1
|
García-Domínguez M. NGF in Neuropathic Pain: Understanding Its Role and Therapeutic Opportunities. Curr Issues Mol Biol 2025; 47:93. [PMID: 39996814 PMCID: PMC11854882 DOI: 10.3390/cimb47020093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/30/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Nerve growth factor (NGF) is one of the essential components that have been implicated in the pathophysiology of neuropathic pain, a condition that develops following nerve injury or dysfunction. This neurotrophin is critical for the survival and maintenance of sensory neurons, and its dysregulation has been implicated in the sensitization of pain pathways. NGF interacts with its receptor TrkA and p75NTR to activate intracellular signaling pathways associated with nociception and the emergence of allodynia and hyperalgesia. Therapeutic approaches employing neutralizing antibodies and molecule inhibitors have been highly effective at both preclinical and clinical levels, hence giving hope again for the use of NGF as an important biomarker and therapeutic target in the management of neuropathic pain. By exploiting the unique properties of NGF and its interactions within the nervous system, new therapeutic modalities could be designed to enhance efficacy while minimizing side effects. In conclusion, taking advantage of the multifaceted dynamics of NGF could provide effective pain management therapies to finally respond to the unmet needs of patients experiencing neuropathic pain.
Collapse
Affiliation(s)
- Mario García-Domínguez
- Program of Immunology and Immunotherapy, CIMA-Universidad de Navarra, 31008 Pamplona, Spain;
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
2
|
Pulskamp TG, Johnson LM, Berlau DJ. Novel non-opioid analgesics in pain management. Pain Manag 2024; 14:641-651. [PMID: 39692452 PMCID: PMC11702995 DOI: 10.1080/17581869.2024.2442292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/11/2024] [Indexed: 12/19/2024] Open
Abstract
Effective pain management has long been hindered by the limitations and risks associated with opioid analgesics, necessitating the exploration of novel, non-opioid alternatives. A comprehensive literature search was conducted using PubMed and Google Scholar during October and November 2024 to identify studies on emerging non-opioid pain management therapeutics. This review evaluates three promising classes of mechanism-specific therapeutics: nerve growth factor (NGF) monoclonal antibodies, transient receptor potential vanilloid 1 (TRPV1) antagonists, and selective sodium channel blockers. By targeting distinct pathways involved in pain sensation, these therapies aim to provide relief for various pain types, including chronic, inflammatory, and neuropathic pain, with potentially fewer side effects. Through a detailed analysis of their mechanisms of action and current evidence, this review highlights the clinical potential of each class, addressing both their efficacy and safety challenges. Ultimately, these emerging therapies represent significant advancements in non-opioid pain management, with the potential to reshape standard approaches to patient care.
Collapse
|
3
|
Morado-Urbina CE, Kato J, Sandor K, Vazquez-Mora JA, Ängeby Möller K, Simon N, Salcido J, Martinez-Martinez A, Munoz-Islas E, Jimenez-Andrade JM, Svensson CI. Sex-dependent effects of the targeted nerve growth factor mutation (R100E) on pain behavior, joint inflammation, and bone erosion in mice. Pain 2024; 165:2814-2828. [PMID: 39324959 PMCID: PMC11562760 DOI: 10.1097/j.pain.0000000000003343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 05/10/2024] [Accepted: 06/07/2024] [Indexed: 09/27/2024]
Abstract
ABSTRACT Nerve growth factor (NGF)-R100E is a mutated form of human recombinant NGF that reduces the binding of NGF to its p75NTR receptor while retaining its affinity toward the TrkA receptor. Here, we used human wild type NGF and NGF-R100E knock-in mice to investigate the effects of this NGF mutation on inflammation-induced pain-related behaviors and bone loss. The hNGF-R100E mutation did not alter the nerve fiber density in the sciatic nerve, ankle joint synovium, and skin of naïve mice. Withdrawal responses to mechanical, thermal, and cold stimuli before and after joint inflammation induced by intra-articular injection of complete Freund adjuvant (CFA) were similar between human recombinant nerve growth factor-wild type and hNGF-R100E male and female mice while weight bearing and gait analysis revealed significant differences. Intriguingly, hNGF-R100E male and female mice showed only mild changes, indicating lower degrees of deep joint-related pain compared to their wild type counterparts. Furthermore, micro-CT analysis demonstrated that hNGF-R100E female mice, but not males, were protected from CFA-induced bone loss, and mRNA analysis showed a different gene regulation indicating a sex-dependent relationship between NGF, inflammation, and bone loss. In conclusion, our study reveals that the hNGF-R100E mutation renders mice insensitive to inflammation-induced impact on joint loading and gait while preserving the development of the peripheral nociceptive neurons and sensitivity to punctate stimulation of the skin. Notably, the mutation uncovers a sex-dependent relationship between NGF and inflammation-induced bone loss. These findings offer valuable insights into NGF as a target for pain management and the interplay between NGF and bone architecture.
Collapse
Affiliation(s)
- Carlos E. Morado-Urbina
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Jungo Kato
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
- Department of Anesthesiology, Keio University School of Medicine, Tokyo, Japan
| | - Katalin Sandor
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Juan Antonio Vazquez-Mora
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Kristina Ängeby Möller
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Nils Simon
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Jaira Salcido
- Unidad Académica Multidisciplinaria Reynosa Aztlán, Universidad Autónoma de Tamaulipas, Reynosa, México
| | - Arisai Martinez-Martinez
- Unidad Académica Multidisciplinaria Reynosa Aztlán, Universidad Autónoma de Tamaulipas, Reynosa, México
| | - Enriqueta Munoz-Islas
- Unidad Académica Multidisciplinaria Reynosa Aztlán, Universidad Autónoma de Tamaulipas, Reynosa, México
| | | | - Camilla I. Svensson
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
4
|
Ritter J, Menger M, Herath SC, Histing T, Kolbenschlag J, Daigeler A, Heinzel JC, Prahm C. Translational evaluation of gait behavior in rodent models of arthritic disorders with the CatWalk device - a narrative review. Front Med (Lausanne) 2023; 10:1255215. [PMID: 37869169 PMCID: PMC10587608 DOI: 10.3389/fmed.2023.1255215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 09/21/2023] [Indexed: 10/24/2023] Open
Abstract
Arthritic disorders have become one of the main contributors to the global burden of disease. Today, they are one of the leading causes of chronic pain and disability worldwide. Current therapies are incapable of treating pain sufficiently and preventing disease progression. The lack of understanding basic mechanisms underlying the initiation, maintenance and progression of arthritic disorders and related symptoms represent the major obstacle in the search for adequate treatments. For a long time, histological evaluation of joint pathology was the predominant outcome parameter in preclinical arthritis models. Nevertheless, quantification of pain and functional limitations analogs to arthritis related symptoms in humans is essential to enable bench to bedside translation and to evaluate the effectiveness of new treatment strategies. As the experience of pain and functional deficits are often associated with altered gait behavior, in the last decades, automated gait analysis has become a well-established tool for the quantitative evaluation of the sequalae of arthritic disorders in animal models. The purpose of this review is to provide a detailed overview on the current literature on the use of the CatWalk gait analysis system in rodent models of arthritic disorders, e.g., Osteoarthritis, Monoarthritis and Rheumatoid Arthritis. Special focus is put on the assessment and monitoring of pain-related behavior during the course of the disease. The capability of evaluating the effect of distinct treatment strategies and the future potential for the application of the CatWalk in rodent models of arthritic disorders is also addressed in this review. Finally, we discuss important consideration and provide recommendations on the use of the CatWalk in preclinical models of arthritic diseases.
Collapse
Affiliation(s)
- Jana Ritter
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Klinik Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Maximilian Menger
- Department of Trauma and Reconstructive Surgery, BG Klinik Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Steven C Herath
- Department of Trauma and Reconstructive Surgery, BG Klinik Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, BG Klinik Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Jonas Kolbenschlag
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Klinik Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Adrien Daigeler
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Klinik Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Johannes C Heinzel
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Klinik Tuebingen, University of Tuebingen, Tuebingen, Germany
- Ludwig Boltzmann Institute for Traumatology - The Research Center in Cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Cosima Prahm
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Klinik Tuebingen, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
5
|
Menges S, Michaelis M, Kleinschmidt-Dörr K. Anti-NGF treatment worsens subchondral bone and cartilage measures while improving symptoms in floor-housed rabbits with osteoarthritis. Front Physiol 2023; 14:1201328. [PMID: 37435308 PMCID: PMC10331818 DOI: 10.3389/fphys.2023.1201328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/14/2023] [Indexed: 07/13/2023] Open
Abstract
Objective: Osteoarthritis (OA) is a common joint disorder often affecting the knee. It is characterized by alterations of various joint tissues including subchondral bone and by chronic pain. Anti-nerve growth factor (NGF) antibodies have demonstrated improvement in pain associated with OA in phase 3 clinical trials but have not been approved due to an increased risk of developing rapidly progressive OA. The aim of this study was to investigate effects of systemic anti-NGF-treatment on structure and symptoms in rabbits with surgically induced joint instability. Methods: This was elicited by anterior cruciate ligament transection and partial resection of the medial meniscus in right knee of 63 female rabbits, housed altogether in a 56 m2 floor husbandry. Rabbits received either 0.1, 1 or 3 mg/kg anti-NGF antibody intra-venously at weeks 1, 5 and 14 after surgery or vehicle. During in-life phase, static incapacitance tests were performed and joint diameter was measured. Following necropsy, gross morphological scoring and micro-computed tomography analysis of subchondral bone and cartilage were performed. Results: After surgery, rabbits unloaded operated joints, which was improved with 0.3 and 3 mg/kg anti-NGF compared to vehicle injection during the first half of the study. The diameter of operated knee joints increased over contralateral measures. This increase was bigger in anti-NGF treated rabbits beginning 2 weeks after the first IV injection and became dose-dependent and more pronounced with time. In the 3 mg/kg anti-NGF group, the bone volume fraction and trabecular thickness increased in the medio-femoral region of operated joints compared to contralateral and to vehicle-treated animals, while cartilage volume and to a lesser extent thickness decreased. Enlarged bony areas were found in right medio-femoral cartilage surfaces of animals receiving 1 and 3 mg/kg anti-NGF. Alterations of all structural parameters were particularly distinct in a subgroup of three rabbits, which also exhibited more prominent symptomatic improvement. Conclusion: This study showed that anti-NGF administration exerted negative impact on structure in destabilized joints of rabbits, while pain-induced unloading of joints was improved. Our findings open up the possibility to better understand the effects of systemic anti-NGF, particularly on subchondral bone, and thus the occurrence of rapidly progressive OA in patients.
Collapse
|
6
|
Koya Y, Tanaka H, Yoshimi E, Takeshita N, Morita S, Morio H, Mori K, Fushiki H, Kamohara M. A novel anti-NGF PEGylated Fab' provides analgesia with lower risk of adverse effects. MAbs 2023; 15:2149055. [PMID: 36458900 PMCID: PMC9721442 DOI: 10.1080/19420862.2022.2149055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Nerve growth factor (NGF) has emerged as a key driver of pain perception in several chronic pain conditions, including osteoarthritis (OA), and plays an important role in the generation and survival of neurons. Although anti-NGF antibodies improve pain control and physical function in patients with clinical chronic pain conditions, anti-NGF IgGs are associated with safety concerns such as effects on fetal and postnatal development and the risk of rapidly progressive osteoarthritis. To overcome these drawbacks, we generated a novel anti-NGF PEGylated Fab' antibody. The anti-NGF PEGylated Fab' showed specific binding to and biological inhibitory activity against NGF, and analgesic effects in adjuvant-induced arthritis model mice in a similar manner to an anti-NGF IgG. In collagen-induced arthritis model mice, the anti-NGF PEGylated Fab' showed higher accumulation in inflamed foot pads than the anti-NGF IgG. In pregnant rats and non-human primates, the anti-NGF PEGylated Fab' was undetectable in fetuses, while the anti-NGF IgG was detected and caused abnormal postnatal development. The PEGylated Fab' and IgG also differed in their ability to form immune complexes in vitro. Additionally, while both PEGylated Fab' and IgG showed analgesic effects in sodium monoiodoacetate-induced arthritic model rats, their effects on edema were surprisingly quite different. While the anti-NGF IgG promoted edema over time, the anti-NGF PEGylated Fab' did not. The anti-NGF PEGylated Fab' (ASP6294) may thus be a potential therapeutic candidate with lower risk of adverse effects for various diseases in which NGF is involved such as OA and chronic back pain.
Collapse
Affiliation(s)
- Yukari Koya
- Drug Discovery Research, Astellas Pharma Inc, Tsukuba, Japan,CONTACT Yukari Koya Astellas Pharma Inc, 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Hirotsugu Tanaka
- Incubation Lab, Astellas Innovation Management LLC, Cambridge, MA, USA
| | - Eiji Yoshimi
- Drug Discovery Research, Astellas Pharma Inc, Tsukuba, Japan
| | | | - Shuji Morita
- Drug Discovery Research, Astellas Pharma Inc, Tsukuba, Japan
| | - Hiroki Morio
- Drug Discovery Research, Astellas Pharma Inc, Tsukuba, Japan
| | - Kanako Mori
- Drug Discovery Research, Astellas Pharma Inc, Tsukuba, Japan
| | - Hiroshi Fushiki
- Drug Discovery Research, Astellas Pharma Inc, Tsukuba, Japan
| | | |
Collapse
|
7
|
Markman JD, Schnitzer TJ, Perrot S, Beydoun SR, Ohtori S, Viktrup L, Yang R, Bramson C, West CR, Verburg KM. Clinical Meaningfulness of Response to Tanezumab in Patients with Chronic Low Back Pain: Analysis From a 56-Week, Randomized, Placebo- and Tramadol-Controlled, Phase 3 Trial. Pain Ther 2022; 11:1267-1285. [PMID: 35962939 PMCID: PMC9633876 DOI: 10.1007/s40122-022-00424-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/28/2022] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION A recent phase 3, randomized, placebo- and tramadol-controlled trial (56-week treatment/24-week safety follow-up) demonstrated efficacy of tanezumab 10 mg in patients with chronic low back pain (CLBP) and a history of inadequate response to standard-of-care analgesics. Here, we report on the clinical meaningfulness of treatment response in this study, focused on secondary measures of pain, interference with daily functions, overall disease status, and satisfaction with treatment. METHODS Patients received placebo (up to week 16; n = 406), subcutaneously administered (SC) tanezumab 5 mg (every 8 weeks; n = 407), SC tanezumab 10 mg (every 8 weeks; n = 407), or orally administered tramadol prolonged-release (100-300 mg/day; n = 605) for 56 weeks. Patient's global assessment of low back pain (PGA-LBP), Brief Pain Inventory-short form (BPI-sf), Treatment Satisfaction Questionnaire for Medication (TSQM), and modified Patient-Reported Treatment Impact (mPRTI) were assessed at weeks 16 and 56. RESULTS At week 16, significant (p < 0.05) improvements over placebo were evident with tanezumab for the PGA-LBP (10 mg) and most BPI-sf (both doses), TSQM (both doses), and mPRTI (both doses) items assessed. Improvements over baseline persisted for the PGA-LBP and BPI-sf at week 56. However, the magnitude of improvements was modestly lower at week 56 relative to week 16. Tramadol did not improve PGA-LBP or BPI-sf scores versus placebo at week 16. Most differences between tanezumab and tramadol at week 56 did not reach the level of statistical significance for all endpoints. CONCLUSIONS The totality of the evidence as captured by measures of pain, interference with daily function, patient overall assessment of disease status, and satisfaction with treatment demonstrates the clinically meaningful benefit of tanezumab for some patients with CLBP compared with placebo. CLINICALTRIALS gov: NCT02528253.
Collapse
Affiliation(s)
- John D Markman
- University of Rochester School of Medicine and Dentistry, 2180 S. Clinton Ave, Rochester, NY, 14618, USA.
| | - Thomas J Schnitzer
- Northwestern University Feinberg School of Medicine, 710 N Lake Shore Drive, Room 1020, Chicago, IL, 60611, USA
| | - Serge Perrot
- Pain Center, Cochin Hospital, INSERM U 987, Paris University, Paris, France
| | - Said R Beydoun
- University of Southern California, 1520 San Pablo Street, Suite 3000, Los Angeles, CA, 90033, USA
| | - Seiji Ohtori
- Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Lars Viktrup
- Eli Lilly & Company, 893 Delaware St, Indianapolis, IN, 46225, USA
| | - Ruoyong Yang
- Pfizer Inc, 235 E 42nd St, New York, NY, 10017, USA
| | | | | | | |
Collapse
|
8
|
Vasconcelos DP, Jabangwe C, Lamghari M, Alves CJ. The Neuroimmune Interplay in Joint Pain: The Role of Macrophages. Front Immunol 2022; 13:812962. [PMID: 35355986 PMCID: PMC8959978 DOI: 10.3389/fimmu.2022.812962] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/15/2022] [Indexed: 12/29/2022] Open
Abstract
Chronic pain associated with joint disorders, such as rheumatoid arthritis (RA), osteoarthritis (OA) and implant aseptic loosening (AL), is a highly debilitating symptom that impacts mobility and quality of life in affected patients. The neuroimmune crosstalk has been demonstrated to play a critical role in the onset and establishment of chronic pain conditions. Immune cells release cytokines and immune mediators that can activate and sensitize nociceptors evoking pain, through interaction with receptors in the sensory nerve terminals. On the other hand, sensory and sympathetic nerve fibers release neurotransmitters that bind to their specific receptor expressed on surface of immune cells, initiating an immunomodulatory role. Macrophages have been shown to be key players in the neuroimmune crosstalk. Moreover, macrophages constitute the dominant immune cell population in RA, OA and AL. Importantly, the targeting of macrophages can result in anti-nociceptive effects in chronic pain conditions. Therefore, the aim of this review is to discuss the nature and impact of the interaction between the inflammatory response and nerve fibers in these joint disorders regarding the genesis and maintenance of pain. The role of macrophages is highlighted. The alteration in the joint innervation pattern and the inflammatory response are also described. Additionally, the immunomodulatory role of sensory and sympathetic neurotransmitters is revised.
Collapse
Affiliation(s)
- Daniela P Vasconcelos
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto- Associação, Porto, Portugal.,Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Clive Jabangwe
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto- Associação, Porto, Portugal.,Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Meriem Lamghari
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto- Associação, Porto, Portugal.,Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,Instituto Ciências Biomédicas Abel Salazar, Universidade de Porto, Porto, Portugal
| | - Cecília J Alves
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto- Associação, Porto, Portugal.,Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| |
Collapse
|
9
|
Gonçalves S, Gowler PR, Woodhams SG, Turnbull J, Hathway G, Chapman V. The challenges of treating osteoarthritis pain and opportunities for novel peripherally directed therapeutic strategies. Neuropharmacology 2022; 213:109075. [DOI: 10.1016/j.neuropharm.2022.109075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/07/2022] [Accepted: 04/21/2022] [Indexed: 12/22/2022]
|
10
|
Han FY, Brockman DA, Nicholson JR, Corradini L, Smith MT. Gait analysis as a robust pain behavioural endpoint in the chronic phase of the monoiodoacetate-induced knee joint pain in the rat. Behav Pharmacol 2022; 33:23-31. [PMID: 35007233 DOI: 10.1097/fbp.0000000000000663] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The monoiodoacetate-induced rat model of osteoarthritis knee pain is widely used. However, there are between-study differences in the pain behavioural endpoints assessed and in the dose of intraarticular monoiodoacetate administered. This study evaluated the robustness of gait analysis as a pain behavioural endpoint in the chronic phase of this model, in comparison with mechanical hyperalgesia in the injected (ipsilateral) joint and development of mechanical allodynia in the ipsilateral hind paws. Groups of Sprague-Dawley rats received a single intraarticular injection of monoiodoacetate at 0.5, 1, 2 or 3 mg or vehicle (saline) into the left (ipsilateral) knee joint. An additional group of rats were not injected (naïve group). The pain behavioural methods used were gait analysis, measurement of pressure algometry thresholds in the ipsilateral knee joints, and assessment of mechanical allodynia in the ipsilateral hind paws using von Frey filaments. These pain behavioural endpoints were assessed premonoiodoacetate injection and for up to 42-days postmonoiodoacetate injection in a blinded manner. Body weights were also assessed as a measure of general health. Good general health was maintained as all rats gained weight at a similar rate for the 42-day study period. In the chronic phase of the model (days 9-42), intraarticular monoiodoacetate at 3 mg evoked robust alterations in multiple gait parameters as well as persistent mechanical allodynia in the ipsilateral hind paws. For the chronic phase of the monoiodoacetate-induced rat model of osteoarthritis knee pain, gait analysis, such as mechanical allodynia in the ipsilateral hind paws, is a robust pain behavioural measure.
Collapse
Affiliation(s)
- Felicity Y Han
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Queensland, Australia
| | - David A Brockman
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Queensland, Australia
| | | | - Laura Corradini
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Maree Therese Smith
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Queensland, Australia
| |
Collapse
|
11
|
Subramanian G, Duclos B, Johnson PD, Williams T, Ross JT, Bowen SJ, Zhu Y, White JA, Hedke C, Huczek D, Collard W, Javens C, Vairagoundar R, Respondek T, Zachary T, Maddux T, Cox MR, Kamerling S, Gonzales AJ. In Pursuit of an Allosteric Human Tropomyosin Kinase A ( hTrkA) Inhibitor for Chronic Pain. ACS Med Chem Lett 2021; 12:1847-1852. [PMID: 34795875 DOI: 10.1021/acsmedchemlett.1c00483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/21/2021] [Indexed: 12/13/2022] Open
Abstract
Human β-nerve growth factor (β-NGF) and its associated receptor, human tropomyosin receptor kinase A (hTrkA), have been demonstrated to be key factors in the perception of pain. However, efficacious small molecule therapies targeting the intracellularly located hTrkA kinase have not been explored thoroughly for pain management. Herein, we report the pharmacological properties of a selective hTrkA allosteric inhibitor, 1. 1 was shown to be active against the full length hTrkA, showing preferential binding for the inactive kinase, and was confirmed through the X-ray of hTrkA···1 bound complex. 1 was also found to inhibit β-NGF induced neurite outgrowth in rat PC12 cells. Daily oral administration of 1 improved the joint compression threshold of rats injected intra-articularly with monoiodoacetate over a 14-day period. The efficacy of 1 in a relevant chronic pain model of osteoarthritis coupled with in vitro confirmation of target mediation makes allosteric hTrkA inhibitors potential candidates for modulating pain.
Collapse
Affiliation(s)
- Govindan Subramanian
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Brian Duclos
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Paul D. Johnson
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Tracey Williams
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Jason T. Ross
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Scott J. Bowen
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Yaqi Zhu
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Julie A. White
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Carolyn Hedke
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Dennis Huczek
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Wendy Collard
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Christopher Javens
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Rajendran Vairagoundar
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Tomasz Respondek
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Theresa Zachary
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Todd Maddux
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Mark R. Cox
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Steven Kamerling
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| | - Andrea J. Gonzales
- Veterinary Medicine Research & Development, Zoetis, 333 Portage Street, Building 300, Kalamazoo, Michigan 49007, United States
| |
Collapse
|
12
|
Magnusdottir R, Gohin S, Ter Heegde F, Hopkinson M, McNally IF, Fisher A, Upton N, Billinton A, Chenu C. Fracture-induced pain-like behaviours in a femoral fracture mouse model. Osteoporos Int 2021; 32:2347-2359. [PMID: 34080043 PMCID: PMC8563675 DOI: 10.1007/s00198-021-05991-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/03/2021] [Indexed: 11/25/2022]
Abstract
UNLABELLED This study is the first comprehensive characterisation of the pain phenotype after fracture using both evoked and naturalistic behaviours in adult male and ovariectomised female mice. It also shows that an anti-nerve growth factor (NGF) therapy could be considered to reduce pain after fracture surgery. INTRODUCTION Bone fractures are common due to the ageing population and very painful even after healing. The phenotype of this pain is still poorly understood. We aimed to characterise it in a femoral fracture model in mice. METHODS We employed both adult male, and female ovariectomised (OVX) mice to mimic osteoporotic fractures. Mice underwent a unilateral femoral fracture maintained by an external fixator or a sham surgery. Pain behaviours, including mechanical and thermal sensitivity, weight bearing and LABORAS, were measured from baseline to 6 weeks after fracture. The effect on pain of an antibody against nerve growth factor (anti-NGF) was assessed. Changes in nerve density at the fracture callus were analysed by immunohistochemistry. RESULTS Following surgery, all groups exhibited high levels of invoked nociception. Mechanical and thermal hyperalgesia were observed from 1 week after surgery, with nociceptive sensitization in the fracture group maintained for the 6 weeks, whereas it resolved in the sham group after 3 weeks. OVX induced reduction in pain thresholds, which was maintained after fracture. The frequency of naturalistic behaviours did not change between groups. Anti-NGF administered before and weekly after surgery alleviated fracture-induced mechanical nociception. The density of nerve fibres in the fracture callus was similar in all groups 6 weeks after surgery. CONCLUSIONS Fractures in rodent models are highly painful in both sexes. This pain-like phenotype is prolonged and should be routinely considered in fracture healing studies as it can affect the study outcome. The anti-NGF alleviates fracture-induced mechanical pain.
Collapse
Affiliation(s)
- R Magnusdottir
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London, NW1 0TU, UK
- Transpharmation Ltd., The London Bioscience Innovation Centre, 2 Royal College Street, London, NW1 0NH, UK
| | - S Gohin
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London, NW1 0TU, UK
| | - F Ter Heegde
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London, NW1 0TU, UK
| | - M Hopkinson
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London, NW1 0TU, UK
| | - I F McNally
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London, NW1 0TU, UK
| | - A Fisher
- Transpharmation Ltd., The London Bioscience Innovation Centre, 2 Royal College Street, London, NW1 0NH, UK
| | - N Upton
- Transpharmation Ltd., The London Bioscience Innovation Centre, 2 Royal College Street, London, NW1 0NH, UK
| | - A Billinton
- Astrazeneca, Neuroscience, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - C Chenu
- Skeletal Biology Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London, NW1 0TU, UK.
| |
Collapse
|
13
|
Han FY, Brockman DA, Nicholson JR, Corradini L, Smith MT. Pharmacological characterization of the chronic phase of the monoiodoacetate-induced rat model of osteoarthritis pain in the knee joint. Clin Exp Pharmacol Physiol 2021; 48:1515-1522. [PMID: 34275162 DOI: 10.1111/1440-1681.13551] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 11/27/2022]
Abstract
For patients with osteoarthritis (OA) of the knee, pain is the most debilitating symptom. Although it has been proposed that the chronic phase of the monoiodoacetate (MIA)-induced rodent model of knee joint pain may be superior to other chronic or acute OA models for assessing the analgesic efficacy of novel molecules, relatively few pharmacological studies have been conducted in the chronic phase of this model. Hence, this study was designed to use pharmacological methods to characterize the chronic phase of the MIA-induced rat model of knee joint OA pain. Rats received a single intraarticular injection of MIA at 2.5 mg or vehicle (saline) into the left (ipsilateral) knee joint. Pain behaviour was assessed by measuring paw withdrawal thresholds (PWTs) in the hindpaws pre-MIA injection and twice-weekly until study completion on day 42. Mechanical allodynia was fully developed in the ipsilateral hindpaws (PWTs ≤6 g) from day 7 and it persisted until day 42. MIA-injected rats with PWTs ≤6 g in the ipsilateral hindpaws received single doses of one of four clinically available drugs that represent four distinct pharmacological classes, viz gabapentin, amitriptyline, meloxicam and morphine, according to a 'washout' protocol with at least 48 hours between successive doses. Gabapentin evoked dose-dependent anti-allodynia as did morphine whereas amitriptyline and meloxicam were inactive. Our findings are aligned with clinical data showing that gabapentin and morphine alleviated OA pain in the knee. The lack of efficacy of amitriptyline is consistent with the loss of descending diffuse noxious inhibitory controls reported by others in this model.
Collapse
Affiliation(s)
- Felicity Y Han
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - David A Brockman
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | | | - Laura Corradini
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Maree T Smith
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
14
|
Sánchez-Robles EM, Girón R, Paniagua N, Rodríguez-Rivera C, Pascual D, Goicoechea C. Monoclonal Antibodies for Chronic Pain Treatment: Present and Future. Int J Mol Sci 2021; 22:ijms221910325. [PMID: 34638667 PMCID: PMC8508878 DOI: 10.3390/ijms221910325] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022] Open
Abstract
Chronic pain remains a major problem worldwide, despite the availability of various non-pharmacological and pharmacological treatment options. Therefore, new analgesics with novel mechanisms of action are needed. Monoclonal antibodies (mAbs) are directed against specific, targeted molecules involved in pain signaling and processing pathways that look to be very effective and promising as a novel therapy in pain management. Thus, there are mAbs against tumor necrosis factor (TNF), nerve growth factor (NGF), calcitonin gene-related peptide (CGRP), or interleukin-6 (IL-6), among others, which are already recommended in the treatment of chronic pain conditions such as osteoarthritis, chronic lower back pain, migraine, or rheumatoid arthritis that are under preclinical research. This narrative review summarizes the preclinical and clinical evidence supporting the use of these agents in the treatment of chronic pain.
Collapse
|
15
|
Bimonte S, Cascella M, Forte CA, Esposito G, Cuomo A. The Role of Anti-Nerve Growth Factor Monoclonal Antibodies in the Control of Chronic Cancer and Non-Cancer Pain. J Pain Res 2021; 14:1959-1967. [PMID: 34234542 PMCID: PMC8253925 DOI: 10.2147/jpr.s302004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022] Open
Abstract
Nerve growth factor (NGF) belongs to the neurotrophin family and plays a fundamental role in the endurance of sensory and sympathetic neurons during embryogenesis. NGF, by interacting with tropomyosin receptor kinase A receptor (TrkA), modulates the pain pathway through the enhancement of the neurotrophic and nociceptor functions. Moreover, it has been demonstrated that NGF is upregulated in patients with chronic pain syndromes, which are difficult to treat. Thus, new non-pharmacological approaches, based on the use of different species-specific monoclonal antibodies (mAbs) targeting the NGF pathway, have been tested for the treatment of chronic pain in preclinical and clinical studies. With regard to preclinical investigations, anti-NGF mAbs have been used for the management of osteoarthritis (OA) and chronic low back pain animal models, with encouraging results. Moreover, anti-NGF mAb therapy is effective in animal models of neuropathic cancer pain. As regards patients with OA, although phase II and phase III clinical trials with tanezumab led to pain reduction, the safety was not observed in all these patients. Here, we review the preclinical and clinical studies on anti-NGF mAb therapy in chronic syndromes, dissect the role of NGF in pain transduction, and highlight the use of anti-NGF mAbs in humans.
Collapse
Affiliation(s)
- Sabrina Bimonte
- Division of Anesthesia and Pain Medicine, Istituto Nazionale dei Tumori, IRCCS Fondazione G. Pascale, Naples, Italy
| | - Marco Cascella
- Division of Anesthesia and Pain Medicine, Istituto Nazionale dei Tumori, IRCCS Fondazione G. Pascale, Naples, Italy
| | - Cira Antonietta Forte
- Division of Anesthesia and Pain Medicine, Istituto Nazionale dei Tumori, IRCCS Fondazione G. Pascale, Naples, Italy
| | - Gennaro Esposito
- Division of Anesthesia and Pain Medicine, Istituto Nazionale dei Tumori, IRCCS Fondazione G. Pascale, Naples, Italy
| | - Arturo Cuomo
- Division of Anesthesia and Pain Medicine, Istituto Nazionale dei Tumori, IRCCS Fondazione G. Pascale, Naples, Italy
| |
Collapse
|
16
|
Yu Y, Lu ST, Sun JP, Zhou W. Safety of Low-Dose Tanezumab in the Treatment of Hip or Knee Osteoarthritis: A Systemic Review and Meta-analysis of Randomized Phase III Clinical Trials. PAIN MEDICINE 2021; 22:585-595. [PMID: 33141224 DOI: 10.1093/pm/pnaa260] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES To evaluate the safety of low-dose tanezumab in the treatment of knee or hip osteoarthritis (OA). METHODS Databases were searched up to September 2019 for phase III randomized controlled trials (RCTs). Eleven phase III RCTs comprising 11,455 patients were eligible. The pooled estimates of safety outcomes were assessed and expressed using relative risks (RRs) and 95% confidence intervals with a random-effects model. RESULTS Tanezumab significantly increased the incidence of rapidly progressive OA (RPOA; RR = 9.07, 95% CI = 1.21-67.90, P = 0.03) and abnormal peripheral sensation (APS; RR = 2.68, 95% CI = 1.64-4.37, P < 0.00001) compared with placebo. No significant difference was found in terms of incidence of total joint replacement (TJR; RR = 1.13, 95% CI = 0.76-1.68, P = 0.55) or withdrawal due to adverse effects (AEs; RR = 1.26, 95% CI = 0.79-2.00, P = 0.33). The tanezumab group showed a statistically higher incidence of RPOA (RR = 3.96, 95% CI = 2.23-7.04, P < 0.00001) and APS (RR = 1.2, 95% CI = 1.44-2.56, P < 0.00001) compared with the nonsteroidal anti-inflammatory drugs and opioids group. No significant difference was found in terms of TJR (RR = 1.51, 95% CI = 0.65-3.47, P = 0.33) and withdrawal (RR = 0.54, 95% CI = 0.20-1.40, P = 0.20). Subgroup analysis revealed that 2.5 mg of tanezumab did not show an advantage over 5 mg of tanezumab in reducing AEs. CONCLUSIONS These results demonstrate that RPOA and APS should be the most concerning AEs when using tanezumab in OA patients. Additional data are needed to define the optimal dose to minimize risk and to determine the optimal subjects to receive this treatment.
Collapse
Affiliation(s)
- Yang Yu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Shi-Tao Lu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jin-Peng Sun
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Wei Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
17
|
Chun JM, Lee AY, Nam JY, Lim KS, Choe MS, Lee MY, Kim C, Kim JS. Effects of Dipsacus asperoides Extract on Monosodium Iodoacetate-Induced Osteoarthritis in Rats Based on Gene Expression Profiling. Front Pharmacol 2021; 12:615157. [PMID: 33927614 PMCID: PMC8076797 DOI: 10.3389/fphar.2021.615157] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/02/2021] [Indexed: 12/24/2022] Open
Abstract
The root of Dipsacus asperoides C. Y. Cheng et T. M. Ai is traditionally used as an analgesic and anti-inflammatory agent to treat pain, rheumatoid arthritis, and bone fractures. However, neither its effects on osteoarthritis (OA) nor its effects on the arthritic cartilage tissue transcriptome have not been fully investigated. In this study, we used a rat model of monosodium iodoacetate- (MIA-) induced OA to investigate the therapeutic effects of a Dipsacus asperoides ethanolic extract (DAE, 200 mg/kg for 21 days). The study first assessed joint diameter, micro-CT scans, and histopathological analysis and then conducted gene expression profiling using RNA sequencing in articular cartilage tissue. We found that DAE treatment ameliorates OA disease phenotypes; it reduced the knee joint diameter and prevented changes in the structural and histological features of the joint, thereby showing that DAE has a protective effect against OA. Based on the results of gene expression profiling and subsequent pathway analysis, we found that several canonical pathways were linked to DAE treatment, including WNT/β-catenin signaling. Taken together, the present results suggest molecular mechanism, involving gene expression changes, by which DAE has a protective effect in a rat model of MIA-induced OA.
Collapse
Affiliation(s)
- Jin Mi Chun
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju-si, Korea
| | - A Yeong Lee
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju-si, Korea
| | - Jae Yong Nam
- Bioinformatics Group, R&D Center, Insilicogen Corporation, Yongin, Korea
| | - Kyung Seob Lim
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Korea
| | - Mu Seog Choe
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu, Korea
| | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu, Korea
| | - Chul Kim
- Korea Future Medicine Division, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - Joong-Sun Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju-si, Korea
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Osteoarthritis is a degenerative joint disease that features pain as a hallmark symptom. This review summarises progress and obstacles in our understanding of pain mechanisms in arthritis. RECENT FINDINGS Pain phenotypes in osteoarthritis are poorly characterized in clinical studies and animal studies are largely carti-centric. Different animal models incur variable disease progression patterns and activation of distinct pain pathways, but studies reporting both structural and pain outcomes permit better translational insights. In patients, classification of osteoarthritis disease severity is only based on structural integrity of the joint, but pain outcomes do not consistently correlate with joint damage. The complexity of this relationship underlines the need for pain detection in criteria for osteoarthritis classification and patient-reported outcome measures. SUMMARY Variable inflammatory and neuropathic components and spatiotemporal evolution underlie the heterogeneity of osteoarthritis pain phenotypes, which must be considered to adequately stratify patients. Revised classification of osteoarthritis at different stages encompassing both structural and pain outcomes would significantly improve detection and diagnosis at both early and late stages of disease. These are necessary advancements in the field that would also improve trial design and provide better understanding of basic mechanisms of disease progression and pain in osteoarthritis.
Collapse
|
19
|
Zhang X, Yang J, Cheng B, Zhao S, Li Y, Kang H, Chen S. Magnetic nanocarriers as a therapeutic drug delivery strategy for promoting pain-related motor functions in a rat model of cartilage transplantation. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:37. [PMID: 33787997 PMCID: PMC8012338 DOI: 10.1007/s10856-021-06508-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 03/09/2021] [Indexed: 05/11/2023]
Abstract
Cartilage is an avascular tissue with low cellularity and insufficient self-repair response. In clinical practice, a large articular cartilage defect is usually fixed by cartilage transplantation. Importantly, the fast repair process has been demanded postoperatively in the area between the host cartilage and the transplanted cartilage. In the past few years, magnetic nanoparticles have drawn great attention due to their biocompatible, biodegradable, and nontoxic properties. In addition, the nanoparticles can easily pass through the cell plasma membrane and increase the cellular uptake efficiency. Here, a therapeutic drug delivery strategy was proposed for cartilage repair. The prepared kartogenin (KGN)-conjugated magnetic nanocarriers (KGN@NCs) promoted the viability of chondrocytes in vitro. In a rat model of cartilage transplantation, intra-articularly delivered KGN@NCs generated cartilage with a flat surface and a high level of aggrecan in vivo. Notably, KGN@NCs were also capable of improving the pain-related motor functions. They promoted the motor functional parameters including the print area and intensity to restore to a normal level compared with the single KGN. Therefore, these therapeutic drug nanocarriers provided the potential for cartilage repair.
Collapse
Affiliation(s)
- Xingyu Zhang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jianjun Yang
- Department of Orthopaedics, Tenth People's Hospital, Tongji University, Shanghai, 200072, China.
| | - Baochang Cheng
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai, 200433, China
| | - Shenli Zhao
- Department of Joint Surgery, Yangpu Hospital Affiliated to Tongji University, Shanghai, 200082, China
| | - Yao Li
- Department of Orthopaedics, Tenth People's Hospital, Nanjing Medical University, Shanghai, 200072, China
| | - Hui Kang
- Department of Orthopaedics, Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
20
|
Tian Y, Onodera T, Terkawi MA, Iwasaki K, Hishimura R, Liang D, Miyazaki T, Iwasaki N. Local Administration of Low-Dose Nerve Growth Factor Antibody Reduced Pain in a Rat Osteoarthritis Model. Int J Mol Sci 2021; 22:ijms22052552. [PMID: 33806315 PMCID: PMC7962042 DOI: 10.3390/ijms22052552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/22/2021] [Accepted: 02/26/2021] [Indexed: 11/23/2022] Open
Abstract
Systemic injection of a nerve growth factor (NGF) antibody has been proven to have a significant relevance in relieving osteoarthritis (OA) pain, while its adverse effects remain a safety concern for patients. A local low-dose injection is thought to minimize adverse effects. In this study, OA was induced in an 8-week-old male Sprague–Dawley (SD) rat joint by monoiodoacetate (MIA) injection for 2 weeks, and the effect of weekly injections of low-dose (1, 10, and 100 µg) NGF antibody or saline (control) was evaluated. Behavioral tests were performed, and at the end of week 6, all rats were sacrificed and their knee joints were collected for macroscopic and histological evaluations. Results showed that 100 µg NGF antibody injection relieved pain in OA rats, as evidenced from improved weight-bearing performance but not allodynia. In contrast, no significant differences were observed in macroscopic and histological scores between rats from different groups, demonstrating that intra-articular treatment does not worsen OA progression. These results suggest that local administration yielded a low effective NGF antibody dose that may serve as an alternative approach to systemic injection for the treatment of patients with OA.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (Y.T.); (M.A.T.); (K.I.); (D.L.); (T.M.); (N.I.)
| | - Tomohiro Onodera
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (Y.T.); (M.A.T.); (K.I.); (D.L.); (T.M.); (N.I.)
- Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 060-0808, Japan
- Correspondence: ; Tel.: +81-11-706-5935; Fax: +81-11-706-6054
| | - Mohamad Alaa Terkawi
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (Y.T.); (M.A.T.); (K.I.); (D.L.); (T.M.); (N.I.)
- Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 060-0808, Japan
| | - Koji Iwasaki
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (Y.T.); (M.A.T.); (K.I.); (D.L.); (T.M.); (N.I.)
| | - Ryosuke Hishimura
- Department of Orthopedic Surgery, Hokkaido University Hospital, Kita14, Nishi5, Kita-Ku, Sapporo 060-8648, Japan;
| | - Dawei Liang
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (Y.T.); (M.A.T.); (K.I.); (D.L.); (T.M.); (N.I.)
| | - Takuji Miyazaki
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (Y.T.); (M.A.T.); (K.I.); (D.L.); (T.M.); (N.I.)
| | - Norimasa Iwasaki
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; (Y.T.); (M.A.T.); (K.I.); (D.L.); (T.M.); (N.I.)
- Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 060-0808, Japan
| |
Collapse
|
21
|
Chun JM, Lee AY, Nam JY, Lee MY, Choe MS, Lim KS, Kim C, Kim JS. Protective effects of Phlomis umbrosa extract on a monosodium iodoacetate-induced osteoarthritis model and prediction of molecular mechanisms using transcriptomics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 81:153429. [PMID: 33310311 DOI: 10.1016/j.phymed.2020.153429] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 11/19/2020] [Accepted: 11/27/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Phlomis umbrosa Turczaninow root has been traditionally used to treat fractures, rheumatoid arthritis, and arthralgia. However, the effects and mechanisms of P. umbrosa on osteoarthritis (OA) remain poorly understood and a functional genomic approach has not been investigated. AIM The purpose of this study was to investigate the effects and mechanisms of P. umbrosa extract (PUE) on OA using transcriptomic analysis. METHODS We performed joint diameter measurements, micro computed tomography, and histopathological analysis of monosodium iodoacetate (MIA)-induced OA rats treated with PUE (200 mg/kg) for 3 weeks. Gene expression profiling in articular cartilage tissue was then performed using RNA sequencing (RNA-seq) followed by signaling pathway analysis of regulatory genes. RESULTS PUE treatment improved OA based on decreased joint diameter, increased joint morphological parameters, and histopathological features. Many genes involved in multiple signal transduction pathway and collagen activation in OA were differentially regulated by PUE. These included genes related to Wnt/β-catenin, OA pathway, and sonic hedgehog signaling activity. Furthermore, PUE treatment downregulated cartilage damage factors (MMP-9, MMP-13, ADAMTs4, and ADMATs5) and upregulated chondrogenesis (COL2A1 and SOX-9) by regulating the transcription factors SOX-9, Ctnnb1, and Epas1. CONCLUSION Based on the results of gene expression profiling, this study highlighted the molecular mechanisms underlying the effects of PUE in MIA-induced OA rats. The findings provide novel insight into the mechanisms by which PUE treatment-induced gene expression changes may influence OA disease progression. Taken together, the results suggest that PUE may be used as a source of therapeutic agents for OA.
Collapse
Affiliation(s)
- Jin Mi Chun
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Republic of Korea; Department of Life Systems, Sookmyung Women's University, Seoul, Republic of Korea
| | - A Yeong Lee
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Republic of Korea
| | - Jae Yong Nam
- Bioinformatics Group, R&D Center, Insilicogen Corporation, Yongin, Republic of Korea
| | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Mu Seog Choe
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Kyung Seob Lim
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Chungbuk, Republic of Korea
| | - Chul Kim
- Future Medicine Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea.
| | - Joong-Sun Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Republic of Korea.
| |
Collapse
|
22
|
Abboud C, Duveau A, Bouali-Benazzouz R, Massé K, Mattar J, Brochoire L, Fossat P, Boué-Grabot E, Hleihel W, Landry M. Animal models of pain: Diversity and benefits. J Neurosci Methods 2020; 348:108997. [PMID: 33188801 DOI: 10.1016/j.jneumeth.2020.108997] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 12/15/2022]
Abstract
Chronic pain is a maladaptive neurological disease that remains a major health problem. A deepening of our knowledge on mechanisms that cause pain is a prerequisite to developing novel treatments. A large variety of animal models of pain has been developed that recapitulate the diverse symptoms of different pain pathologies. These models reproduce different pain phenotypes and remain necessary to examine the multidimensional aspects of pain and understand the cellular and molecular basis underlying pain conditions. In this review, we propose an overview of animal models, from simple organisms to rodents and non-human primates and the specific traits of pain pathologies they model. We present the main behavioral tests for assessing pain and investing the underpinning mechanisms of chronic pathological pain. The validity of animal models is analysed based on their ability to mimic human clinical diseases and to predict treatment outcomes. Refine characterization of pathological phenotypes also requires to consider pain globally using specific procedures dedicated to study emotional comorbidities of pain. We discuss the limitations of pain models when research findings fail to be translated from animal models to human clinics. But we also point to some recent successes in analgesic drug development that highlight strategies for improving the predictive validity of animal models of pain. Finally, we emphasize the importance of using assortments of preclinical pain models to identify pain subtype mechanisms, and to foster the development of better analgesics.
Collapse
Affiliation(s)
- Cynthia Abboud
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France; Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France; Faculty of Arts and Sciences, Holy Spirit University of Kaslik (USEK), Lebanon
| | - Alexia Duveau
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Rabia Bouali-Benazzouz
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Karine Massé
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Joseph Mattar
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Lebanon
| | - Louison Brochoire
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Pascal Fossat
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Eric Boué-Grabot
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Walid Hleihel
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Lebanon; Faculty of Arts and Sciences, Holy Spirit University of Kaslik (USEK), Lebanon
| | - Marc Landry
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France.
| |
Collapse
|
23
|
Alves CJ, Couto M, Sousa DM, Magalhães A, Neto E, Leitão L, Conceição F, Monteiro AC, Ribeiro-da-Silva M, Lamghari M. Nociceptive mechanisms driving pain in a post-traumatic osteoarthritis mouse model. Sci Rep 2020; 10:15271. [PMID: 32943744 PMCID: PMC7499425 DOI: 10.1038/s41598-020-72227-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/20/2020] [Indexed: 11/29/2022] Open
Abstract
In osteoarthritis (OA), pain is the dominant clinical symptom, yet the therapeutic approaches remain inadequate. The knowledge of the nociceptive mechanisms in OA, which will allow to develop effective therapies for OA pain, is of utmost need. In this study, we investigated the nociceptive mechanisms involved in post-traumatic OA pain, using the destabilization of the medial meniscus (DMM) mouse model. Our results revealed the development of peripheral pain sensitization, reflected by augmented mechanical allodynia. Along with the development of pain behaviour, we observed an increase in the expression of calcitonin gene-related peptide (CGRP) in both the sensory nerve fibers of the periosteum and the dorsal root ganglia. Interestingly, we also observed that other nociceptive mechanisms commonly described in non-traumatic OA phenotypes, such as infiltration of the synovium by immune cells, neuropathic mechanisms and also central sensitization were not present. Overall, our results suggest that CGRP in the sensory nervous system is underlying the peripheral sensitization observed after traumatic knee injury in the DMM model, highlighting the CGRP as a putative therapeutic target to treat pain in post-traumatic OA. Moreover, our findings suggest that the nociceptive mechanisms involved in driving pain in post-traumatic OA are considerably different from those in non-traumatic OA.
Collapse
Affiliation(s)
- C J Alves
- Neuro-Skeletal Circuits Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal. .,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.
| | - M Couto
- Neuro-Skeletal Circuits Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - D M Sousa
- Neuro-Skeletal Circuits Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - A Magalhães
- Neuro-Skeletal Circuits Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - E Neto
- Neuro-Skeletal Circuits Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - L Leitão
- Neuro-Skeletal Circuits Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto Ciências Biomédicas Abel Salazar (ICBAS), Universidade de Porto, Porto, Portugal
| | - F Conceição
- Neuro-Skeletal Circuits Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto Ciências Biomédicas Abel Salazar (ICBAS), Universidade de Porto, Porto, Portugal
| | - A C Monteiro
- Neuro-Skeletal Circuits Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - M Ribeiro-da-Silva
- Neuro-Skeletal Circuits Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Medicina, Universidade do Porto (FMUP), Porto, Portugal.,Serviço de Ortopedia e Traumatologia, Centro Hospitalar São João, Porto, Portugal
| | - M Lamghari
- Neuro-Skeletal Circuits Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto Ciências Biomédicas Abel Salazar (ICBAS), Universidade de Porto, Porto, Portugal
| |
Collapse
|
24
|
Nerve growth factor antibody for the treatment of osteoarthritis pain and chronic low-back pain: mechanism of action in the context of efficacy and safety. Pain 2020; 160:2210-2220. [PMID: 31145219 PMCID: PMC6756297 DOI: 10.1097/j.pain.0000000000001625] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Chronic pain continues to be a significant global burden despite the availability of a variety of nonpharmacologic and pharmacologic treatment options. Thus, there is a need for new analgesics with novel mechanisms of action. In this regard, antibodies directed against nerve growth factor (NGF-Abs) are a new class of agents in development for the treatment of chronic pain conditions such as osteoarthritis and chronic low-back pain. This comprehensive narrative review summarizes evidence supporting pronociceptive functions for NGF that include contributing to peripheral and central sensitization through tropomyosin receptor kinase A activation and stimulation of local neuronal sprouting. The potential role of NGF in osteoarthritis and chronic low-back pain signaling is also examined to provide a mechanistic basis for the observed efficacy of NGF-Abs in clinical trials of these particular pain states. Finally, the safety profile of NGF-Abs in terms of common adverse events, joint safety, and nerve structure/function is discussed.
Collapse
|
25
|
Barker PA, Mantyh P, Arendt-Nielsen L, Viktrup L, Tive L. Nerve Growth Factor Signaling and Its Contribution to Pain. J Pain Res 2020; 13:1223-1241. [PMID: 32547184 PMCID: PMC7266393 DOI: 10.2147/jpr.s247472] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Nerve growth factor (NGF) is a neurotrophic protein essential for the growth, differentiation, and survival of sympathetic and sensory afferent neurons during development. A substantial body of evidence, based on both animal and human studies, demonstrates that NGF plays a pivotal role in modulation of nociception in adulthood. This has spurred development of a variety of novel analgesics that target the NGF signaling pathway. Here, we present a narrative review designed to summarize how NGF receptor activation and downstream signaling alters nociception through direct sensitization of nociceptors at the site of injury and changes in gene expression in the dorsal root ganglion that collectively increase nociceptive signaling from the periphery to the central nervous system. This review illustrates that NGF has a well-known and multifunctional role in nociceptive processing, although the precise signaling pathways downstream of NGF receptor activation that mediate nociception are complex and not completely understood. Additionally, much of the existing knowledge derives from studies performed in animal models and may not accurately represent the human condition. However, available data establish a role for NGF in the modulation of nociception through effects on the release of inflammatory mediators, nociceptive ion channel/receptor activity, nociceptive gene expression, and local neuronal sprouting. The role of NGF in nociception and the generation and/or maintenance of chronic pain has led to it becoming a novel and attractive target of pain therapeutics for the treatment of chronic pain conditions.
Collapse
Affiliation(s)
- Philip A Barker
- Department of Biology, University of British Columbia, Kelowna, BC, Canada
| | - Patrick Mantyh
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Lars Arendt-Nielsen
- Department of Health Science and Technology and the Center for Sensory-Motor Interaction/Center for Neuroplasticity and Pain, Aalborg University, Aalborg, Denmark
| | | | | |
Collapse
|
26
|
An update on targets for treating osteoarthritis pain: NGF and TRPV1. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2020; 6:129-145. [PMID: 34178580 DOI: 10.1007/s40674-020-00146-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Purpose of review a)Osteoarthritis (OA) is the most common form of arthritis, and pain is the primary symptom of the disease, yet analgesic options for treating OA pain remain limited. In this review, we aimed to give an update on the current clinical and preclinical studies targeting two pathways that are being investigated for treating OA pain: the nerve growth factor (NGF) pathway and the transient receptor potential vanilloid-1 (TRPV1) pathway. Recent findings b)Antibodies against NGF, small molecule inhibitors of TrkA, TRPV1 agonists, and TRPV1 antagonists are all in different stages of clinical and pre-clinical testing for the treatment of OA pain. NGF antibodies have shown efficacy in the primary endpoints tested compared to placebo, however, rapidly progressive OA has been consistently observed in a subset of patients and the cause remains unclear. TRPV1 agonists have also demonstrated reduced pain with no serious adverse events - the most common adverse events include a burning or warming sensation upon administration. Summary c)Targeting the NGF and TRPV1 pathways appear effective for reducing OA pain, but further work is needed to better understand which patients may benefit most from these treatments. The anti-NGF antibody tanezumab and the TRPV1 agonist CNTX-4975 have both received fast-track designation from the FDA for the treatment of OA pain.
Collapse
|
27
|
Malfait AM, Miller RE, Block JA. Targeting neurotrophic factors: Novel approaches to musculoskeletal pain. Pharmacol Ther 2020; 211:107553. [PMID: 32311372 DOI: 10.1016/j.pharmthera.2020.107553] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022]
Abstract
Chronic pain represents a substantial unmet medical need globally. In recent years, the quest for a new generation of novel, safe, mechanism-based analgesic treatments has focused on neurotrophic factors, a large group of secreted proteins that control the growth and survival of different populations of neurons, but that postnatally are involved in the genesis and maintenance of pain, with biological activity in both the periphery and the central nervous system. In this narrative review, we discuss the two families of neurotrophic proteins that have been extensively studied for their role in pain: first, the neurotrophins, nerve growth factor (NGF) and brain-derived growth factor (BDNF), and secondly, the GDNF family of ligands (GFLs). We provide an overview of the pain pathway, and the pain-producing effects of these different proteins. We summarize accumulating preclinical and clinical findings with a focus on musculoskeletal pain, and on osteoarthritis in particular, because the musculoskeletal system is the most prevalent source of chronic pain and of disability, and clinical testing of these novel agents - often biologics- is most advanced in this area.
Collapse
Affiliation(s)
- Anne-Marie Malfait
- Division of Rheumatology, Rush University Medical Center, 1611 W Harrison Street, Suite 510, Chicago, IL 60612, United States of America
| | - Rachel E Miller
- Division of Rheumatology, Rush University Medical Center, 1611 W Harrison Street, Suite 510, Chicago, IL 60612, United States of America
| | - Joel A Block
- Division of Rheumatology, Rush University Medical Center, 1611 W Harrison Street, Suite 510, Chicago, IL 60612, United States of America.
| |
Collapse
|
28
|
McDougall JJ. Osteoarthritis is a neurological disease – an hypothesis. OSTEOARTHRITIS AND CARTILAGE OPEN 2019; 1:100005. [DOI: 10.1016/j.ocarto.2019.100005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/17/2019] [Indexed: 12/20/2022] Open
|
29
|
da Silva JT, Evangelista BG, Venega RA, Seminowicz DA, Chacur M. Anti-NGF treatment can reduce chronic neuropathic pain by changing peripheral mediators and brain activity in rats. Behav Pharmacol 2019; 30:79-88. [DOI: 10.1097/fbp.0000000000000422] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
30
|
Majuta LA, Mitchell SA, Kuskowski MA, Mantyh PW. Anti-nerve growth factor does not change physical activity in normal young or aging mice but does increase activity in mice with skeletal pain. Pain 2018; 159:2285-2295. [PMID: 29994990 PMCID: PMC6233725 DOI: 10.1097/j.pain.0000000000001330] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Anti-nerve growth factor (anti-NGF) therapy has shown significant promise in attenuating several types of skeletal pain. However, whether anti-NGF therapy changes the level of physical activity in individuals with or without skeletal pain is largely unknown. Here, automated day/night activity boxes monitored the effects of anti-NGF treatment on physical activity in normal young (3 months old) and aging (18-23 months old) mice and mice with bone fracture pain. Although aging mice were clearly less active and showed loss of bone mass compared with young mice, anti-NGF treatment had no effect on any measure of day/night activity in either the young or aging mice. By contrast, in mice with femoral fracture pain, anti-NGF treatment produced a clear increase (10%-27%) in horizontal activity, vertical rearing, and velocity of travel compared with the Fracture + Vehicle group. These results suggest, just as in humans, mice titrate their level of physical activity to their level of skeletal pain. The level of skeletal pain may in part be determined by the level of free NGF that seems to rise after injury but not normal aging of the skeleton. In terms of bone healing, animals that received anti-NGF showed an increase in the size of calcified callus but no increase in the number of displaced fractures or time to cortical union. As physical activity is the best nondrug treatment for many patients with skeletal pain, anti-NGF may be useful in reducing pain and promoting activity in these patients.
Collapse
Affiliation(s)
- Lisa A. Majuta
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724
| | | | | | - Patrick W. Mantyh
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724
- Cancer Center, University of Arizona, Tucson, AZ 85724
| |
Collapse
|
31
|
Enomoto M, Mantyh PW, Murrell J, Innes JF, Lascelles BDX. Anti-nerve growth factor monoclonal antibodies for the control of pain in dogs and cats. Vet Rec 2018; 184:23. [PMID: 30368458 PMCID: PMC6326241 DOI: 10.1136/vr.104590] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/10/2018] [Accepted: 09/19/2018] [Indexed: 12/13/2022]
Abstract
Nerve growth factor (NGF) is essential for the survival of sensory and sympathetic neurons during development. However, in the adult, NGF and its interaction with tropomyosin receptor kinase A receptor (TrkA) has been found to play a critical role in nociception and nervous system plasticity in pain conditions. Thus, various monoclonal antibody (mAb) therapies targeting this pathway have been investigated in the development of new pharmacotherapies for chronic pain. Although none of the mAbs against NGF are yet approved for use in humans, they look very promising for the effective control of pain. Recently, species-specific anti-NGF mAbs for the management of osteoarthritis (OA)-associated pain in dogs and cats has been developed, and early clinical trials have been conducted. Anti-NGF therapy looks to be both very effective and very promising as a novel therapy against chronic pain in dogs and cats. This review outlines the mechanism of action of NGF, the role of NGF in osteoarthritis, research in rodent OA models and the current status of the development of anti-NGF mAbs in humans. Furthermore, we describe and discuss the recent development of species-specific anti-NGF mAbs for the treatment of OA-associated pain in veterinary medicine.
Collapse
Affiliation(s)
- Masataka Enomoto
- Translational Research in Pain, Comparative Pain Research and Education Centre, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Patrick W Mantyh
- Cancer Center's Cancer Biology Program, Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Joanna Murrell
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | | | - B Duncan X Lascelles
- Translational Research in Pain, Comparative Pain Research and Education Centre, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA.,Comparative Medicine Institute, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA.,Center for Pain Research and Innovation, UNC School of Dentistry, Chapel Hill, North Carolina, USA.,Center for Translational Pain Research, Department of Anesthesiology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
32
|
Abstract
Chronic pain is a significant problem worldwide and is the most common disability in the United States. It is well known that the immune system plays a critical role in the development and maintenance of many chronic pain conditions. The involvement of the immune system can be through the release of autoantibodies, in the case of rheumatoid arthritis, or via cytokines, chemokines, and other inflammatory mediators (i.e. substance P, histamine, bradykinin, tumor necrosis factor, interleukins, and prostaglandins). Immune cells, such as T cells, B cells and their antibodies, and microglia are clearly key players in immune-related pain. The purpose of this review is to briefly discuss the immune system involvement in pain and to outline how it relates to rheumatoid arthritis, osteoarthritis, fibromyalgia, complex regional pain syndrome, multiple sclerosis, and diabetic neuropathy. The immune system plays a major role in many debilitating chronic pain conditions and we believe that animal models of disease and their treatments should be more directly focused on these interactions.
Collapse
Affiliation(s)
- Stacie K Totsch
- Department of Psychology, University of Alabama at Birmingham, Birmingham, USA
| | | |
Collapse
|
33
|
Upadhyay J, Geber C, Hargreaves R, Birklein F, Borsook D. A critical evaluation of validity and utility of translational imaging in pain and analgesia: Utilizing functional imaging to enhance the process. Neurosci Biobehav Rev 2018; 84:407-423. [PMID: 28807753 PMCID: PMC5729102 DOI: 10.1016/j.neubiorev.2017.08.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/22/2017] [Accepted: 08/04/2017] [Indexed: 02/07/2023]
Abstract
Assessing clinical pain and metrics related to function or quality of life predominantly relies on patient reported subjective measures. These outcome measures are generally not applicable to the preclinical setting where early signs pointing to analgesic value of a therapy are sought, thus introducing difficulties in animal to human translation in pain research. Evaluating brain function in patients and respective animal model(s) has the potential to characterize mechanisms associated with pain or pain-related phenotypes and thereby provide a means of laboratory to clinic translation. This review summarizes the progress made towards understanding of brain function in clinical and preclinical pain states elucidated using an imaging approach as well as the current level of validity of translational pain imaging. We hypothesize that neuroimaging can describe the central representation of pain or pain phenotypes and yields a basis for the development and selection of clinically relevant animal assays. This approach may increase the probability of finding meaningful new analgesics that can help satisfy the significant unmet medical needs of patients.
Collapse
Affiliation(s)
| | - Christian Geber
- Department of Neurology, University Medical Centre Mainz, Mainz, Germany; DRK Schmerz-Zentrum Mainz, Mainz, Germany
| | - Richard Hargreaves
- Center for Pain and the Brain, United States; Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, Boston Harvard Medical School, Boston, MA 02115, United States
| | - Frank Birklein
- Department of Neurology, University Medical Centre Mainz, Mainz, Germany
| | - David Borsook
- Center for Pain and the Brain, United States; Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, Boston Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
34
|
Miyagi M, Ishikawa T, Kamoda H, Suzuki M, Inoue G, Sakuma Y, Oikawa Y, Orita S, Uchida K, Takahashi K, Takaso M, Ohtori S. Efficacy of nerve growth factor antibody in a knee osteoarthritis pain model in mice. BMC Musculoskelet Disord 2017; 18:428. [PMID: 29100502 PMCID: PMC5670727 DOI: 10.1186/s12891-017-1792-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 10/30/2017] [Indexed: 01/06/2023] Open
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.
Collapse
Affiliation(s)
- Masayuki Miyagi
- Department of Orthopaedic Surgery, Kitasato University, School of Medicine, 1-15-1, Kitasato, Minami-ku, Sagamihara city, Kanagawa, 252-0374, Japan.
| | - Tetsuhiro Ishikawa
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroto Kamoda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Miyako Suzuki
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Gen Inoue
- Department of Orthopaedic Surgery, Kitasato University, School of Medicine, 1-15-1, Kitasato, Minami-ku, Sagamihara city, Kanagawa, 252-0374, Japan
| | - Yoshihiro Sakuma
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yasuhiro Oikawa
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Sumihisa Orita
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kentaro Uchida
- Department of Orthopaedic Surgery, Kitasato University, School of Medicine, 1-15-1, Kitasato, Minami-ku, Sagamihara city, Kanagawa, 252-0374, Japan
| | - Kazuhisa Takahashi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masashi Takaso
- Department of Orthopaedic Surgery, Kitasato University, School of Medicine, 1-15-1, Kitasato, Minami-ku, Sagamihara city, Kanagawa, 252-0374, Japan
| | - Seiji Ohtori
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
35
|
Miller RE, Malfait AM. Osteoarthritis pain: What are we learning from animal models? Best Pract Res Clin Rheumatol 2017; 31:676-687. [PMID: 30509413 PMCID: PMC6284232 DOI: 10.1016/j.berh.2018.03.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 02/27/2018] [Accepted: 03/08/2018] [Indexed: 12/15/2022]
Abstract
All experimental models of osteoarthritis (OA)-like joint damage are accompanied by behaviors indicative of pain. In experimental knee OA, evoked pain responses to exogenously applied stimuli suggest that animals become sensitized to mechanical stimuli. Neurobiological techniques including electrophysiology and in vivo calcium imaging confirm that joint damage is associated with mechanical stimuli through peripheral sensitization. Several mediators present in the OA joint can cause peripheral sensitization, most notably the neurotrophin nerve growth factor (NGF). Furthermore, experimental OA is associated with neuroinflammation in the peripheral nervous system and central nervous system (CNS), including macrophage infiltration of the dorsal root ganglia and microglial activation in the spinal cord. Increasingly, researchers are employing models that are slowly progressive, and this approach has revealed that distinct pain mechanisms operate in a time-dependent manner, which may have important translational significance. While the study of pain in experimental OA is rapidly evolving, with the application of increasingly sophisticated techniques to assess pain and unravel the neurobiology of its genesis, important gaps and limitations in our current approaches exist, which our research community needs to address.
Collapse
Affiliation(s)
- Rachel E Miller
- Department of Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, United States
| | - Anne-Marie Malfait
- Department of Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W Harrison Street, Suite 510, Chicago, IL, 60612, United States.
| |
Collapse
|
36
|
Nerve growth factor blockade for the management of osteoarthritis pain: what can we learn from clinical trials and preclinical models? Curr Opin Rheumatol 2017; 29:110-118. [PMID: 27672741 DOI: 10.1097/bor.0000000000000354] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW Anti-nerve growth factor (NGF) antibodies hold tremendous potential for the management of osteoarthritis pain, but clinical trials have revealed serious adverse effects that are incompletely understood. This review discusses clinical trial results along with preclinical studies that have assessed NGF blockade in experimental osteoarthritis, in order to provide insight for future studies. RECENT FINDINGS Systematic reviews have revealed that anti-NGF therapy, including tanezumab, is efficacious in improving pain and function, but serious adverse events, including rapidly progressive osteoarthritis and osteonecrosis, resulted in a moratorium on trials that was only recently lifted. Within the past year, preclinical testing has revealed effects of NGF blockade on both pain behaviors and joint structure in experimental models of osteoarthritis. Similar to clinical trial results, these studies in laboratory animals demonstrated analgesic efficacy of NGF blockade. Interestingly, several animal studies have suggested detrimental effects on joint integrity as a result of treatment, particularly when treatment is started early in the disease, when joint damage is mild to moderate. SUMMARY NGF blockade continues to represent a promising new approach for the treatment of osteoarthritis pain, but the actual benefits and risks remain to be fully elucidated. Preclinical models may suggest patient populations that could be best served while limiting side-effects, but future work should further investigate the mechanisms of benefits and unwanted side-effects.
Collapse
|
37
|
MicroRNA-29a Counteracts Synovitis in Knee Osteoarthritis Pathogenesis by Targeting VEGF. Sci Rep 2017; 7:3584. [PMID: 28620193 PMCID: PMC5472675 DOI: 10.1038/s41598-017-03616-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 05/02/2017] [Indexed: 12/11/2022] Open
Abstract
Synovitis contributes to the development of osteoarthritis (OA) of the knee. MicroRNAs regulate joint microenvironment homeostasis and deterioration. This study was undertaken to characterize the actions of microRNA-29a (miR-29a) to synovial remodeling in OA joints. Synovial specimens isolated from patients with end-stage OA knees showed abundant fibrotic matrix and vessel histopathology concomitant with weak miR-29a expression. In vitro, miR-29a knockdown caused synovial fibroblasts to exhibit high expressions of collagen III, TGF-β1, MMP9, MMP13, and ADAMTS5, whereas miR-29a overexpression diminished these joint-deleterious factors. In collagenase-mediated OA pathogenesis, miR-29a-overexpressing transgenic mice showed minor responses to hyperplasia, macrophage infiltration, fibrosis, hyperangiogenesis, and VEGF expression in synovial lesions. These effects mitigated articular cartilage loss and gait aberrance of injured joints. Intra-articular administration of miR-29a precursor lessened the collagenase aggravation of excessive synovial remodeling reactions and thereby sustained joint tissue integrity. miR-29a lowered VEGF production and angiogenic activities in synovial fibroblasts through targeting the 3′-UTR of VEGF. Taken together, miR-29a deficiency exacerbated synovitis pathogenesis in the end-stage OA knees. miR-29a signaling fends off excessive synovial angiogenesis and fibrosis, which delays joint destruction. This study sheds new light on the protective effects against synovial deterioration and the therapeutic advantage of miR-29a in OA knees.
Collapse
|
38
|
Biologic drugs as analgesics for the management of osteoarthritis. Semin Arthritis Rheum 2017; 46:687-691. [DOI: 10.1016/j.semarthrit.2016.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 11/23/2016] [Accepted: 12/01/2016] [Indexed: 01/05/2023]
|
39
|
Blaney Davidson EN, van Caam APM, van der Kraan PM. Osteoarthritis year in review 2016: biology. Osteoarthritis Cartilage 2017; 25:175-180. [PMID: 28100421 DOI: 10.1016/j.joca.2016.09.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/31/2016] [Accepted: 09/01/2016] [Indexed: 02/02/2023]
Abstract
This review highlights a selection of literature in the area of osteoarthritis biology published between the 2015 and 2016 Osteoarthritis Research Society International (OARSI) World Congress. Highlights were selected from a pubmed search covering cartilage, bone, inflammation and pain. A personal selection was made based, amongst other things, on topics presented during the 2015 conference. This covers circadian rhythm, TGF-β signaling, autophagy, SIRT6, exercise, lubricin, TLR's, pain and NGF. Furthermore, in this review we have made an effort to connect these seemingly distant topics into one scheme of connections between them, revealing a theoretical big picture underneath.
Collapse
|
40
|
Abstract
Worldwide, osteoarthritis (OA) is one of the leading causes of chronic pain, for which adequate relief is not available. Ongoing peripheral input from the affected joint is a major factor in OA-associated pain. Therefore, this review focuses predominantly on peripheral targets emerging in the preclinical and clinical arena. Nerve growth factor is the most advanced of these targets, and its blockade has shown tremendous promise in clinical trials in knee OA. A number of different types of ion channels, including voltage-gated sodium channels and calcium channels, transient receptor potential channels, and acid-sensing ion channels, are important for neuronal excitability and play a role in pain genesis. Few channel blockers have been tested in preclinical models of OA, with varying results. Finally, we discuss some examples of G-protein coupled receptors, which may offer attractive therapeutic strategies for OA pain, including receptors for bradykinin, calcitonin gene-related peptide, and chemokines. Since many of the pathways described above can be selectively and potently targeted, they offer an exciting opportunity for pain management in OA, either systemically or locally.
Collapse
Affiliation(s)
- Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W Harrison Street, Suite 510, Chicago, IL, 60612, USA.
| | - Richard J Miller
- Department of Pharmacology, Northwestern University, Robert H. Lurie Medical Research Center, 303 E. Superior, Chicago, IL, 60613, USA
| |
Collapse
|
41
|
Abstract
Osteoarthritis (OA) is a multifactorial chronic joint disease, and so far, there are no approved disease-modifying anti-OA drugs (DMOADs). There is an urgent need to develop therapies for different phenotypes of OA. Monoclonal antibodies (mAb) may slow structural progression, control inflammation and relieve pain, and thus have the potential to be DMOADs. Areas covered: In this review, the authors searched the literature on PubMed, EMBASE and the Cochrane Library using keywords, including mAbs, biological agents, OA and osteoarthritis, electronically up to May 2016. They also included abstracts of international conferences. Furthermore, they reviewed experimental and clinical studies of various mAbs targeting different pathological mechanisms of OA, including ADAMTS, Interleukine-1, tumour necrosis factor, never growth factor and vascular endothelial growth factor. Expert opinion: MAbs for the treatment of OA are under intense investigation and the results for some mAbs (e.g., anti-nerve growth factor mAbs, anti- vascular endothelial growth factor mAbs) are promising. The authors believe that mAb therapy can be a targeted therapeutic approach for the treatment of OA. Future clinical trials are required to evaluate the therapeutic efficacy of these agents by the appropriate selection of specific phenotype for targeted therapy based on the mechanism of drug action.
Collapse
Affiliation(s)
- Shuang Zheng
- a Arthritis Research Institute & Department of Rheumatology , 1st Affiliated Hospital of Anhui Medical University , Hefei , China
- b Menzies Institute for Medical Research , University of Tasmania , Hobart , Australia
| | - David J Hunter
- c Institute of Bone and Joint Research, Kolling Institute and Royal North Shore Hospital , University of Sydney , Sydney , Australia
| | - Jianhua Xu
- a Arthritis Research Institute & Department of Rheumatology , 1st Affiliated Hospital of Anhui Medical University , Hefei , China
| | - Changhai Ding
- a Arthritis Research Institute & Department of Rheumatology , 1st Affiliated Hospital of Anhui Medical University , Hefei , China
- b Menzies Institute for Medical Research , University of Tasmania , Hobart , Australia
| |
Collapse
|
42
|
Adams BL, Guo W, Gors RT, Knopp KL. Pharmacological interrogation of a rodent forced ambulation model: leveraging gait impairment as a measure of pain behavior pre-clinically. Osteoarthritis Cartilage 2016; 24:1928-1939. [PMID: 27450884 DOI: 10.1016/j.joca.2016.05.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 04/15/2016] [Accepted: 05/11/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The aim of this study was to investigate whether inflammogen-induced temporal and spatial gait changes in a rodent forced-ambulation paradigm were sensitive to pharmacological intervention with both clinically validated and novel analgesics. METHODS Using the GaitScan (CleverSys Inc., Reston, VA) treadmill system, we identified four functional endpoints inspired by clinical literature and sensitive to unilateral joint injury induced by intra-articular Complete Freund's Adjuvant (CFA). These endpoints included: range of motion, normalized stance distance, stance/swing ratio, and paw print size as a measure of guarding; collectively, these measures are proposed to serve as a high fidelity index of joint pain. We then examined the ability of known analgesic mechanisms to attenuate gait impairment as measured by this index. RESULTS Clinically efficacious opioids, Nonsteroidal anti-inflammatory drugs (NSAIDs), and the yet unapproved anti-NGF antibody dose-dependently attenuated the CFA)-induced gait deficits, while a TNF-alpha fusion protein blocker had no effect on gait, but did produce a reduction in swelling. As well, the time course for gait impairment in the model appears to be distinct from the traditional endpoint of tactile hypersensitivity, offering the potential to assess a novel functional pain phenotype. CONCLUSIONS In response to the call for more functional pain measures, we submit this composite gait score as a novel endpoint to interrogate joint pain pre-clinically. As the etiology of human osteoarthritis (OA) remains unclear, this model/endpoint cannot attempt to improve construct validity, but may provide an additional dimension to interrogate pain-induced gait deficits.
Collapse
Affiliation(s)
- B L Adams
- Neuroscience Research, Lilly Research Laboratories, Indianapolis, IN 46285, USA
| | - W Guo
- Neuroscience Research, Lilly Research Laboratories, Indianapolis, IN 46285, USA
| | - R T Gors
- Neuroscience Research, Lilly Research Laboratories, Indianapolis, IN 46285, USA
| | - K L Knopp
- Neuroscience Research, Lilly Research Laboratories, Indianapolis, IN 46285, USA.
| |
Collapse
|
43
|
El-Tawil S, Arendt E, Parker D. Position statement: the epidemiology, pathogenesis and risk factors of osteoarthritis of the knee. J ISAKOS 2016. [DOI: 10.1136/jisakos-2015-000002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
44
|
Malfait AM. Osteoarthritis year in review 2015: biology. Osteoarthritis Cartilage 2016; 24:21-6. [PMID: 26707989 PMCID: PMC4693144 DOI: 10.1016/j.joca.2015.09.010] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/04/2015] [Accepted: 09/14/2015] [Indexed: 02/07/2023]
Abstract
This review highlights a selection of recently published literature in the area of osteoarthritis biology. Major themes transpiring from a PubMed search covering the year between the 2014 and the 2015 Osteoarthritis Research Society International (OARSI) World Congress are explored. Inflammation emerged as a significant theme, revealing complex pathways that drive dramatic changes in cartilage homeostasis and in the synovium. Highlights include a homeostatic role for CXC chemokines in cartilage, identification of the zinc-ZIP8-MTF1 axis as an essential regulator of cartilage catabolism, and the discovery that a small aggrecan fragment can have catabolic and pro-inflammatory effects through Toll-like receptor 2. Synovitis can promote joint damage, partly through alarmins such as S100A8. Synovitis and synovial expression of the pro-algesic neurotrophin, Nerve Growth Factor, are associated with pain. Increasingly, researchers are considering specific pathogenic pathways that may operate in distinct subsets of osteoarthritis associated with distinct risk factors, including obesity, age, and joint injury. In obesity, the contribution of metabolic factors and diet is under intense investigation. The role of autophagy and oxidative stress in age-related osteoarthritis has been further explored. This approach may open avenues for targeted treatment of distinct phenotypes of osteoarthritis. Finally, a small selection of novel analgesic targets in the periphery is briefly discussed, including calcitonin gene-related peptide and the neuronal sodium voltage-gated channels, Nav1.7 and Nav1.8.
Collapse
Affiliation(s)
- A M Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA; Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
45
|
Miller RE, Tran PB, Sondoqah A, Raghu P, Ishihara S, Miller RJ, Malfait AM. The Role of Peripheral Nociceptive Neurons in the Pathophysiology of Osteoarthritis Pain. Curr Osteoporos Rep 2015; 13:318-26. [PMID: 26233284 PMCID: PMC4596062 DOI: 10.1007/s11914-015-0280-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Knee osteoarthritis is characterized by progressive damage and remodeling of all tissues in the knee joint. Pain is the main symptom associated with knee osteoarthritis. Recent clinical and pre-clinical studies have provided novel insights into the mechanisms that drive the pain associated with joint destruction. In this narrative review, we describe current knowledge regarding the changes in the peripheral and central nervous systems that occur during the progression of osteoarthritis and discuss how therapeutic interventions may provide pain relief.
Collapse
Affiliation(s)
- Rachel E. Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
- Department of Biochemistry, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
| | - Phuong B. Tran
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
| | - Alia Sondoqah
- Department of Biochemistry, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
| | - Padmanabhan Raghu
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
| | - Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
| | - Richard J. Miller
- Department of Pharmacology, Northwestern University, Lurie 8-125, 303 E. Superior St, Chicago, IL 60611
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
- Department of Biochemistry, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
- Corresponding author Anne-Marie Malfait, MD, PhD, Associate Professor of Medicine, , T: 312-563-2925, F: 312-563-2267
| |
Collapse
|