Skip to main content
Top
Published in: BMC Musculoskeletal Disorders 1/2017

Open Access 01-12-2017 | Research article

Efficacy of nerve growth factor antibody in a knee osteoarthritis pain model in mice

Authors: Masayuki Miyagi, Tetsuhiro Ishikawa, Hiroto Kamoda, Miyako Suzuki, Gen Inoue, Yoshihiro Sakuma, Yasuhiro Oikawa, Sumihisa Orita, Kentaro Uchida, Kazuhisa Takahashi, Masashi Takaso, Seiji Ohtori

Published in: BMC Musculoskeletal Disorders | Issue 1/2017

Login to get access

Abstract

Background

Nerve growth factor (NGF) is not only an important factor in nerve growth but also a major contributor to the production of inflammation. It has been reported that inhibiting NGF could reduce several types of pain in several animal models. Here, we aimed to clarify the efficacy of NGF antibody in a knee osteoarthritis (OA) pain model in mice.

Method

Six-week-old male C57BR/J mice were used (n = 30). Ten mice comprised the control group, which received saline injection into the right knee joints; the other 20 mice comprised the experimental group, which received monoiodoacetate (MIA) injection into the right knee joints. Three weeks after surgery, the 20 experimental mice were randomly placed into treatment groups which received either sterile saline (non-treat group: 10 mg/kg, i.p.) or an anti-NGF antibody (anti-NGF group: 10 mg/kg, i.p.). Simultaneously, all mice received fluorogold (FG) retrograde neurotracer injection into their right joints. In a behavioral study, we evaluated gait using the CatWalk quantitative gait analysis system before surgery, 3 weeks after surgery (before treatment), 4 weeks after surgery (one week after surgery), and 5 weeks after surgery (2 weeks after surgery). In immunohistochemical analysis, the right dorsal root ganglia (DRGs) from the L4–L6 levels were resected 5 weeks after surgery (2 weeks after surgery). They were immunostained for calcitonin gene-related peptide (CGRP), and the number of FG-labeled or CGRP-immunoreactive (IR) DRG neurons was counted.

Results

On gait analysis using the CatWalk system, duty cycle, swing speed, and print area were decreased in non-treat group compared with those in control group and improved in the anti-NGF group compared with those in non-treat group. CGRP expression in DRGs was up-regulated in non-treat group compared with that in control group and suppressed in the anti-NGF group compared with that in non-treat group (both p < 0.05).

Conclusions

MIA injection into the knee joint induced gait impairment and the up-regulation of CGRP in DRG neurons in a knee OA pain model in mice. Intraperitoneal injection of anti-NGF antibody suppressed this impairment of gait and up-regulation of CGRP in DRG neurons. These finding suggest that anti-NGF therapy might be valuable in the treatment of OA pain in the knee.
Literature
1.
go back to reference Vaishya R, Pariyo GB, Agarwal AK, Vijay V. Non-operative management of osteoarthritis of the knee joint. J Clin. Orthop Trauma. 2016;7(3):170–6. Vaishya R, Pariyo GB, Agarwal AK, Vijay V. Non-operative management of osteoarthritis of the knee joint. J Clin. Orthop Trauma. 2016;7(3):170–6.
2.
go back to reference Miyagi M, Ishikawa T, Kamoda H, Suzuki M, Inoue G, Sakuma Y, Oikawa Y, Uchida K, Suzuki T, Takahashi K, Takaso M, Ohtori S. The efficacy of nerve growth factor antibody in a mouse model of neuropathic cancer pain. Exp.Anim 2016; 65 (4):337–343. Miyagi M, Ishikawa T, Kamoda H, Suzuki M, Inoue G, Sakuma Y, Oikawa Y, Uchida K, Suzuki T, Takahashi K, Takaso M, Ohtori S. The efficacy of nerve growth factor antibody in a mouse model of neuropathic cancer pain. Exp.Anim 2016; 65 (4):337–343.
3.
go back to reference Mendell LM. Neurotrophins and sensory neurons: role in development, maintenance and injury. A thematic summary. Philos.Trans.R.Soc.Lond B Biol.Sci. 1996;351(1338):463–7.CrossRefPubMed Mendell LM. Neurotrophins and sensory neurons: role in development, maintenance and injury. A thematic summary. Philos.Trans.R.Soc.Lond B Biol.Sci. 1996;351(1338):463–7.CrossRefPubMed
4.
5.
go back to reference Lewin GR, Rueff A, Mendell LM. Peripheral and central mechanisms of NGF-induced hyperalgesia. Eur.J Neurosci. 1994;6(12):1903–12.CrossRefPubMed Lewin GR, Rueff A, Mendell LM. Peripheral and central mechanisms of NGF-induced hyperalgesia. Eur.J Neurosci. 1994;6(12):1903–12.CrossRefPubMed
6.
go back to reference Ramer MS, Bisby MA. Adrenergic innervation of rat sensory ganglia following proximal or distal painful sciatic neuropathy: distinct mechanisms revealed by anti-NGF treatment. Eur. J Neurosci. 1999;11(3):837–46. Ramer MS, Bisby MA. Adrenergic innervation of rat sensory ganglia following proximal or distal painful sciatic neuropathy: distinct mechanisms revealed by anti-NGF treatment. Eur. J Neurosci. 1999;11(3):837–46.
7.
go back to reference Ro LS, Chen ST, Tang LM, Jacobs JM. Effect of NGF and anti-NGF on neuropathic pain in rats following chronic constriction injury of the sciatic nerve. Pain. 1999;79(2–3):265–74.CrossRefPubMed Ro LS, Chen ST, Tang LM, Jacobs JM. Effect of NGF and anti-NGF on neuropathic pain in rats following chronic constriction injury of the sciatic nerve. Pain. 1999;79(2–3):265–74.CrossRefPubMed
8.
go back to reference Wild KD, Bian D, Zhu D, Davis J, Bannon AW, Zhang TJ, Louis JC. Antibodies to nerve growth factor reverse established tactile allodynia in rodent models of neuropathic pain without tolerance. J Pharmacol.Exp.Ther. 2007;322(1):282–7.CrossRefPubMed Wild KD, Bian D, Zhu D, Davis J, Bannon AW, Zhang TJ, Louis JC. Antibodies to nerve growth factor reverse established tactile allodynia in rodent models of neuropathic pain without tolerance. J Pharmacol.Exp.Ther. 2007;322(1):282–7.CrossRefPubMed
9.
go back to reference Ochiai N, Ohtori S, Sasho T, Nakagawa K, Takahashi K, Takahashi N, Murata R, Takahashi K, Moriya H, Wada Y, Saisu T. Extracorporeal shock wave therapy improves motor dysfunction and pain originating from knee osteoarthritis in rats. Osteoarthritis. Cartilage. 2007;15(9):1093–6.CrossRef Ochiai N, Ohtori S, Sasho T, Nakagawa K, Takahashi K, Takahashi N, Murata R, Takahashi K, Moriya H, Wada Y, Saisu T. Extracorporeal shock wave therapy improves motor dysfunction and pain originating from knee osteoarthritis in rats. Osteoarthritis. Cartilage. 2007;15(9):1093–6.CrossRef
10.
go back to reference Muramatsu Y, Sasho T, Saito M, Yamaguchi S, Akagi R, Mukoyama S, Akatsu Y, Katsuragi J, Fukawa T, Endo J, Hoshi H, Yamamoto Y, Takahashi K. Preventive effects of hyaluronan from deterioration of gait parameters in surgically induced mice osteoarthritic knee model. Osteoarthritis. Cartilage. 2014;22(6):831–5.CrossRef Muramatsu Y, Sasho T, Saito M, Yamaguchi S, Akagi R, Mukoyama S, Akatsu Y, Katsuragi J, Fukawa T, Endo J, Hoshi H, Yamamoto Y, Takahashi K. Preventive effects of hyaluronan from deterioration of gait parameters in surgically induced mice osteoarthritic knee model. Osteoarthritis. Cartilage. 2014;22(6):831–5.CrossRef
11.
go back to reference Bendele AM. Animal models of osteoarthritis. J Musculoskelet. Neuronal.Interact. 2001;1(4):363–76. Bendele AM. Animal models of osteoarthritis. J Musculoskelet. Neuronal.Interact. 2001;1(4):363–76.
12.
go back to reference Daans M, Luyten FP, Lories RJ. GDF5 deficiency in mice is associated with instability-driven joint damage, gait and subchondral bone changes. Ann.Rheum.Dis. 2011;70(1):208–13.CrossRefPubMed Daans M, Luyten FP, Lories RJ. GDF5 deficiency in mice is associated with instability-driven joint damage, gait and subchondral bone changes. Ann.Rheum.Dis. 2011;70(1):208–13.CrossRefPubMed
13.
go back to reference Combe R, Bramwell S, Field MJ. The monosodium iodoacetate model of osteoarthritis: a model of chronic nociceptive pain in rats? Neurosci.Lett. 2004;370(2–3):236–40.CrossRefPubMed Combe R, Bramwell S, Field MJ. The monosodium iodoacetate model of osteoarthritis: a model of chronic nociceptive pain in rats? Neurosci.Lett. 2004;370(2–3):236–40.CrossRefPubMed
14.
go back to reference Guingamp C, Gegout-Pottie P, Philippe L, Terlain B, Netter P, Gillet P. Mono-iodoacetate-induced experimental osteoarthritis: a dose-response study of loss of mobility, morphology, and biochemistry. Arthritis Rheum. 1997;40(9):1670–9.CrossRefPubMed Guingamp C, Gegout-Pottie P, Philippe L, Terlain B, Netter P, Gillet P. Mono-iodoacetate-induced experimental osteoarthritis: a dose-response study of loss of mobility, morphology, and biochemistry. Arthritis Rheum. 1997;40(9):1670–9.CrossRefPubMed
15.
go back to reference Guzman RE, Evans MG, Bove S, Morenko B, Kilgore K. Mono-iodoacetate-induced histologic changes in subchondral bone and articular cartilage of rat femorotibial joints: an animal model of osteoarthritis. Toxicol.Pathol. 2003;31(6):619–24.CrossRefPubMed Guzman RE, Evans MG, Bove S, Morenko B, Kilgore K. Mono-iodoacetate-induced histologic changes in subchondral bone and articular cartilage of rat femorotibial joints: an animal model of osteoarthritis. Toxicol.Pathol. 2003;31(6):619–24.CrossRefPubMed
16.
go back to reference Orita S, Ishikawa T, Miyagi M, Ochiai N, Inoue G, Eguchi Y, Kamoda H, Arai G, Toyone T, Aoki Y, Kubo T, Takahashi K, Ohtori S. Pain-related sensory innervation in monoiodoacetate-induced osteoarthritis in rat knees that gradually develops neuronal injury in addition to inflammatory pain. BMCMusculoskeletDisord. 2011;12:134. Orita S, Ishikawa T, Miyagi M, Ochiai N, Inoue G, Eguchi Y, Kamoda H, Arai G, Toyone T, Aoki Y, Kubo T, Takahashi K, Ohtori S. Pain-related sensory innervation in monoiodoacetate-induced osteoarthritis in rat knees that gradually develops neuronal injury in addition to inflammatory pain. BMCMusculoskeletDisord. 2011;12:134.
17.
go back to reference Ferland CE, Laverty S, Beaudry F, Vachon P. Gait analysis and pain response of two rodent models of osteoarthritis. Pharmacol.Biochem.Behav. 2011;97(3):603–10.CrossRefPubMed Ferland CE, Laverty S, Beaudry F, Vachon P. Gait analysis and pain response of two rodent models of osteoarthritis. Pharmacol.Biochem.Behav. 2011;97(3):603–10.CrossRefPubMed
18.
go back to reference Ferreira-Gomes J, Adaes S, Castro-Lopes JM. Assessment of movement-evoked pain in osteoarthritis by the knee-bend and CatWalk tests: a clinically relevant study. J Pain. 2008;9(10):945–54.CrossRefPubMed Ferreira-Gomes J, Adaes S, Castro-Lopes JM. Assessment of movement-evoked pain in osteoarthritis by the knee-bend and CatWalk tests: a clinically relevant study. J Pain. 2008;9(10):945–54.CrossRefPubMed
19.
go back to reference Hamers FP, Lankhorst AJ, van Laar TJ, Veldhuis WB, Gispen WH. Automated quantitative gait analysis during overground locomotion in the rat: its application to spinal cord contusion and transection injuries. J Neurotrauma. 2001;18(2):187–201.CrossRefPubMed Hamers FP, Lankhorst AJ, van Laar TJ, Veldhuis WB, Gispen WH. Automated quantitative gait analysis during overground locomotion in the rat: its application to spinal cord contusion and transection injuries. J Neurotrauma. 2001;18(2):187–201.CrossRefPubMed
20.
go back to reference Nwosu LN, Mapp PI, Chapman V, Walsh DA. Relationship between structural pathology and pain behaviour in a model of osteoarthritis (OA). Osteoarthritis. Cartilage. 2016;24(11):1910–7.CrossRef Nwosu LN, Mapp PI, Chapman V, Walsh DA. Relationship between structural pathology and pain behaviour in a model of osteoarthritis (OA). Osteoarthritis. Cartilage. 2016;24(11):1910–7.CrossRef
21.
go back to reference Miyagi M, Ishikawa T, Kamoda H, Orita S, Kuniyoshi K, Ochiai N, Kishida S, Nakamura J, Eguchi Y, Arai G, Suzuki M, Aoki Y, Toyone T, Takahashi K, Inoue G, Ohtori S. Assessment of gait in a rat model of myofascial inflammation using the CatWalk system. Spine (Phila Pa 1976). 2011;36(21):1760–4.CrossRef Miyagi M, Ishikawa T, Kamoda H, Orita S, Kuniyoshi K, Ochiai N, Kishida S, Nakamura J, Eguchi Y, Arai G, Suzuki M, Aoki Y, Toyone T, Takahashi K, Inoue G, Ohtori S. Assessment of gait in a rat model of myofascial inflammation using the CatWalk system. Spine (Phila Pa 1976). 2011;36(21):1760–4.CrossRef
22.
go back to reference Miyagi M, Ishikawa T, Orita S, Eguchi Y, Kamoda H, Arai G, Suzuki M, Inoue G, Aoki Y, Toyone T, Takahashi K, Ohtori S. Disk injury in rats produces persistent increases in pain-related neuropeptides in dorsal root ganglia and spinal cord glia but only transient increases in inflammatory mediators: pathomechanism of chronic diskogenic low back pain. Spine (Phila Pa 1976). 2011;36(26):2260–6.CrossRef Miyagi M, Ishikawa T, Orita S, Eguchi Y, Kamoda H, Arai G, Suzuki M, Inoue G, Aoki Y, Toyone T, Takahashi K, Ohtori S. Disk injury in rats produces persistent increases in pain-related neuropeptides in dorsal root ganglia and spinal cord glia but only transient increases in inflammatory mediators: pathomechanism of chronic diskogenic low back pain. Spine (Phila Pa 1976). 2011;36(26):2260–6.CrossRef
23.
go back to reference Myers SL, Brandt KD, O'Connor BL, Visco DM, Albrecht ME. Synovitis and osteoarthritic changes in canine articular cartilage after anterior cruciate ligament transection. Effect of surgical hemostasis. Arthritis Rheum. 1990;33(9):1406–15.CrossRefPubMed Myers SL, Brandt KD, O'Connor BL, Visco DM, Albrecht ME. Synovitis and osteoarthritic changes in canine articular cartilage after anterior cruciate ligament transection. Effect of surgical hemostasis. Arthritis Rheum. 1990;33(9):1406–15.CrossRefPubMed
24.
go back to reference Marijnissen AC, van Roermund PM, TeKoppele JM, Bijlsma JW, Lafeber FP. The canine 'groove' model, compared with the ACLT model of osteoarthritis. Osteoarthritis. Cartilage. 2002;10(2):145–55.CrossRef Marijnissen AC, van Roermund PM, TeKoppele JM, Bijlsma JW, Lafeber FP. The canine 'groove' model, compared with the ACLT model of osteoarthritis. Osteoarthritis. Cartilage. 2002;10(2):145–55.CrossRef
25.
go back to reference Janusz MJ, Bendele AM, Brown KK, Taiwo YO, Hsieh L, Heitmeyer SA. Induction of osteoarthritis in the rat by surgical tear of the meniscus: inhibition of joint damage by a matrix metalloproteinase inhibitor. Osteoarthritis. Cartilage. 2002;10(10):785–91.CrossRef Janusz MJ, Bendele AM, Brown KK, Taiwo YO, Hsieh L, Heitmeyer SA. Induction of osteoarthritis in the rat by surgical tear of the meniscus: inhibition of joint damage by a matrix metalloproteinase inhibitor. Osteoarthritis. Cartilage. 2002;10(10):785–91.CrossRef
26.
go back to reference Shelton DL, Zeller J, Ho WH, Pons J, Rosenthal A. Nerve growth factor mediates hyperalgesia and cachexia in auto-immune arthritis. Pain. 2005;116(1–2):8–16.CrossRefPubMed Shelton DL, Zeller J, Ho WH, Pons J, Rosenthal A. Nerve growth factor mediates hyperalgesia and cachexia in auto-immune arthritis. Pain. 2005;116(1–2):8–16.CrossRefPubMed
27.
go back to reference Cendelin J, Voller J, Vozeh F. Ataxic gait analysis in a mouse model of the olivocerebellar degeneration. Behav. Brain Res. 2010;210(1):8–15. Cendelin J, Voller J, Vozeh F. Ataxic gait analysis in a mouse model of the olivocerebellar degeneration. Behav. Brain Res. 2010;210(1):8–15.
28.
go back to reference Chuang CS, Su HL, Cheng FC, Hsu SH, Chuang CF, Liu CS. Quantitative evaluation of motor function before and after engraftment of dopaminergic neurons in a rat model of Parkinson's disease. J Biomed.Sci. 2010;17:9.CrossRefPubMedPubMedCentral Chuang CS, Su HL, Cheng FC, Hsu SH, Chuang CF, Liu CS. Quantitative evaluation of motor function before and after engraftment of dopaminergic neurons in a rat model of Parkinson's disease. J Biomed.Sci. 2010;17:9.CrossRefPubMedPubMedCentral
29.
go back to reference Koopmans GC, Deumens R, Honig WM, Hamers FP, Steinbusch HW, Joosten EA. The assessment of locomotor function in spinal cord injured rats: the importance of objective analysis of coordination. J Neurotrauma. 2005;22(2):214–25.CrossRefPubMed Koopmans GC, Deumens R, Honig WM, Hamers FP, Steinbusch HW, Joosten EA. The assessment of locomotor function in spinal cord injured rats: the importance of objective analysis of coordination. J Neurotrauma. 2005;22(2):214–25.CrossRefPubMed
30.
go back to reference Vrinten DH, Hamers FF. CatWalk' automated quantitative gait analysis as a novel method to assess mechanical allodynia in the rat; a comparison with von Frey testing. Pain. 2003;102(1–2):203–9.CrossRefPubMed Vrinten DH, Hamers FF. CatWalk' automated quantitative gait analysis as a novel method to assess mechanical allodynia in the rat; a comparison with von Frey testing. Pain. 2003;102(1–2):203–9.CrossRefPubMed
31.
go back to reference Miyagi M, Ishikawa T, Kamoda H, Suzuki M, Sakuma Y, Orita S, Oikawa Y, Aoki Y, Toyone T, Takahashi K, Inoue G, Ohtori S. Assessment of pain behavior in a rat model of intervertebral disc injury using the CatWalk gait analysis system. Spine (Phila Pa 1976). 2013;38(17):1459–65.CrossRef Miyagi M, Ishikawa T, Kamoda H, Suzuki M, Sakuma Y, Orita S, Oikawa Y, Aoki Y, Toyone T, Takahashi K, Inoue G, Ohtori S. Assessment of pain behavior in a rat model of intervertebral disc injury using the CatWalk gait analysis system. Spine (Phila Pa 1976). 2013;38(17):1459–65.CrossRef
32.
go back to reference Averill S, McMahon SB, Clary DO, Reichardt LF, Priestley JV. Immunocytochemical localization of trkA receptors in chemically identified subgroups of adult rat sensory neurons. Eur. J Neurosci. 1995;7(7):1484–94. Averill S, McMahon SB, Clary DO, Reichardt LF, Priestley JV. Immunocytochemical localization of trkA receptors in chemically identified subgroups of adult rat sensory neurons. Eur. J Neurosci. 1995;7(7):1484–94.
33.
go back to reference Pezet S, McMahon SB. Neurotrophins: mediators and modulators of pain. Annu.Rev.Neurosci. 2006;29:507–38.CrossRefPubMed Pezet S, McMahon SB. Neurotrophins: mediators and modulators of pain. Annu.Rev.Neurosci. 2006;29:507–38.CrossRefPubMed
34.
go back to reference Ishikawa G, Koya Y, Tanaka H, Nagakura Y. Long-term analgesic effect of a single dose of anti-NGF antibody on pain during motion without notable suppression of joint edema and lesion in a rat model of osteoarthritis. Osteoarthritis. Cartilage. 2015;23(6):925–32.CrossRef Ishikawa G, Koya Y, Tanaka H, Nagakura Y. Long-term analgesic effect of a single dose of anti-NGF antibody on pain during motion without notable suppression of joint edema and lesion in a rat model of osteoarthritis. Osteoarthritis. Cartilage. 2015;23(6):925–32.CrossRef
35.
go back to reference Schnitzer TJ, Marks JAA. Systematic review of the efficacy and general safety of antibodies to NGF in the treatment of OA of the hip or knee. Osteoarthritis. Cartilage. 2015;23(Suppl 1):S8–17.CrossRef Schnitzer TJ, Marks JAA. Systematic review of the efficacy and general safety of antibodies to NGF in the treatment of OA of the hip or knee. Osteoarthritis. Cartilage. 2015;23(Suppl 1):S8–17.CrossRef
36.
go back to reference Manni L, Lundeberg T, Fiorito S, Bonini S, Vigneti E, Aloe L. Nerve growth factor release by human synovial fibroblasts prior to and following exposure to tumor necrosis factor-alpha, interleukin-1 beta and cholecystokinin-8: the possible role of NGF in the inflammatory response. Clin.Exp.Rheumatol. 2003;21(5):617–24.PubMed Manni L, Lundeberg T, Fiorito S, Bonini S, Vigneti E, Aloe L. Nerve growth factor release by human synovial fibroblasts prior to and following exposure to tumor necrosis factor-alpha, interleukin-1 beta and cholecystokinin-8: the possible role of NGF in the inflammatory response. Clin.Exp.Rheumatol. 2003;21(5):617–24.PubMed
37.
go back to reference Takano S, Uchida K, Miyagi M, Inoue G, Fujimaki H, Aikawa J, Iwase D, Minatani A, Iwabuchi K, Takaso M. Nerve growth factor regulation by TNF-alpha and IL-1beta in synovial macrophages and fibroblasts in osteoarthritic mice. J Immunol Res. 2016;2016:5706359.CrossRefPubMedPubMedCentral Takano S, Uchida K, Miyagi M, Inoue G, Fujimaki H, Aikawa J, Iwase D, Minatani A, Iwabuchi K, Takaso M. Nerve growth factor regulation by TNF-alpha and IL-1beta in synovial macrophages and fibroblasts in osteoarthritic mice. J Immunol Res. 2016;2016:5706359.CrossRefPubMedPubMedCentral
38.
go back to reference Ashraf S, Mapp PI, Burston J, Bennett AJ, Chapman V, Walsh DA. Augmented pain behavioural responses to intra-articular injection of nerve growth factor in two animal models of osteoarthritis. Ann.Rheum.Dis. 2014;73(9):1710–8.CrossRefPubMed Ashraf S, Mapp PI, Burston J, Bennett AJ, Chapman V, Walsh DA. Augmented pain behavioural responses to intra-articular injection of nerve growth factor in two animal models of osteoarthritis. Ann.Rheum.Dis. 2014;73(9):1710–8.CrossRefPubMed
39.
go back to reference McNamee KE, Burleigh A, Gompels LL, Feldmann M, Allen SJ, Williams RO, Dawbarn D, Vincent TL, Inglis JJ. Treatment of murine osteoarthritis with TrkAd5 reveals a pivotal role for nerve growth factor in non-inflammatory joint pain. Pain. 2010;149(2):386–92.CrossRefPubMed McNamee KE, Burleigh A, Gompels LL, Feldmann M, Allen SJ, Williams RO, Dawbarn D, Vincent TL, Inglis JJ. Treatment of murine osteoarthritis with TrkAd5 reveals a pivotal role for nerve growth factor in non-inflammatory joint pain. Pain. 2010;149(2):386–92.CrossRefPubMed
40.
go back to reference Xu L, Nwosu LN, Burston JJ, Millns PJ, Sagar DR, Mapp PI, Meesawatsom P, Li L, Bennett AJ, Walsh DA, Chapman V. The anti-NGF antibody muMab 911 both prevents and reverses pain behaviour and subchondral osteoclast numbers in a rat model of osteoarthritis pain. Osteoarthritis. Cartilage. 2016;24(9):1587–95.CrossRef Xu L, Nwosu LN, Burston JJ, Millns PJ, Sagar DR, Mapp PI, Meesawatsom P, Li L, Bennett AJ, Walsh DA, Chapman V. The anti-NGF antibody muMab 911 both prevents and reverses pain behaviour and subchondral osteoclast numbers in a rat model of osteoarthritis pain. Osteoarthritis. Cartilage. 2016;24(9):1587–95.CrossRef
41.
go back to reference Cattaneo A. Tanezumab, a recombinant humanized mAb against nerve growth factor for the treatment of acute and chronic pain. Curr.Opin.Mol.Ther. 2010;12(1):94–106.PubMed Cattaneo A. Tanezumab, a recombinant humanized mAb against nerve growth factor for the treatment of acute and chronic pain. Curr.Opin.Mol.Ther. 2010;12(1):94–106.PubMed
42.
go back to reference Balanescu AR, Feist E, Wolfram G, Davignon I, Smith MD, Brown MT, West CR. 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(9):1665–72.CrossRefPubMed Balanescu AR, Feist E, Wolfram G, Davignon I, Smith MD, Brown MT, West CR. 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(9):1665–72.CrossRefPubMed
Metadata
Title
Efficacy of nerve growth factor antibody in a knee osteoarthritis pain model in mice
Authors
Masayuki Miyagi
Tetsuhiro Ishikawa
Hiroto Kamoda
Miyako Suzuki
Gen Inoue
Yoshihiro Sakuma
Yasuhiro Oikawa
Sumihisa Orita
Kentaro Uchida
Kazuhisa Takahashi
Masashi Takaso
Seiji Ohtori
Publication date
01-12-2017
Publisher
BioMed Central
Published in
BMC Musculoskeletal Disorders / Issue 1/2017
Electronic ISSN: 1471-2474
DOI
https://doi.org/10.1186/s12891-017-1792-x

Other articles of this Issue 1/2017

BMC Musculoskeletal Disorders 1/2017 Go to the issue