole inhibitors of the TrkA kinase: structure-activity relationship, computer modeling, optimization, and identification of highly potent antagonists. Bioorg Med Chem Lett 2006; 16: 3444–8

    Article  CAS  Google Scholar 

  113. Zhang HS, Krisky D, McNamara AR, et al. An engineered zinc finger protein transcriptional repressor of TrkA reduces nociception in a mouse model of bone cancer pain [poster no. 182.4/NN20]. Neuroscience 2007; 2007 Nov 3–7; San Diego (CA)

  114. Sangamo BioSciences Inc. Human therapeutics overview [online]. Available from URL: http://www.sangamo.com/human_human_overview.html#intract [Accessed 2008 Aug 12]

Download references

Acknowledgments

Judy Watson is funded by Bristol Research into Alzheimer’s and Care of the Elderly. Shelley Allen is a Sigmund Gestetner Senior Research Fellow. David Dawbarn and Shelley Allen are inventors on University of Bristol patents involving TrkAd5 (TrkAIg2).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to DrDavid Dawbarn.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Watson, J.J., Allen, S.J. & Dawbarn, D. Targeting Nerve Growth Factor in Pain. BioDrugs 22, 349–359 (2008). https://doi.org/10.2165/0063030-200822060-00002

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/0063030-200822060-00002

Keywords

Navigation