1
|
Wang D, Liu W, Venkatesan JK, Madry H, Cucchiarini M. Therapeutic Controlled Release Strategies for Human Osteoarthritis. Adv Healthc Mater 2024:e2402737. [PMID: 39506433 DOI: 10.1002/adhm.202402737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis is a progressive, irreversible debilitating whole joint disease that affects millions of people worldwide. Despite the availability of various options (non-pharmacological and pharmacological treatments and therapy, orthobiologics, and surgical interventions), none of them can definitively cure osteoarthritis in patients. Strategies based on the controlled release of therapeutic compounds via biocompatible materials may provide powerful tools to enhance the spatiotemporal delivery, expression, and activities of the candidate agents as a means to durably manage the pathological progression of osteoarthritis in the affected joints upon convenient intra-articular (injectable) delivery while reducing their clearance, dissemination, or side effects. The goal of this review is to describe the current knowledge and advancements of controlled release to treat osteoarthritis, from basic principles to applications in vivo using therapeutic recombinant molecules and drugs and more innovatively gene sequences, providing a degree of confidence to manage the disease in patients in a close future.
Collapse
Affiliation(s)
- Dan Wang
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| |
Collapse
|
2
|
Bihlet AR, Byrjalsen I, Mundbjerg K, Rovsing H, Axelsen TM, Andersen JR, Metnik A, Bachtell N, Brett A, Alexandersen P. A phase 2b double-blind placebo-controlled randomized clinical trial of SB-061, an aggrecan mimetic, in patients with symptomatic knee osteoarthritis. Osteoarthritis Cartilage 2024; 32:1471-1480. [PMID: 38960141 DOI: 10.1016/j.joca.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/04/2024] [Accepted: 06/11/2024] [Indexed: 07/05/2024]
Abstract
OBJECTIVES To evaluate the efficacy and safety of intra-articular injections of a novel aggrecan mimetic, SB-061, in subjects with knee osteoarthritis (OA). METHODS This was a randomized, placebo-controlled, double-blind phase II study comparing intra-articular injections of SB-061 with placebo (isotonic saline) for 52 weeks, administered at baseline, Wk 16, and Wk 32. Eligible subjects had a KL grade of 2 or 3 on X-ray of the target knee and a Western Ontario McMaster Universities Osteoarthritis Index (WOMAC) pain score ≥20 out of 50 at screening and baseline visits. Subjects having any other knee condition were excluded. Use of analgesics was prohibited, except for rescue medication. The primary endpoint was change from baseline (CFB) in WOMAC pain at Week 8. Secondary endpoints were CFB in WOMAC function and total, ICOAP, Patient Global Assessment, and 20-meter walk test. Exploratory endpoints included structural CFB in magnetic resonance imaging entities. RESULTS A total of 288 subjects were randomized to SB-061 (n = 145) or placebo (n = 143), and 252 (87.5%) completed injections. The groups were comparable at baseline. The primary endpoint was not met, as no significant difference in the CFB of the WOMAC pain score at Week 8 between groups was observed, nor at any other time point during the study. Similarly, neither of the secondary or exploratory endpoints indicated any significant difference between groups. The frequency and type of adverse events were similar between groups. SB-061 was well-tolerated. CONCLUSION Intra-articular injections of SB-061 administered at baseline, Week 16, and Week 32, over one year in subjects with knee OA, were safe but did not show any statistically significant effect on knee pain nor on other symptomatic or structural entities compared to placebo. TRIAL REGISTRATION NUMBER EUDRACT NO 2019-004515-31.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anna Metnik
- NBCD A/S, Telefonvej 8D, 2860 Søborg, Denmark.
| | - Nathan Bachtell
- Third Harmonic Bio, 1700 Montgomery Street, Suite #210, San Francisco, CA 94111, USA.
| | - Alan Brett
- Imorphics Ltd, Worthington House, Towers Business Park, Wilmslow Road, Manchester M20 2HJ, UK.
| | | |
Collapse
|
3
|
Liang W, Feng R, Li X, Duan X, Feng S, Chen J, Li Y, Chen J, Liu Z, Wang X, Ruan G, Tang S, Ding C, Huang B, Zou Z, Chen T. A RANKL-UCHL1-sCD13 negative feedback loop limits osteoclastogenesis in subchondral bone to prevent osteoarthritis progression. Nat Commun 2024; 15:8792. [PMID: 39389988 PMCID: PMC11466963 DOI: 10.1038/s41467-024-53119-2] [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: 10/08/2023] [Accepted: 09/27/2024] [Indexed: 10/12/2024] Open
Abstract
Abnormal subchondral bone remodeling plays a pivotal role in the progression of osteoarthritis (OA). Here, we analyzed subchondral bone samples from OA patients and observed a significant upregulation of ubiquitin carboxy-terminal hydrolase L1 (UCHL1) specifically in subchondral bone osteoclasts. Notably, we found a strong correlation between UCHL1 expression and osteoclast activity in the subchondral bone during OA progression in both human and murine models. Conditional UCHL1 deletion in osteoclast precursors exacerbated OA progression, while its overexpression, mediated by adeno-associated virus 9, alleviated this process in male mice. Mechanistically, RANKL stimulates UCHL1 expression in osteoclast precursors, subsequently stabilizing CD13, augmenting soluble CD13 (sCD13) release, and triggering an autocrine inhibitory effect on the MAPK pathway, thereby suppressing osteoclast formation. These findings unveil a previously unidentified negative feedback loop, RANKL-UCHL1-sCD13, that modulates osteoclast formation and presents a potential therapeutic target for OA.
Collapse
Affiliation(s)
- Wenquan Liang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
- Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Ru Feng
- Department of Rehabilitation medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Xiaojia Li
- Department of Rehabilitation medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xingwei Duan
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shourui Feng
- State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jun Chen
- Department of Rehabilitation Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yicheng Li
- Department of Rehabilitation medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Junqi Chen
- Department of Rehabilitation medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zezheng Liu
- Department of Orthopaedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiaogang Wang
- Department of Orthopaedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Guangfeng Ruan
- Clinical Research Centre, Guangzhou First People's Hospital, Guangzhou, China
| | - Su'an Tang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Bin Huang
- Department of Orthopaedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| | - Zhipeng Zou
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Tianyu Chen
- Department of Orthopaedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
4
|
Adu Y, Ring D, Teunis T. Randomized Controlled Trials Studying Nonoperative Treatments of Osteoarthritis Often Use Misleading and Uninformative Control Groups: A Systematic Review. Clin Orthop Relat Res 2024:00003086-990000000-01759. [PMID: 39453403 DOI: 10.1097/corr.0000000000003273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Because there are no known treatments that alter the natural course of the pathophysiology of osteoarthritis, nonoperative treatment needs to be compared with known effective treatments that seek to mitigate symptoms or with similarly invasive inert (placebo) treatments to determine effectiveness. Comparing a treatment to an uninformative control group may inappropriately legitimize and support the use of potentially ineffective treatments. We therefore investigated the prevalence of inappropriate control groups in musculoskeletal research and asked whether these are associated with reporting a positive treatment effect. QUESTIONS/PURPOSES We systematically reviewed randomized trials of nonoperative treatments of osteoarthritis and asked: (1) What proportion of randomized trials use uninformative control groups (defined as a treatment less invasive than the tested treatment, or a treatment that might possibly not outperform placebo but is not acknowledged as such)? (2) Is the use of uninformative control groups independently associated with reporting a positive treatment effect (defined as p < 0.05 in favor of the intervention, or as making a recommendation favoring the intervention over the control treatment)? METHODS In a systematic review following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we searched PubMed, Cochrane, and Embase up to September 2023 for randomized controlled trials published between 2020 to 2022 that compared one or more nonoperative treatments for the symptoms of osteoarthritis. We excluded studies that contained a surgical treatment group. We identified 103 trials that met eligibility criteria, with a total of 15,491 patients. The risk of bias was high in 60% (n = 62) of trials using the Cochrane Risk of Bias Tool, version 2. Although the high risk of bias in the included studies is concerning, it does not invalidate our design; instead, it highlights that some studies may use flawed methods to recommend treatments with unproven effectiveness beyond nonspecific effects because the kinds of bias observed would tend to increase the apparent benefit of the treatment(s) being evaluated. We used logistic regression to test the association of uninformative control groups with a positive treatment effect, accounting for potential confounders such as conflict of interest and study bias using the Cochrane Risk of Bias score. RESULTS The use of uninformative control groups (treatments less invasive than the tested treatment, or treatments that might not outperform placebo but are not acknowledged as such) was found in 46% (47 of 103) of included studies. After accounting for potential confounding, there was no association between reporting positive treatment effects and the use of an uninformative control group. Studies with a low risk of bias had a lower likelihood of reporting a positive treatment effect (OR 0.2 [95% confidence interval 0.05 to 0.9]; p = 0.04, model pseudo R2 = 0.21). CONCLUSION The finding that recent studies that mimic high-level evidence often use uninformative control groups that do not adequately account for nonspecific effects (perceived treatment benefits unrelated to a treatment's direct physiological effects) points to a high risk of legitimizing ineffective treatments. This raises the ethical imperative for patients, clinicians, journal peer reviewers, and journal editors to hold researchers to the standard of an adequate, informative control group. Awareness and risk of bias checklists might help patients and clinicians forgo new treatments based on seemingly high-level evidence that may carry only iatrogenic, financial, and psychological harm (false hope, in particular). LEVEL OF EVIDENCE Level I, therapeutic study.
Collapse
Affiliation(s)
- Yaw Adu
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - David Ring
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Teun Teunis
- Department of Plastic Surgery, University Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Ilyas S, Lee J, Hwang Y, Choi Y, Lee D. Deciphering Cathepsin K inhibitors: a combined QSAR, docking and MD simulation based machine learning approaches for drug design. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:771-793. [PMID: 39382544 DOI: 10.1080/1062936x.2024.2405626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024]
Abstract
Cathepsin K (CatK), a lysosomal cysteine protease, contributes to skeletal abnormalities, heart diseases, lung inflammation, and central nervous system and immune disorders. Currently, CatK inhibitors are associated with severe adverse effects, therefore limiting their clinical utility. This study focuses on exploring quantitative structure-activity relationships (QSAR) on a dataset of CatK inhibitors (1804) compiled from the ChEMBL database to predict the inhibitory activities. After data cleaning and pre-processing, a total of 1568 structures were selected for exploratory data analysis which revealed physicochemical properties, distributions and statistical significance between the two groups of inhibitors. PubChem fingerprinting with 11 different machine-learning classification models was computed. The comparative analysis showed the ET model performed well with accuracy values for the training set (0.999), cross-validation (0.970) and test set (0.977) in line with OECD guidelines. Moreover, to gain structural insights on the origin of CatK inhibition, 15 diverse molecules were selected for molecular docking. The CatK inhibitors (1 and 2) exhibited strong binding energies of -8.3 and -7.2 kcal/mol, respectively. MD simulation (300 ns) showed strong structural stability, flexibility and interactions in selected complexes. This synergy between QSAR, docking, MD simulation and machine learning models strengthen our evidence for developing novel and resilient CatK inhibitors.
Collapse
Affiliation(s)
- S Ilyas
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, Seongnam-si, Korea
| | - J Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, Seongnam-si, Korea
| | - Y Hwang
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, Seongnam-si, Korea
| | - Y Choi
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, Seongnam-si, Korea
| | - D Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, Seongnam-si, Korea
| |
Collapse
|
6
|
Kingsbury SR, Tharmanathan P, Keding A, Watt FE, Scott DL, Roddy E, Birrell F, Arden NK, Bowes M, Arundel C, Watson M, Ronaldson SJ, Hewitt C, Doherty M, Moots RJ, O'Neill TW, Green M, Patel G, Garrood T, Edwards CJ, Walmsley PJ, Sheeran T, Torgerson DJ, Conaghan PG. Pain Reduction With Oral Methotrexate in Knee Osteoarthritis : A Randomized, Placebo-Controlled Clinical Trial. Ann Intern Med 2024; 177:1145-1156. [PMID: 39074374 DOI: 10.7326/m24-0303] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Treatments for osteoarthritis (OA) are limited. Previous small studies suggest that the antirheumatic drug methotrexate may be a potential treatment for OA pain. OBJECTIVE To assess symptomatic benefits of methotrexate in knee OA (KOA). DESIGN A multicenter, randomized, double-blind, placebo-controlled trial done between 13 June 2014 and 13 October 2017. (ISRCTN77854383; EudraCT: 2013-001689-41). SETTING 15 secondary care musculoskeletal clinics in the United Kingdom. PARTICIPANTS A total of 207 participants with symptomatic, radiographic KOA and knee pain (severity ≥4 out of 10) on most days in the past 3 months with inadequate response to current medication were approached for inclusion. INTERVENTION Participants were randomly assigned 1:1 to oral methotrexate once weekly (6-week escalation 10 to 25 mg) or matched placebo over 12 months and continued usual analgesia. MEASUREMENTS The primary end point was average knee pain (numerical rating scale [NRS] 0 to 10) at 6 months, with 12-month follow-up to assess longer-term response. Secondary end points included knee stiffness and function outcomes and adverse events (AEs). RESULTS A total of 155 participants (64% women; mean age, 60.9 years; 50% Kellgren-Lawrence grade 3 to 4) were randomly assigned to methotrexate (n = 77) or placebo (n = 78). Follow-up was 86% (n = 134; methotrexate: 66, placebo: 68) at 6 months. Mean knee pain decreased from 6.4 (SD, 1.80) at baseline to 5.1 (SD, 2.32) at 6 months in the methotrexate group and from 6.8 (SD, 1.62) to 6.2 (SD, 2.30) in the placebo group. The primary intention-to-treat analysis showed a statistically significant pain reduction of 0.79 NRS points in favor of methotrexate (95% CI, 0.08 to 1.51; P = 0.030). There were also statistically significant treatment group differences in favor of methotrexate at 6 months for Western Ontario and McMaster Universities Osteoarthritis Index stiffness (0.60 points [CI, 0.01 to 1.18]; P = 0.045) and function (5.01 points [CI, 1.29 to 8.74]; P = 0.008). Treatment adherence analysis supported a dose-response effect. Four unrelated serious AEs were reported (methotrexate: 2, placebo: 2). LIMITATION Not permitting oral methotrexate to be changed to subcutaneous delivery for intolerance. CONCLUSION Oral methotrexate added to usual medications demonstrated statistically significant reduction in KOA pain, stiffness, and function at 6 months. PRIMARY FUNDING SOURCE Versus Arthritis.
Collapse
Affiliation(s)
- Sarah R Kingsbury
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, and National Institute for Health and Care Research (NIHR) Leeds Biomedical Research Centre, Leeds, United Kingdom (S.R.K., P.G.C.)
| | - Puvan Tharmanathan
- York Trials Unit, Department of Health Sciences, Faculty of Science, University of York, Heslington, York, United Kingdom (P.T., A.K., C.A., M.W., S.J.R., C.H., D.J.T.)
| | - Ada Keding
- York Trials Unit, Department of Health Sciences, Faculty of Science, University of York, Heslington, York, United Kingdom (P.T., A.K., C.A., M.W., S.J.R., C.H., D.J.T.)
| | - Fiona E Watt
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Hammersmith Campus, Imperial College London, and Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, and Department of Rheumatology, Oxford University Hospitals NHS Foundation Trust, and Centre for Sport, Exercise and Osteoarthritis Research Versus Arthritis, Oxford, United Kingdom (F.E.W.)
| | - David L Scott
- King's College London, London, United Kingdom (D.L.S.)
| | - Edward Roddy
- Primary Care Centre Versus Arthritis, Keele University, and Haywood Academic Rheumatology Centre, Midlands Partnership University NHS Foundation Trust, Keele, United Kingdom (E.R.)
| | - Fraser Birrell
- Medical Research Council-Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing, Newcastle University, and Northumbria Healthcare NHS Foundation Trust, Newcastle upon Tyne, United Kingdom (F.B.)
| | - Nigel K Arden
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, and Centre for Sport, Exercise and Osteoarthritis Research Versus Arthritis, University of Oxford, Oxford, United Kingdom (N.K.A.)
| | - Mike Bowes
- Imorphics, Manchester, United Kingdom (M.B.)
| | - Catherine Arundel
- York Trials Unit, Department of Health Sciences, Faculty of Science, University of York, Heslington, York, United Kingdom (P.T., A.K., C.A., M.W., S.J.R., C.H., D.J.T.)
| | - Michelle Watson
- York Trials Unit, Department of Health Sciences, Faculty of Science, University of York, Heslington, York, United Kingdom (P.T., A.K., C.A., M.W., S.J.R., C.H., D.J.T.)
| | - Sarah J Ronaldson
- York Trials Unit, Department of Health Sciences, Faculty of Science, University of York, Heslington, York, United Kingdom (P.T., A.K., C.A., M.W., S.J.R., C.H., D.J.T.)
| | - Catherine Hewitt
- York Trials Unit, Department of Health Sciences, Faculty of Science, University of York, Heslington, York, United Kingdom (P.T., A.K., C.A., M.W., S.J.R., C.H., D.J.T.)
| | - Michael Doherty
- Academic Rheumatology and Pain Centre Versus Arthritis, University of Nottingham, Nottingham, United Kingdom (M.D.)
| | - Robert J Moots
- Faculty of Heath Social Care and Medicine, Edge Hill University, Ormskirk, and Department of Rheumatology, Aintree University Hospital, Liverpool, United Kingdom (R.J.M.)
| | - Terence W O'Neill
- Centre for Epidemiology Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, Manchester Academic Health Science Centre, University of Manchester, and NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom (T.W.O.)
| | - Michael Green
- Harrogate and District NHS Foundation Trust, Harrogate, and York Teaching Hospital NHS Foundation Trust, York, United Kingdom (M.G.)
| | - Gulam Patel
- Rheumatology Department, Ashford and St. Peter's Hospital NHS Trust, Chertsey, United Kingdom (G.P.)
| | - Toby Garrood
- Department of Rheumatology, Guy's and St. Thomas' NHS Trust, London, United Kingdom (T.G.)
| | - Christopher J Edwards
- NIHR Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom (C.J.E.)
| | - Phil J Walmsley
- Department of Orthopaedics, Victoria Hospital Kirkcaldy and School of Medicine, St. Andrews University, Fife and Fife NHS Trust, Kirkcaldy, United Kingdom (P.J.W.)
| | - Tom Sheeran
- Department of Rheumatology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, United Kingdom (T.S.)
| | - David J Torgerson
- York Trials Unit, Department of Health Sciences, Faculty of Science, University of York, Heslington, York, United Kingdom (P.T., A.K., C.A., M.W., S.J.R., C.H., D.J.T.)
| | - Philip G Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, and National Institute for Health and Care Research (NIHR) Leeds Biomedical Research Centre, Leeds, United Kingdom (S.R.K., P.G.C.)
| |
Collapse
|
7
|
D’Agostino V, Sorriento A, Cafarelli A, Donati D, Papalexis N, Russo A, Lisignoli G, Ricotti L, Spinnato P. Ultrasound Imaging in Knee Osteoarthritis: Current Role, Recent Advancements, and Future Perspectives. J Clin Med 2024; 13:4930. [PMID: 39201072 PMCID: PMC11355885 DOI: 10.3390/jcm13164930] [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: 06/26/2024] [Revised: 08/04/2024] [Accepted: 08/19/2024] [Indexed: 09/02/2024] Open
Abstract
While conventional radiography and MRI have a well-established role in the assessment of patients with knee osteoarthritis, ultrasound is considered a complementary and additional tool. Moreover, the actual usefulness of ultrasound is still a matter of debate in knee osteoarthritis assessment. Despite that, ultrasound offers several advantages and interesting aspects for both current clinical practice and future perspectives. Ultrasound is potentially a helpful tool in the detection of anomalies such as cartilage degradation, osteophytes, and synovitis in cases of knee osteoarthritis. Furthermore, local diagnostic and minimally invasive therapeutic operations pertaining to knee osteoarthritis can be safely guided by real-time ultrasound imaging. We are constantly observing a growing knowledge and awareness among radiologists and other physicians, concerning ultrasound imaging. Ultrasound studies can be extremely useful to track the response to various therapies. For this specific aim, tele-ultrasonography may constitute an easy tool aiding precise and repeated follow-up controls. Moreover, raw radio-frequency data from US backscattering signals contain more information than B-mode imaging. This paves the way for quantitative in-depth analyses of cartilage, bone, and other articular structures. Overall, ultrasound technologies and their rapid evolution have the potential to make a difference at both the research and clinical levels. This narrative review article describes the potential of such technologies and their possible future implications.
Collapse
Affiliation(s)
- Valerio D’Agostino
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Via GC Pupilli 1, 40136 Bologna, Italy
- Radiology Unit, Policlinico Ospedaliero “Umberto I”, Nocera Inferiore, 84014 Salerno, Italy
| | - Angela Sorriento
- The BioRobotics Institute, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Department of Excellence in Robotics & AI, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Andrea Cafarelli
- The BioRobotics Institute, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Department of Excellence in Robotics & AI, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Danilo Donati
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Nicolas Papalexis
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Via GC Pupilli 1, 40136 Bologna, Italy
| | - Alessandro Russo
- Clinica 2, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Gina Lisignoli
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Leonardo Ricotti
- The BioRobotics Institute, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Department of Excellence in Robotics & AI, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Paolo Spinnato
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Via GC Pupilli 1, 40136 Bologna, Italy
| |
Collapse
|
8
|
Pan L, Nagib L, Ganguly S, Moorthy A, Tahir H. A comprehensive review of phase 2/3 trials in osteoarthritis: an expert opinion. Expert Opin Emerg Drugs 2024:1-13. [PMID: 39087391 DOI: 10.1080/14728214.2024.2386174] [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: 05/31/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024]
Abstract
INTRODUCTION Osteoarthritis (OA) is a chronic, degenerative, and debilitating disease associated with significant long-term morbidity and disability. The pathogenesis of OA is not completely understood but involves an interplay between environmental risk factors, joint mechanics, abnormal pain pathways and upregulation of inflammatory signaling pathways. Current therapeutic options for patients are limited to conservative management, minimal pharmacological options or surgical management, with significant caveats to all approaches. AREAS COVERED In this review, we have set out to investigate current phase II/III clinical trials by undertaking a PubMed search. Examined clinical trials have explored a myriad of potential therapeutics from conventional disease-modifying anti-rheumatic drugs and biologics usually used in the treatment of inflammatory arthritides, to more novel approaches targeting inflammatory pathways implicated in OA, cartilage degeneration or pain pathways. EXPERT OPINION Unfortunately, most completed phase II/III clinical trials have shown little impact on patient pain scores, with the exception of the traditional DMARD methotrexate and Sprifermin. Methotrexate has been shown to be beneficial when used in the correct patient cohort (MRI proven synovitis). Sprifermin has the longest follow-up data of 5 years and has been shown to reduce loss of MRI-measured cartilage thickness and pain scores.
Collapse
Affiliation(s)
- Liyang Pan
- General internal medicine, Imperial College Healthcare NHS Trust, London, UK
| | - Lydia Nagib
- General internal medicine, Royal Free London NHS Foundation Trust, London, UK
| | - Sujata Ganguly
- Department of rheumatology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Arumugam Moorthy
- Department of rheumatology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Hasan Tahir
- General internal medicine, Royal Free London NHS Foundation Trust, London, UK
- Department of rheumatology, University College London, London, UK
| |
Collapse
|
9
|
Oo WM. Prospects of Disease-Modifying Osteoarthritis Drugs. Rheum Dis Clin North Am 2024; 50:483-518. [PMID: 38942581 DOI: 10.1016/j.rdc.2024.03.003] [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] [Indexed: 06/30/2024]
Abstract
Osteoarthritis (OA) causes a massive disease burden with a global prevalence of nearly 23% in 2020 and an unmet need for adequate treatment, given a lack of disease-modifying drugs (DMOADs). The author reviews the prospects of active DMOAD candidates in the phase 2/3 clinical trials of drug development pipeline based on key OA pathogenetic mechanisms directed to inflammation-driven, bone-driven, and cartilage-driven endotypes. The challenges and possible research opportunities are stated in terms of the formulation of a research question known as the PICO approach: (1) population, (2) interventions, (3) comparison or placebo, and (4) outcomes.
Collapse
Affiliation(s)
- Win Min Oo
- Department of Physical Medicine and Rehabilitation, Mandalay General Hospital, University of Medicine, Mandalay, Mandalay, Myanmar; Rheumatology Department, Royal North Shore Hospital, Institute of Bone and Joint Research, Kolling Institute, The University of Sydney, Sydney, Australia.
| |
Collapse
|
10
|
Saxer F, Demanse D, Brett A, Laurent D, Mindeholm L, Conaghan P, Schieker M. Prognostic value of B-score for predicting joint replacement in the context of osteoarthritis phenotypes: Data from the osteoarthritis initiative. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100458. [PMID: 38495348 PMCID: PMC10944111 DOI: 10.1016/j.ocarto.2024.100458] [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] [Received: 07/12/2023] [Revised: 01/27/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024] Open
Abstract
Objective Developing new therapies for knee osteoarthritis (KOA) requires improved prediction of disease progression. This study evaluated the prognostic value of clinical clusters and machine-learning derived quantitative 3D bone shape B-score for predicting total and partial knee replacement (KR). Design This retrospective study used longitudinal data from the Osteoarthritis Initiative. A previous study used patients' clinical profiles to delineate phenotypic clusters. For these clusters, the distribution of B-scores was assessed (employing Tukey's method). The value of both cluster allocation and B-score for KR-prediction was then evaluated using multivariable Cox regression models and Kaplan-Meier curves for time-to-event analyses. The impact of using B-score vs. cluster was evaluated using a likelihood ratio test for the multivariable Cox model; global performances were assessed by concordance statistics (Harrell's C-index) and time dependent receiver operating characteristic (ROC) curves. Results B-score differed significantly for the individual clinical clusters (p < 0.001). Overall, 9.4% of participants had a KR over 9 years, with a shorter time to event in clusters with high B-score at baseline. Those clusters were characterized clinically by a high rate of comorbidities and potential signs of inflammation. Both phenotype and B-score independently predicted KR, with better prediction if combined (P < 0.001). B-score added predictive value in groups with less pain and radiographic severity but limited physical activity. Conclusions B-scores correlated with phenotypes based on clinical patient profiles. B-score and phenotype independently predicted KR surgery, with higher predictive value if combined. This can be used for patient stratification in drug development and potentially risk prediction in clinical practice.
Collapse
Affiliation(s)
- F. Saxer
- Novartis Biomedical Research, Novartis Campus, 4002, Basel, Switzerland
- Medical Faculty, University of Basel, 4002, Basel, Switzerland
| | - D. Demanse
- Novartis Pharma AG, 4002, Basel, Switzerland
| | - A. Brett
- Imorphics, Worthington House, Towers Business Park, Wilmslow Road, Manchester, M20 2HJ, UK
| | - D. Laurent
- Novartis Biomedical Research, Biomarker Development, 4002, Basel, Switzerland
| | - L. Mindeholm
- Novartis Biomedical Research, Novartis Campus, 4002, Basel, Switzerland
| | - P.G. Conaghan
- Leeds Institute of Rheumatic & Musculoskeletal Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, UK
| | - M. Schieker
- Novartis Biomedical Research, Novartis Campus, 4002, Basel, Switzerland
- Medical Faculty, Ludwig-Maximilians-University, Munich, 80336, Germany
| |
Collapse
|
11
|
Zhang M, Wang Z, Ding C. Pharmacotherapy for osteoarthritis-related pain: current and emerging therapies. Expert Opin Pharmacother 2024; 25:1209-1227. [PMID: 38938057 DOI: 10.1080/14656566.2024.2374464] [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: 04/06/2024] [Accepted: 06/26/2024] [Indexed: 06/29/2024]
Abstract
INTRODUCTION Osteoarthritis (OA) related pain has affected millions of people worldwide. However, the current pharmacological options for managing OA-related pain have not achieved a satisfactory effect. AREAS COVERED This narrative review provides an overview of the current and emerging drugs for OA-related pain. It covers the drugs' mechanism of action, safety, efficacy, and limitations. The National Library of Medicine (PubMed) database was primarily searched from 2000 to 2024. EXPERT OPINION Current treatment options are limited and suboptimal for OA pain management. Topical nonsteroidal anti-inflammatory drugs (NSAIDs) are the recognized and first-line treatment in the management of OA-related pain, and other drugs are inconsistent recommendations by guidelines. Emerging treatment options are promising for OA-related pain, including nerve growth factor (NGF) inhibitors, ion channel inhibitors, and calcitonin gene-related peptide (CGRP) antagonists. Besides, drugs repurposing from antidepressants and antiepileptic analgesics are shedding light on the management of OA-related pain. The management of OA-related pain is challenging as pain is heterogeneous and subjective. A more comprehensive strategy combined with non-pharmacological therapy needs to be considered, and tailored management options to individualized patients.
Collapse
Affiliation(s)
- Mengdi Zhang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiqiang Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
12
|
Eckstein F, Wluka AE, Wirth W, Cicuttini F. 30 Years of MRI-based cartilage & bone morphometry in knee osteoarthritis: From correlation to clinical trials. Osteoarthritis Cartilage 2024; 32:439-451. [PMID: 38331162 DOI: 10.1016/j.joca.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/20/2023] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
OBJECTIVE The first publication on morphometric analysis of articular cartilage using magnetic resonance imaging (MRI) in 1994 set the scene for a game change in osteoarthritis (OA) research. The current review highlights milestones in cartilage and bone morphometry, summarizing the rapid progress made in imaging, its application to understanding joint (patho-)physiology, and its use in interventional clinical trials. METHODS Based on a Pubmed search of articles from 1994 to 2023, the authors subjectively selected representative work illustrating important steps in the development or application of magnetic resonance-based cartilage and bone morphometry, with a focus on studies in humans, and on the knee. Research on OA-pathophysiology is addressed only briefly, given length constraints. Compositional and semi-quantitative assessment are not covered here. RESULTS The selected articles are presented in historical order as well as by content. We review progress in the technical aspects of image acquisition, segmentation and analysis, advances in understanding tissue growth, physiology, function, and adaptation, and a selection of clinical trials examining the efficacy of interventions on knee cartilage and bone. A perspective is provided of how lessons learned may be applied to future research and clinical management. CONCLUSIONS Over the past 30 years, MRI-based morphometry of cartilage and bone has contributed to a paradigm shift in understanding articular tissue physiology and OA pathophysiology, and to the development of new treatment strategies. It is likely that these technologies will continue to play a key role in the development and (accelerated) approval of therapy, potentially targeted to different OA phenotypes.
Collapse
Affiliation(s)
- Felix Eckstein
- Department of Imaging & Functional Musculoskeletal Research, Center of Anatomy and Cell Biology, Paracelsus Medical University (PMU), Salzburg, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University, Salzburg, Austria; Chondrometrics GmbH, Freilassing, Bavaria, Germany.
| | - Anita E Wluka
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Wolfgang Wirth
- Department of Imaging & Functional Musculoskeletal Research, Center of Anatomy and Cell Biology, Paracelsus Medical University (PMU), Salzburg, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University, Salzburg, Austria; Chondrometrics GmbH, Freilassing, Bavaria, Germany
| | - Flavia Cicuttini
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
13
|
Jiang T, Zhang J, Ruan B, Xi X, Yang Z, Liu J, Zhao H, Xu X, Jiang M. Trachelogenin alleviates osteoarthritis by inhibiting osteoclastogenesis and enhancing chondrocyte survival. Chin Med 2024; 19:37. [PMID: 38429848 PMCID: PMC10905921 DOI: 10.1186/s13020-024-00909-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/20/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent global health concern associated with the loss of articular cartilage and subchondral bone. The lack of disease-modifying drugs for OA necessitates the exploration of novel therapeutic options. Our previous study has demonstrated that traditional Chinese medical herb Trachelospermum jasminoides (Lindl.) Lem. extract suppressed osteoclastogenesis and identified trachelogenin (TCG) as a representative compound. Here, we delved into TCG's potential to alleviate OA. METHODS We initially validated the in vivo efficacy of TCG in alleviating OA using a rat OA model. Subsequently, we isolated primary bone marrow-derived macrophages in vitro to investigate TCG's impact on osteoclastogenesis. We further employed a small molecule pull-down assay to verify TCG's binding target within osteoclasts. Finally, we isolated primary mouse chondrocytes in vitro to study TCG's regulatory effects and mechanisms on chondrocyte survival. RESULTS TCG preserved subchondral bone integrity and protected articular cartilage in a rat OA model. Subsequently, in vitro experiments unveiled TCG's capability to inhibit osteoclastogenesis and function through binding to Ras association proximate 1 (Rap1) and inhibiting its activation. Further study demonstrated that TCG inhibited Rap1/integrin αvβ3/c-Src/Pyk2 signaling cascade, and consequently led to failed F-actin ring formation. Besides, TCG promoted the proliferation of mouse primary chondrocytes while suppressing apoptosis in vitro. This is attributed to TCG's ability to upregulate HIF1α, thereby promoting glycolysis. CONCLUSION TCG exerted inhibitory effects on osteoclastogenesis through binding to Rap1 and inhibiting Rap1 activation, consequently preventing subchondral bone loss. Moreover, TCG enhanced chondrocyte survival by upregulating HIF1α and promoting glycolysis. These dual mechanisms collectively provide a novel approach to prevented against cartilage degradation.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiahui Zhang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beite Ruan
- The Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaobing Xi
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuo Yang
- Chemical Biology Core Facility, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China
| | - Jianmin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyan Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xing Xu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Min Jiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
14
|
Roemer FW, Wirth W, Demehri S, Kijowski R, Jarraya M, Hayashi D, Eckstein F, Guermazi A. Imaging Biomarkers of Osteoarthritis. Semin Musculoskelet Radiol 2024; 28:14-25. [PMID: 38330967 DOI: 10.1055/s-0043-1776432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Currently no disease-modifying osteoarthritis drug has been approved for the treatment of osteoarthritis (OA) that can reverse, hold, or slow the progression of structural damage of OA-affected joints. The reasons for failure are manifold and include the heterogeneity of structural disease of the OA joint at trial inclusion, and the sensitivity of biomarkers used to measure a potential treatment effect.This article discusses the role and potential of different imaging biomarkers in OA research. We review the current role of radiography, as well as advances in quantitative three-dimensional morphological cartilage assessment and semiquantitative whole-organ assessment of OA. Although magnetic resonance imaging has evolved as the leading imaging method in OA research, recent developments in computed tomography are also discussed briefly. Finally, we address the experience from the Foundation for the National Institutes of Health Biomarker Consortium biomarker qualification study and the future role of artificial intelligence.
Collapse
Affiliation(s)
- Frank W Roemer
- Department of Radiology, Chobanian & Avedisian Boston University School of Medicine, Boston, Massachusetts
- Department of Radiology, Universitätsklinikum Erlangen & Friedrich Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang Wirth
- Center of Anatomy, and Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University, Salzburg, Austria
- Chondrometrics, GmbH, Freilassing, Germany
| | - Shadpour Demehri
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Richard Kijowski
- Department of Radiology, New York University Grossmann School of Medicine, New York, New York
| | - Mohamed Jarraya
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daichi Hayashi
- Department of Radiology, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Felix Eckstein
- Center of Anatomy, and Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University, Salzburg, Austria
- Chondrometrics, GmbH, Freilassing, Germany
| | - Ali Guermazi
- Department of Radiology, Chobanian & Avedisian Boston University School of Medicine, Boston, Massachusetts
- Department of Radiology, Boston VA Healthcare System, West Roxbury, Massachusetts
| |
Collapse
|
15
|
Bourne LE, Hesketh A, Sharma A, Bucca G, Bush PG, Staines KA. The effects of physiological and injurious hydrostatic pressure on murine ex vivo articular and growth plate cartilage explants: an RNAseq study. Front Endocrinol (Lausanne) 2023; 14:1278596. [PMID: 38144567 PMCID: PMC10740163 DOI: 10.3389/fendo.2023.1278596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Introduction Chondrocytes are continuously exposed to loads placed upon them. Physiological loads are pivotal to the maintenance of articular cartilage health, while abnormal loads contribute to pathological joint degradation. Similarly, the growth plate cartilage is subject to various loads during growth and development. Due to the high-water content of cartilage, hydrostatic pressure is considered one of the main biomechanical influencers on chondrocytes and has been shown to play an important role in the mechano-regulation of cartilage. Methods Herein, we conducted RNAseq analysis of ex vivo hip cap (articular), and metatarsal (growth plate) cartilage cultures subjected to physiological (5 MPa) and injurious (50 MPa) hydrostatic pressure, using the Illumina platform (n = 4 replicates). Results Several hundreds of genes were shown to be differentially modulated by hydrostatic pressure, with the majority of these changes evidenced in hip cap cartilage cultures (375 significantly upregulated and 322 downregulated in 5 MPa versus control; 1022 upregulated and 724 downregulated in 50 MPa versus control). Conversely, fewer genes were differentially affected by hydrostatic pressure in the metatarsal cultures (5 significantly upregulated and 23 downregulated in 5 MPa versus control; 7 significantly upregulated and 19 downregulated in 50 MPa versus control). Using Gene Ontology annotations for Biological Processes, in the hip cap data we identified a number of pathways that were modulated by both physiological and injurious hydrostatic pressure. Pathways upregulated in response to 50 MPa versus control, included those involved in the generation of precursor metabolites and cellular respiration. Biological processes that were downregulated in this tissue included ossification, connective tissue development, and chondrocyte differentiation. Discussion Collectively our data highlights the divergent chondrocyte phenotypes in articular and growth plate cartilage. Further, we show that the magnitude of hydrostatic pressure application has distinct effects on gene expression and biological processes in hip cap cartilage explants. Finally, we identified differential expression of a number of genes that have previously been identified as osteoarthritis risk genes, including Ctsk, and Chadl. Together these data may provide potential genetic targets for future investigations in osteoarthritis research and novel therapeutics.
Collapse
Affiliation(s)
- Lucie E. Bourne
- Centre for Lifelong Health, School of Applied Sciences, University of Brighton, Brighton, United Kingdom
| | - Andrew Hesketh
- Centre for Lifelong Health, School of Applied Sciences, University of Brighton, Brighton, United Kingdom
| | - Aikta Sharma
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Giselda Bucca
- Centre for Lifelong Health, School of Applied Sciences, University of Brighton, Brighton, United Kingdom
| | - Peter G. Bush
- Centre for Lifelong Health, School of Applied Sciences, University of Brighton, Brighton, United Kingdom
| | - Katherine A. Staines
- Centre for Lifelong Health, School of Applied Sciences, University of Brighton, Brighton, United Kingdom
| |
Collapse
|
16
|
Bjerre-Bastos JJ, Hamrouni N, Henrotin Y, Thudium CS, Bihlet AR. Joint biomarker response to mechanical stimuli in osteoarthritis - A scoping review. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100390. [PMID: 37885822 PMCID: PMC10598037 DOI: 10.1016/j.ocarto.2023.100390] [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] [Received: 01/09/2023] [Accepted: 08/04/2023] [Indexed: 10/28/2023] Open
Abstract
Objective Arthritic cartilage is primed for mechanical damage. Joint biochemical markers (JBM) could provide insight into the impact of mechanical stimulation on joint tissue turnover in osteoarthritis (OA) of potential use in clinical OA research and practice. However, existing studies of the acute impact of physical activities (PA) on JBM often contain risks of substantial bias. The purpose of this scoping review was to critically review and discuss existing reports of acute joint tissue turnover as reflected in JBM in relation to PA in OA and propose considerations for future research. Design We searched PubMed, Embase, and Scopus and reference lists for original reports on the acute impact of PA on JBM in human OA. Identified studies were reviewed by two reviewers forming the basis for the discussion of methodology. Results Search in databases resulted in nine eligible papers after full-text evaluation. Two additional papers were identified through reference lists, resulting in 11 papers included in this review. Ten investigated knee OA and one investigated hand OA. Biomarkers described were related to turnover of type II collagen, aggrecan, and cartilage oligomeric matrix protein. Conclusions The literature is dominated by small, simplistic studies, but suggests that mechanical stimulation can induce acute changes in joint biomarkers. In order to diminish the existing bias in future studies, it is important to recognize methodological considerations e.g. patient and biomarker selection as well as peri-interventional control. Common potential sources of bias include the acute shift in plasma volume due to cardiovascular stress and postural changes.
Collapse
Affiliation(s)
- Jonathan J. Bjerre-Bastos
- Xlab, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- NBCD A/S, Herlev, Denmark
- Department of Orthopedic Surgery, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Nizar Hamrouni
- Department of Orthopedic Surgery, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Yves Henrotin
- Musculoskeletal Innovative Research Lab (mSKIL), Arthropôle Liège, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège, CHU Sart-Tilman, 4000 Liège, Belgium
- Physical Therapy and Rehabilitation Department, Princess Paola Hospital, Vivalia, Rue du Vivier, 6900 Marche-en-Famenne, Belgium
- Artialis SA, CHU Sart-Tilman, GIGA Tower, level 3, 4000 Liège, Belgium
| | | | | |
Collapse
|
17
|
Wirth W, Ladel C, Maschek S, Wisser A, Eckstein F, Roemer F. Quantitative measurement of cartilage morphology in osteoarthritis: current knowledge and future directions. Skeletal Radiol 2023; 52:2107-2122. [PMID: 36380243 PMCID: PMC10509082 DOI: 10.1007/s00256-022-04228-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/29/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022]
Abstract
Quantitative measures of cartilage morphology ("cartilage morphometry") extracted from high resolution 3D magnetic resonance imaging (MRI) sequences have been shown to be sensitive to osteoarthritis (OA)-related change and also to treatment interventions. Cartilage morphometry is therefore nowadays widely used as outcome measure for observational studies and randomized interventional clinical trials. The objective of this narrative review is to summarize the current status of cartilage morphometry in OA research, to provide insights into aspects relevant for the design of future studies and clinical trials, and to give an outlook on future developments. It covers the aspects related to the acquisition of MRIs suitable for cartilage morphometry, the analysis techniques needed for deriving quantitative measures from the MRIs, the quality assurance required for providing reliable cartilage measures, and the appropriate participant recruitment criteria for the enrichment of study cohorts with knees likely to show structural progression. Finally, it provides an overview over recent clinical trials that relied on cartilage morphometry as a structural outcome measure for evaluating the efficacy of disease-modifying OA drugs (DMOAD).
Collapse
Affiliation(s)
- Wolfgang Wirth
- Department of Imaging & Functional Musculoskeletal Research, Institute of Anatomy & Cell Biology, Paracelsus Medical University Salzburg & Nuremberg, Strubergasse 21, 5020 Salzburg, Austria
- Ludwig Boltzmann Inst. for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
- Chondrometrics GmbH, Freilassing, Germany
| | | | - Susanne Maschek
- Department of Imaging & Functional Musculoskeletal Research, Institute of Anatomy & Cell Biology, Paracelsus Medical University Salzburg & Nuremberg, Strubergasse 21, 5020 Salzburg, Austria
- Chondrometrics GmbH, Freilassing, Germany
| | - Anna Wisser
- Department of Imaging & Functional Musculoskeletal Research, Institute of Anatomy & Cell Biology, Paracelsus Medical University Salzburg & Nuremberg, Strubergasse 21, 5020 Salzburg, Austria
- Ludwig Boltzmann Inst. for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
- Chondrometrics GmbH, Freilassing, Germany
| | - Felix Eckstein
- Department of Imaging & Functional Musculoskeletal Research, Institute of Anatomy & Cell Biology, Paracelsus Medical University Salzburg & Nuremberg, Strubergasse 21, 5020 Salzburg, Austria
- Ludwig Boltzmann Inst. for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
- Chondrometrics GmbH, Freilassing, Germany
| | - Frank Roemer
- Quantitative Imaging Center, Department of Radiology, Boston University School of Medicine, Boston, MA USA
- Department of Radiology, Universitätsklinikum Erlangen and Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
18
|
Hunter DJ, Collins JE, Deveza L, Hoffmann SC, Kraus VB. Biomarkers in osteoarthritis: current status and outlook - the FNIH Biomarkers Consortium PROGRESS OA study. Skeletal Radiol 2023; 52:2323-2339. [PMID: 36692532 PMCID: PMC10509067 DOI: 10.1007/s00256-023-04284-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 01/25/2023]
Abstract
Currently, no disease-modifying therapies are approved for osteoarthritis (OA) use. One obstacle to trial success in this field has been our existing endpoints' limited validity and responsiveness. To overcome this impasse, the Foundation for the NIH OA Biomarkers Consortium is focused on investigating biomarkers for a prognostic context of use for subsequent qualification through regulatory agencies. This narrative review describes this activity and the work underway, focusing on the PROGRESS OA study.
Collapse
Affiliation(s)
- David J Hunter
- Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine, University of Sydney, Australia and Rheumatology Department, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia.
| | - Jamie E Collins
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Leticia Deveza
- Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine, University of Sydney, Australia and Rheumatology Department, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Steven C Hoffmann
- Foundation for the National Institutes of Health, Bethesda, North, MD, USA
| | - Virginia B Kraus
- Duke Molecular Physiology Institute, and Department of Medicine|, Duke University, Durham, NC, USA
| |
Collapse
|
19
|
Karsdal MA, Tambiah J, Felson D, Ladel C, Nikolov NP, Hodgins D, Bihlet AR, Neogi T, Baatenburg de Jong C, Bay-Jensen AC, Baron R, Laslop A, Mobasheri A, Kraus VB. Reflections from the OARSI 2022 clinical trials symposium: The pain of OA-Deconstruction of pain and patient-reported outcome measures for the benefit of patients and clinical trial design. Osteoarthritis Cartilage 2023; 31:1293-1302. [PMID: 37380011 PMCID: PMC11184959 DOI: 10.1016/j.joca.2023.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 06/01/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) drug development is hampered by a number of challenges. One of the main challenges is the apparent discordance between pain and structure, which has had a significant impact on drug development programs and has led to hesitance among stakeholders. Since 2017, the Clinical Trials Symposium (CTS) has been hosted under the Osteoarthritis Research Society International (OARSI) leadership. OARSI and the CTS steering committee yearly invite and encourage discussions on selected special subject matter between regulators, drug developers, clinicians, clinical researchers, biomarker specialists, and basic scientists to progress drug development in the OA field. METHOD The main topic for the 2022 OARSI CTS was to elucidate the many facets of pain in OA and to enable a discussion between regulators (Food and Drug Administration (FDA) and the European Medicines Agency (EMA)) and drug developers to clarify outcomes and study designs for OA drug development. RESULTS Signs or symptoms indicative of nociceptive pain occur in 50-70% of OA patients, neuropathic-like pain in 15-30% of patients, and nociplastic pain in 15-50% of patients. Weight-bearing knee pain is associated with bone marrow lesions and effusions. There are currently no simple objective functional tests whose improvements correlate with patient perceptions. CONCLUSIONS The CTS participants, in collaboration with the FDA and EMA, raised several suggestions that they consider key to future clinical trials in OA including the need for more precise differentiation of pain symptoms and mechanisms, and methods to reduce placebo responses in OA trials.
Collapse
Affiliation(s)
- M A Karsdal
- Nordic Bioscience, Herlev, Denmark; Southern Danish University, Odense, Denmark.
| | - J Tambiah
- Biosplice Therapeutics, San Diego, USA
| | - D Felson
- Boston University School of Medicine, Boston, MA, USA
| | - C Ladel
- CHL4special Consultancy, Darmstadt, Germany
| | - N P Nikolov
- Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - D Hodgins
- Dynamic Metrics Limited, Codicote, UK
| | | | - T Neogi
- Boston University School of Medicine, Boston, MA, USA
| | | | | | - R Baron
- University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Kiel, Germany
| | - A Laslop
- Committee for Medicinal Products for Human Use (CHMP), European Medicines Agency, Amsterdam, the Netherlands; Bundesamt für Sicherheit im Gesundheitswesen (BASG), Vienna, Austria
| | - A Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania; Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liege, Belgium
| | - V B Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
20
|
Neogi T, Colloca L. Placebo effects in osteoarthritis: implications for treatment and drug development. Nat Rev Rheumatol 2023; 19:613-626. [PMID: 37697077 PMCID: PMC10615856 DOI: 10.1038/s41584-023-01021-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2023] [Indexed: 09/13/2023]
Abstract
Osteoarthritis (OA) is the most common form of arthritis worldwide, affecting ~500 million people, yet there are no effective treatments to halt its progression. Without any structure-modifying agents, management of OA focuses on ameliorating pain and improving function. Treatment approaches typically have modest efficacy, and many patients have contraindications to recommended pharmacological treatments. Drug development for OA is hindered by the gradual and progressive nature of the disease and the targeting of established disease in clinical trials. Additionally, new medications for OA cannot receive regulatory approval without demonstrating improvements in both structure (pathological features of OA) and symptoms (reduced pain and/or improved function). In clinical trials, people with OA show high 'placebo responses', which hamper the ability to identify new effective treatments. Placebo responses refer to the individual variability in response to placebos given in the context of clinical trials and other settings. Placebo effects refer specifically to short-lasting improvements in symptoms that occur because of physiological changes. To mitigate the effects of the placebo phenomenon, we must first understand what it is, how it manifests, how to identify placebo responders in OA trials and how these insights can be used to improve clinical trials in OA. Leveraging placebo responses and effects in clinical practice might provide additional avenues to augment symptom management of OA.
Collapse
Affiliation(s)
- Tuhina Neogi
- Section of Rheumatology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Luana Colloca
- Department of Pain and Translation Symptom Science, School of Nursing, University of Maryland, Baltimore, MD, USA.
- Placebo Beyond Opinions Center, School of Nursing, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
21
|
Skiöldebrand E, Adepu S, Lützelschwab C, Nyström S, Lindahl A, Abrahamsson-Aurell K, Hansson E. A randomized, triple-blinded controlled clinical study with a novel disease-modifying drug combination in equine lameness-associated osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100381. [PMID: 37416846 PMCID: PMC10320210 DOI: 10.1016/j.ocarto.2023.100381] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 07/08/2023] Open
Abstract
Objective This study aimed to test a novel treatment combination (TC) (equivalent to sildenafil, mepivacaine, and glucose) with disease-modifying properties compared to Celestone® bifas® (CB) in a randomized triple-blinded phase III clinical study in horses with mild osteoarthritis (OA). Joint biomarkers (reflecting the articular cartilage and subchondral bone remodelling) and clinical lameness were used as readouts to evaluate the treatment efficacy. Methods Twenty horses with OA-associated lameness in the carpal joint were included in the study and received either TC (n = 10) or CB (n = 10) drug intra-articularly-twice in the middle carpal joint with an interval of 2 weeks (visit 1 & 2). Clinical lameness was assessed both objectively (Lameness locator) and subjectively (visually). Synovial fluid and serum were sampled for quantification of the extracellular matrix (ECM) neo-epitope joint biomarkers represented by biglycan (BGN262) and cartilage oligomeric matrix protein (COMP156). Another two weeks later clinical lameness was recorded, and serum was collected for biomarkers analysis. The overall health status was compared pre and post-intervention by interviewing the trainer. Results Post-intervention, SF BGN262 levels significantly declined in TC (P = 0.002) and COMP156 levels significantly increased in CB (P = 0.002). The flexion test scores improved in the TC compared to CB (P =0.033) and also had an improved trotting gait quality (P =0.044). No adverse events were reported. Conclusion This is the first clinical study presenting companion diagnostics assisting in identifying OA phenotype and evaluating the efficacy and safety of a novel disease-modifying osteoarthritic drug.
Collapse
Affiliation(s)
- E. Skiöldebrand
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - S. Adepu
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - C. Lützelschwab
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - S. Nyström
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital, Gothenburg University, Gothenburg, Sweden
| | - A. Lindahl
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital, Gothenburg University, Gothenburg, Sweden
| | - K. Abrahamsson-Aurell
- Hallands Djursjukhus Kungsbacka Hästklinik, Älvsåkers Byväg 20, 434 95 Kungsbacka, Sweden
| | - E. Hansson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
22
|
Muthu S, Korpershoek JV, Novais EJ, Tawy GF, Hollander AP, Martin I. Failure of cartilage regeneration: emerging hypotheses and related therapeutic strategies. Nat Rev Rheumatol 2023:10.1038/s41584-023-00979-5. [PMID: 37296196 DOI: 10.1038/s41584-023-00979-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2023] [Indexed: 06/12/2023]
Abstract
Osteoarthritis (OA) is a disabling condition that affects billions of people worldwide and places a considerable burden on patients and on society owing to its prevalence and economic cost. As cartilage injuries are generally associated with the progressive onset of OA, robustly effective approaches for cartilage regeneration are necessary. Despite extensive research, technical development and clinical experimentation, no current surgery-based, material-based, cell-based or drug-based treatment can reliably restore the structure and function of hyaline cartilage. This paucity of effective treatment is partly caused by a lack of fundamental understanding of why articular cartilage fails to spontaneously regenerate. Thus, research studies that investigate the mechanisms behind the cartilage regeneration processes and the failure of these processes are critical to instruct decisions about patient treatment or to support the development of next-generation therapies for cartilage repair and OA prevention. This Review provides a synoptic and structured analysis of the current hypotheses about failure in cartilage regeneration, and the accompanying therapeutic strategies to overcome these hurdles, including some current or potential approaches to OA therapy.
Collapse
Affiliation(s)
- Sathish Muthu
- Orthopaedic Research Group, Coimbatore, Tamil Nadu, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, New Delhi, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore, India
| | - Jasmijn V Korpershoek
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Emanuel J Novais
- Unidade Local de Saúde do Litoral Alentejano, Orthopedic Department, Santiago do Cacém, Portugal
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gwenllian F Tawy
- Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester, UK
| | - Anthony P Hollander
- Institute of Lifecourse and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| |
Collapse
|
23
|
Gezer HH, Ostor A. What is new in pharmacological treatment for osteoarthritis? Best Pract Res Clin Rheumatol 2023; 37:101841. [PMID: 37302928 DOI: 10.1016/j.berh.2023.101841] [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: 05/03/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023]
Abstract
Osteoarthritis (OA) is a degenerative joint disease in which structural changes of hyaline articular cartilage, subchondral bone, ligaments, capsule, synovium, muscles, and periarticular changes are involved. The knee is the most commonly affected joint, followed by the hand, hip, spine, and feet. Different pathological mechanisms are at play in each of these various involvement sites. Although systemic inflammation is more prominent in hand OA, knee and hip OA have been associated with excessive joint load and injury. As OA has varied phenotypes and the primarily affected tissues differ, treatment options must be tailored accordingly. In recent years, ongoing efforts have been made to develop disease-modifying options that halt or slow disease progression. Many are still in clinical trials, and as insights into the pathogenesis of OA evolve, novel therapeutic strategies will be developed. In this chapter, we provide an overview of the novel and emerging strategies in the management of OA.
Collapse
Affiliation(s)
- Halise Hande Gezer
- Marmara University School of Medicine, PMR Department Rheumatology Division, Istanbul, Turkiye
| | - Andrew Ostor
- Cabrini Medical Centre, Monash University, Melbourne & ANU, Canberra, Australia.
| |
Collapse
|
24
|
Li S, Cao P, Chen T, Ding C. Latest insights in disease-modifying osteoarthritis drugs development. Ther Adv Musculoskelet Dis 2023; 15:1759720X231169839. [PMID: 37197024 PMCID: PMC10184265 DOI: 10.1177/1759720x231169839] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 03/29/2023] [Indexed: 05/19/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent and severely debilitating disease with an unmet medical need. In order to alleviate OA symptoms or prevent structural progression of OA, new drugs, particularly disease-modifying osteoarthritis drugs (DMOADs), are required. Several drugs have been reported to attenuate cartilage loss or reduce subchondral bone lesions in OA and thus potentially be DMOADs. Most biologics (including interleukin-1 (IL-1) and tumor necrosis factor (TNF) inhibitors), sprifermin, and bisphosphonates failed to yield satisfactory results when treating OA. OA clinical heterogeneity is one of the primary reasons for the failure of these clinical trials, which can require different therapeutic approaches based on different phenotypes. This review describes the latest insights into the development of DMOADs. We summarize in this review the efficacy and safety profiles of various DMOADs targeting cartilage, synovitis, and subchondral bone endotypes in phase 2 and 3 clinical trials. To conclude, we summarize the reasons for clinical trial failures in OA and suggest possible solutions.
Collapse
Affiliation(s)
- Shengfa Li
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peihua Cao
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tianyu Chen
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Changhai Ding
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, 261 Industry Road, Guangzhou 510515, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- Clinical Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
25
|
Schnitzer T, Pueyo M, Deckx H, van der Aar E, Bernard K, Hatch S, van der Stoep M, Grankov S, Phung D, Imbert O, Chimits D, Muller K, Hochberg MC, Bliddal H, Wirth W, Eckstein F, Conaghan PG. Evaluation of S201086/GLPG1972, an ADAMTS-5 inhibitor, for the treatment of knee osteoarthritis in ROCCELLA: a phase 2 randomized clinical trial. Osteoarthritis Cartilage 2023:S1063-4584(23)00737-9. [PMID: 37059327 DOI: 10.1016/j.joca.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/16/2023]
Abstract
OBJECTIVE To evaluate the efficacy and safety of the anti-catabolic ADAMTS-5 inhibitor S201086/GLPG1972 for the treatment of symptomatic knee osteoarthritis. DESIGN ROCCELLA (NCT03595618) was a randomized, double-blind, placebo-controlled, dose-ranging, phase 2 trial in adults (aged 40-75 years) with knee osteoarthritis. Participants had moderate-to-severe pain in the target knee, Kellgren-Lawrence grade 2 or 3 and Osteoarthritis Research Society International joint space narrowing (grade 1 or 2). Participants were randomized 1:1:1:1 to once-daily oral S201086/GLPG1972 75, 150 or 300 mg, or placebo for 52 weeks. The primary endpoint was change from baseline to week 52 in central medial femorotibial compartment cartilage thickness (cMFTC) assessed quantitatively by magnetic resonance imaging. Secondary endpoints included change from baseline to week 52 in radiographic joint space width, Western Ontario and McMaster Universities Osteoarthritis Index total and subscores, and pain (visual analogue scale). Treatment-emergent adverse events (TEAEs) were also recorded. RESULTS Overall, 932 participants were enrolled. No significant differences in cMFTC cartilage loss were observed between placebo and S201086/GLPG1972 therapeutic groups: placebo vs 75 mg, P = 0.165; vs 150 mg, P = 0.939; vs 300 mg, P = 0.682. No significant differences in any of the secondary endpoints were observed between placebo and treatment groups. Similar proportions of participants across treatment groups experienced TEAEs. CONCLUSIONS Despite enrolment of participants who experienced substantial cartilage loss over 52 weeks, during the same time period, S201086/GLPG1972 did not significantly reduce rates of cartilage loss or modify symptoms in adults with symptomatic knee osteoarthritis.
Collapse
Affiliation(s)
- T Schnitzer
- Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| | - M Pueyo
- Institut de Recherches Internationales Servier (IRIS), Suresnes, France
| | - H Deckx
- Galapagos NV, Mechelen, Belgium.
| | | | - K Bernard
- Institut de Recherches Internationales Servier (IRIS), Suresnes, France.
| | - S Hatch
- Galapagos Inc., Waltham, Massachusetts, USA.
| | | | - S Grankov
- Institut de Recherches Internationales Servier (IRIS), Suresnes, France.
| | - D Phung
- Galapagos NV, Mechelen, Belgium.
| | - O Imbert
- Institut de Recherches Internationales Servier (IRIS), Suresnes, France.
| | - D Chimits
- Institut de Recherches Internationales Servier (IRIS), Suresnes, France.
| | - K Muller
- Galapagos NV, Mechelen, Belgium.
| | - M C Hochberg
- Departments of Medicine and Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| | - H Bliddal
- The Parker Institute, Copenhagen, Denmark.
| | - W Wirth
- Chondrometrics GmbH, Ainring, Germany; Institute of Anatomy and Cell Biology and Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University, Salzburg, Austria.
| | - F Eckstein
- Chondrometrics GmbH, Ainring, Germany; Institute of Anatomy and Cell Biology and Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University, Salzburg, Austria.
| | - P G Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, and National Institute for Health and Care Research (NIHR) Leeds Biomedical Research Centre, Leeds, UK.
| |
Collapse
|
26
|
Lin J, Jia S, Zhang W, Nian M, Liu P, Yang L, Zuo J, Li W, Zeng H, Zhang X. Recent Advances in Small Molecule Inhibitors for the Treatment of Osteoarthritis. J Clin Med 2023; 12:jcm12051986. [PMID: 36902773 PMCID: PMC10004353 DOI: 10.3390/jcm12051986] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/23/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
Osteoarthritis refers to a degenerative disease with joint pain as the main symptom, and it is caused by various factors, including fibrosis, chapping, ulcers, and loss of articular cartilage. Traditional treatments can only delay the progression of osteoarthritis, and patients may need joint replacement eventually. As a class of organic compound molecules weighing less than 1000 daltons, small molecule inhibitors can target proteins as the main components of most drugs clinically. Small molecule inhibitors for osteoarthritis are under constant research. In this regard, by reviewing relevant manuscripts, small molecule inhibitors targeting MMPs, ADAMTS, IL-1, TNF, WNT, NF-κB, and other proteins were reviewed. We summarized these small molecule inhibitors with different targets and discussed disease-modifying osteoarthritis drugs based on them. These small molecule inhibitors have good inhibitory effects on osteoarthritis, and this review will provide a reference for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Shicheng Jia
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shantou University Medical College, Shantou 515041, China
| | - Weifei Zhang
- Department of Bone and Joint, Peking University Shenzhen Hospital, Shenzhen 518036, China
- National & Local Joint Engineering Research Center of Orthopedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Mengyuan Nian
- Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Peng Liu
- Department of Bone and Joint, Peking University Shenzhen Hospital, Shenzhen 518036, China
- National & Local Joint Engineering Research Center of Orthopedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Li Yang
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Jianwei Zuo
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Wei Li
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Correspondence: (W.L.); (H.Z.); (X.Z.)
| | - Hui Zeng
- Department of Bone and Joint, Peking University Shenzhen Hospital, Shenzhen 518036, China
- National & Local Joint Engineering Research Center of Orthopedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Correspondence: (W.L.); (H.Z.); (X.Z.)
| | - Xintao Zhang
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Correspondence: (W.L.); (H.Z.); (X.Z.)
| |
Collapse
|
27
|
Dilley JE, Bello MA, Roman N, McKinley T, Sankar U. Post-traumatic osteoarthritis: A review of pathogenic mechanisms and novel targets for mitigation. Bone Rep 2023. [DOI: 10.1016/j.bonr.2023.101658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
28
|
Wen P, Liu R, Wang J, Wang Y, Song W, Zhang Y. Bibliometric insights from publications on subchondral bone research in osteoarthritis. Front Physiol 2022; 13:1095868. [PMID: 36620224 PMCID: PMC9814489 DOI: 10.3389/fphys.2022.1095868] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Background: The role of subchondral bone in the pathogenesis of osteoarthritis has received continuous attention worldwide. To date, no comprehensive bibliometric analysis of this topic has been carried out. The purpose of this study was to investigate the knowledge landscape, hot spots, and research trends in subchondral bone research through bibliometrics. Methods: Web of Science Core Collection database was used to collect articles and reviews on subchondral bone in osteoarthritis published between 2003 and 2022. CiteSpace, VOSviewer, Scimago Graphica, and a bibliometric online analysis platform (http://bibliometric.com/) were used to visualize the knowledge network of countries, institutions, authors, references, and keywords in this field. Both curve fitting and statistical plotting were performed using OriginPro, while correlation analysis was done using SPSS. Results: A total of 3,545 articles and reviews were included. The number of publications on subchondral bone showed an exponential growth trend. The US produced the most (980), followed by China (862) and the United Kingdom (364). Scientific output and gross domestic product were significantly correlated (r = .948, p < .001). The University of California System and Professor Pelletier Jean-Pierre were the most prolific institutions and influential authors, respectively. The most active and influential journal for subchondral bone research was Osteoarthritis and Cartilage. The majority of papers were financed by NSFC (474, 13.4%), followed by HHS (445, 12.6%), and NIH (438, 12.4%). In recent years, hot keywords have focused on the research of pathomechanisms (e.g., inflammation, apoptosis, pathogenesis, cartilage degeneration/repair, angiogenesis, TGF beta) and therapeutics (e.g., regeneration, stromal cell, mesenchymal stem cell). Conclusion: Subchondral bone research in osteoarthritis is flourishing. Current topics and next research trends would be centered on the pathomechanisms of cellular and molecular interactions in the subchondral bone microenvironment in the development of osteoarthritis and the exploration of targeted treatment medicines for the altered subchondral bone microenvironment.
Collapse
Affiliation(s)
| | | | | | | | - Wei Song
- *Correspondence: Wei Song, ; Yumin Zhang,
| | | |
Collapse
|
29
|
Kim H, Seo J, Lee Y, Park K, Perry TA, Arden NK, Mobasheri A, Choi H. The current state of the osteoarthritis drug development pipeline: a comprehensive narrative review of the present challenges and future opportunities. Ther Adv Musculoskelet Dis 2022; 14:1759720X221085952. [PMID: 36504595 PMCID: PMC9732806 DOI: 10.1177/1759720x221085952] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 02/18/2022] [Indexed: 12/12/2022] Open
Abstract
In this narrative review article, we critically assess the current state of the osteoarthritis (OA) drug development pipeline. We discuss the current state-of-the-art in relation to the development and evaluation of candidate disease-modifying OA drugs (DMOADs) and the limitations associated with the tools and methodologies that are used to assess outcomes in OA clinical trials. We focus on the definition of DMOADs, highlight the need for an updated definition in the form of a consensus statement from all the major stakeholders, including academia, industry, regulatory agencies, and patient organizations, and provide a summary of the results of recent clinical trials of novel DMOAD candidates. We propose that DMOADs should be more appropriately targeted and investigated according to the emerging clinical phenotypes and molecular endotypes of OA. Based on the findings from recent clinical trials, we propose key topics and directions for the development of future DMOADs.
Collapse
Affiliation(s)
- Heungdeok Kim
- Institute of Bio Innovation Research, Kolon
Life Science, Inc., Seoul, South Korea
| | - Jinwon Seo
- Institute of Bio Innovation Research, Kolon
Life Science, Inc., Seoul, South Korea
| | - Yunsin Lee
- Institute of Bio Innovation Research, Kolon
Life Science, Inc., Seoul, South Korea
| | - Kiwon Park
- Institute of Bio Innovation Research, Kolon
Life Science, Inc., Seoul, South Korea
| | - Thomas A. Perry
- Centre for Osteoarthritis Pathogenesis Versus
Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford,
UK
| | - Nigel K. Arden
- Versus Arthritis Centre for Sport, Exercise and
Osteoarthritis, University of Oxford, Oxford, UK
- Botnar Research Centre, Nuffield Orthopaedic
Centre, Oxford, UK
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and
Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State
Research Institute Center for Innovative Medicine, Vilnius, Lithuania
- Department of Orthopedics and Department of
Rheumatology and Clinical Immunology, University Medical Center Utrecht,
Utrecht, The Netherlands
- Department of Joint Surgery, The First
Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- World Health Organization Collaborating Center
for Public Health Aspects of Musculoskeletal Health and Aging, Université de
Liège, Liège, Belgium
| | - Heonsik Choi
- Healthcare Research Institute, Kolon Advanced
Research Center, Kolon Industries, Inc., 110 Magokdong-ro, Gangseo-gu, Seoul
07793, South Korea
| |
Collapse
|
30
|
Han S. Osteoarthritis year in review 2022: biology. Osteoarthritis Cartilage 2022; 30:1575-1582. [PMID: 36150676 DOI: 10.1016/j.joca.2022.09.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 02/02/2023]
Abstract
The field of osteoarthritis (OA) biology is rapidly evolving and brilliant progress has been made this year as well. Landmark studies of OA biology published in 2021 and early 2022 were selected through PubMed search by personal opinion. These papers were classified by their molecular mechanisms, and it was largely divided into the intracellular signaling mechanisms and the inter-compartment interaction in chondrocyte homeostasis and OA progression. The intracellular signaling mechanisms involving OA progression included (1) Piezo1/transient receptor potential channels of the vanilloid subtype (TRPV) 4-mediated calcium signaling, (2) mechanical load-F-box and WD repeat domain containing 7 (FBXW7) in chondrocyte senescence, (3) mechanical loading-primary cilia-hedgehog signaling, (4) low grade inflammation by toll-like receptor (TLR)-CD14-lipopolysaccharide-binding protein (LBP) complex and inhibitor of NF-κB kinase (IKK) β-nuclear factor kappa B (NF-κB) signaling, (5) selenium pathway and reactive oxygen species (ROS) production, (6) G protein-coupled receptor (GPCR) and cyclic adenosine monophosphate (cAMP) signaling, (7) peroxisome proliferator-activated receptor α (PPARα)-acyl-CoA thioesterase 12 (ACOT12)-mediated de novo lipogenesis and (8) hypoxia-disruptor of telomeric silencing 1-like (DOT1L)-H3-lysine 79 (H3K79) methylation pathway. The studies on inter-compartment or intercellular interaction in OA progression included the following subjects; (1) the anabolic role of lubricin, glycoprotein from superficial zone cells, (2) osteoclast-chondrocyte interaction via exosomal miRNA and sphingosine 1-phosphate (S1P), (3) senescent fibroblast-like synoviocyte and chondrocyte interaction, (4) synovial macrophage and chondrocyte interaction through Flightless I, (5) αV integrin-mediated transforming growth factor beta (TGFβ) activation by mechanical loading, and (6) osteocytic TGFβ in subchondral bone thickening. Despite the disastrous Covid-19 pandemic, many outstanding studies have expanded the boundary of OA biology. They provide both critical insight into the pathophysiology as well as clues for the treatment of OA.
Collapse
Affiliation(s)
- S Han
- Laboratory for for Arthritis and Cartilage Biology, Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
31
|
Biasizzo M, Javoršek U, Vidak E, Zarić M, Turk B. Cysteine cathepsins: A long and winding road towards clinics. Mol Aspects Med 2022; 88:101150. [PMID: 36283280 DOI: 10.1016/j.mam.2022.101150] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/03/2022]
Abstract
Biomedical research often focuses on properties that differentiate between diseased and healthy tissue; one of the current focuses is elevated expression and altered localisation of proteases. Among these proteases, dysregulation of cysteine cathepsins can frequently be observed in inflammation-associated diseases, which tips the functional balance from normal physiological to pathological manifestations. Their overexpression and secretion regularly exhibit a strong correlation with the development and progression of such diseases, making them attractive pharmacological targets. But beyond their mostly detrimental role in inflammation-associated diseases, cysteine cathepsins are physiologically highly important enzymes involved in various biological processes crucial for maintaining homeostasis and responding to different stimuli. Consequently, several challenges have emerged during the efforts made to translate basic research data into clinical applications. In this review, we present both physiological and pathological roles of cysteine cathepsins and discuss the clinical potential of cysteine cathepsin-targeting strategies for disease management and diagnosis.
Collapse
Affiliation(s)
- Monika Biasizzo
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Urban Javoršek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Eva Vidak
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Miki Zarić
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
32
|
Shentu CY, Yan G, Xu DC, Chen Y, Peng LH. Emerging pharmaceutical therapeutics and delivery technologies for osteoarthritis therapy. Front Pharmacol 2022; 13:945876. [PMID: 36467045 PMCID: PMC9712996 DOI: 10.3389/fphar.2022.945876] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 11/03/2022] [Indexed: 10/03/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common joint degenerative diseases in the world. At present, the management of OA depends on the lifestyle modification and joint replacement surgery, with the lifespan of prosthesis quite limited yet. Effective drug treatment of OA is essential. However, the current drugs, such as the non-steroidal anti-inflammatory drugs and acetaminophen, as well as glucosamine, chondroitin sulfate, hyaluronic acid, are accompanied by obvious side effects, with the therapeutic efficacy to be enhanced. Recently, novel reagents such as IL-1 antagonists and nerve growth factor inhibitors have entered clinical trials. Moreover, increasing evidence demonstrated that active ingredients of natural plants have great potential for treating OA. Meanwhile, the use of novel drug delivery strategies may overcome the shortcomings of conventional preparations and enhance the bioavailability of drugs, as well as decrease the side effects significantly. This review therefore summarizes the pathological mechanisms, management strategies, and research progress in the drug molecules including the newly identified active ingredient derived from medicinal plants for OA therapy, with the drug delivery technologies also summarized, with the expectation to provide the summary and outlook for developing the next generation of drugs and preparations for OA therapy.
Collapse
Affiliation(s)
- Cheng-Yu Shentu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ge Yan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Dong-Chen Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yong Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Li-Hua Peng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| |
Collapse
|
33
|
Emulsion Gel: a Dual Drug Delivery Platform for Osteoarthritis Treatment. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00282-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
34
|
Fan T, Chen S, Zeng M, Li J, Wang X, Ruan G, Cao P, Zhang Y, Chen T, Ou Q, Wang Q, Wluka AE, Cicuttini F, Ding C, Zhu Z. Osteophytes mediate the associations between cartilage morphology and changes in knee symptoms in patients with knee osteoarthritis. Arthritis Res Ther 2022; 24:217. [PMID: 36076236 PMCID: PMC9454107 DOI: 10.1186/s13075-022-02905-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Aims To investigate whether the associations between cartilage defects and cartilage volumes with changes in knee symptoms were mediated by osteophytes. Methods Data from the Vitamin D Effects on Osteoarthritis (VIDEO) study were analyzed as a cohort. The Western Ontario and McMaster Universities Osteoarthritis Index was used to assess knee symptoms at baseline and follow-up. Osteophytes, cartilage defects, and cartilage volumes were measured using magnetic resonance imaging at baseline. Associations between cartilage morphology and changes in knee symptoms were assessed using linear regression models, and mediation analysis was used to test whether these associations were mediated by osteophytes. Results A total of 334 participants (aged 50 to 79 years) with symptomatic knee osteoarthritis were included in the analysis. Cartilage defects were significantly associated with change in total knee pain, change in weight-bearing pain, and change in non-weight-bearing pain after adjustment for age, sex, body mass index, and intervention. Cartilage volume was significantly associated with change in weight-bearing pain and change in physical dysfunction after adjustment. Lateral tibiofemoral and patellar osteophyte mediated the associations of cartilage defects with change in total knee pain (49–55%) and change in weight-bearing pain (61–62%) and the association of cartilage volume with change in weight-bearing pain (27–30%) and dysfunction (24–25%). Both cartilage defects and cartilage volume had no direct effects on change in knee symptoms. Conclusions The significant associations between cartilage morphology and changes in knee symptoms were indirect and were partly mediated by osteophytes. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02905-8.
Collapse
Affiliation(s)
- Tianxiang Fan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shibo Chen
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Muhui Zeng
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jia Li
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoshuai Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guangfeng Ruan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Peihua Cao
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan Zhang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Tianyu Chen
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qianhua Ou
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qianyi Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Anita E Wluka
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Flavia Cicuttini
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China. .,Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia. .,Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia. .,Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Zhaohua Zhu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China. .,Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
35
|
Park J, Lee SY. A review of osteoarthritis signaling intervention using small-molecule inhibitors. Medicine (Baltimore) 2022; 101:e29501. [PMID: 35960127 PMCID: PMC9371536 DOI: 10.1097/md.0000000000029501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Numerous small-molecule inhibitors (SMIs) have been approved as adjuvant or first-line therapies for malignancies. Based on cancer treatment using SMIs, next-generation SMIs that can be used to optimize the therapeutic index, overcome drug resistance, and establish combination therapies are in development. Osteoarthritis (OA) is the most common chronic joint disease with senescence, and there are various approaches to OA treatment; however, the gold standard treatment is controversial. Therefore, in this manuscript, we demonstrated the potential of using SMIs in OA treatment and described the general strategies for using SMIs in OA treatment.
Collapse
Affiliation(s)
- Junyong Park
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Sang Yeob Lee
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, Dong-A University, Busan, Republic of Korea
- * Correspondence: Sang Yeob Lee, MD, PhD, Division of Rheumatology, Department of Internal Medicine, College of Medicine, Dong-A University, 26 Daeshingongwon-ro, Seo-Gu, Busan 49201, Republic of Korea (e-mail: )
| |
Collapse
|
36
|
Kinetic Characterization of Cerium and Gallium Ions as Inhibitors of Cysteine Cathepsins L, K, and S. Int J Mol Sci 2022; 23:ijms23168993. [PMID: 36012257 PMCID: PMC9409168 DOI: 10.3390/ijms23168993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022] Open
Abstract
Heavy metal ions can disrupt biological functions via multiple molecular mechanisms, including inhibition of enzymes. We investigate the interactions of human papain-like cysteine endopeptidases cathepsins L, K, and S with gallium and cerium ions, which are associated with medical applications. We compare these results with zinc and lead, which are known to inhibit thiol enzymes. We show that Ga3+, Ce3+, and Ce4+ ions inhibit all tested peptidases with inhibition constants in the low micromolar range (between 0.5 µM and 10 µM) which is comparable to Zn2+ ions, whereas inhibition constants of Pb2+ ions are one order of magnitude higher (30 µM to 150 µM). All tested ions are linear specific inhibitors of cathepsin L, but cathepsins K and S are inhibited by Ga3+, Ce3+, and Ce4+ ions via hyperbolic inhibition mechanisms. This indicates a mode of interaction different from that of Zn2+ and Pb2+ ions, which act as linear specific inhibitors of all peptidases. All ions also inhibit the degradation of insoluble elastin, which is a common target of these peptidases in various inflammatory diseases. Our results suggest that these ions and their compounds have the potential to be used as cysteine cathepsin inhibitors in vitro and possibly in vivo.
Collapse
|
37
|
Cheng J, Sun Y, Ma Y, Ao Y, Hu X, Meng Q. Engineering of MSC-Derived Exosomes: A Promising Cell-Free Therapy for Osteoarthritis. MEMBRANES 2022; 12:membranes12080739. [PMID: 36005656 PMCID: PMC9413347 DOI: 10.3390/membranes12080739] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is characterized by progressive cartilage degeneration with increasing prevalence and unsatisfactory treatment efficacy. Exosomes derived from mesenchymal stem cells play an important role in alleviating OA by promoting cartilage regeneration, inhibiting synovial inflammation and mediating subchondral bone remodeling without the risk of immune rejection and tumorigenesis. However, low yield, weak activity, inefficient targeting ability and unpredictable side effects of natural exosomes have limited their clinical application. At present, various approaches have been applied in exosome engineering to regulate their production and function, such as pretreatment of parental cells, drug loading, genetic engineering and surface modification. Biomaterials have also been proved to facilitate efficient delivery of exosomes and enhance treatment effectiveness. Here, we summarize the current understanding of the biogenesis, isolation and characterization of natural exosomes, and focus on the large-scale production and preparation of engineered exosomes, as well as their therapeutic potential in OA, thus providing novel insights into exploring advanced MSC-derived exosome-based cell-free therapy for the treatment of OA.
Collapse
Affiliation(s)
- Jin Cheng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
| | - Yixin Sun
- Peking Unversity First Hospital, Peking University Health Science Center, Beijing 100034, China;
| | - Yong Ma
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
| | - Yingfang Ao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
| | - Xiaoqing Hu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
- Correspondence: (X.H.); (Q.M.); Tel.: +86-010-8226-5680 (Q.M.)
| | - Qingyang Meng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
- Correspondence: (X.H.); (Q.M.); Tel.: +86-010-8226-5680 (Q.M.)
| |
Collapse
|
38
|
Zou N, Liu R, Li C. Cathepsin K+ Non-Osteoclast Cells in the Skeletal System: Function, Models, Identity, and Therapeutic Implications. Front Cell Dev Biol 2022; 10:818462. [PMID: 35912093 PMCID: PMC9326176 DOI: 10.3389/fcell.2022.818462] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Cathepsin K (Ctsk) is a cysteine protease of the papain superfamily initially identified in differentiated osteoclasts; it plays a critical role in degrading the bone matrix. However, subsequent in vivo and in vitro studies based on animal models elucidate novel subpopulations of Ctsk-expressing cells, which display markers and properties of mesenchymal stem/progenitor cells. This review introduces the function, identity, and role of Ctsk+ cells and their therapeutic implications in related preclinical osseous disorder models. It also summarizes the available in vivo models for studying Ctsk+ cells and their progeny. Further investigations of detailed properties and mechanisms of Ctsk+ cells in transgenic models are required to guide potential therapeutic targets in multiple diseases in the future.
Collapse
Affiliation(s)
- Nanyu Zou
- Department of Endocrinology, Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, China
| | - Ran Liu
- Department of Endocrinology, Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, China
| | - Changjun Li
- Department of Endocrinology, Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- *Correspondence: Changjun Li,
| |
Collapse
|
39
|
Khoshdel A, Forootan M, Afsharinasab M, Rezaian M, Abbasifard M. Assessment of the circulatory concentrations of cathepsin D, cathepsin K, and alpha-1 antitrypsin in patients with knee osteoarthritis. Ir J Med Sci 2022:10.1007/s11845-022-03061-3. [PMID: 35749030 DOI: 10.1007/s11845-022-03061-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 06/08/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Evidence has shown that cysteine protease enzymes, such as cathepsin D, cathepsin A, cathepsin K, and alpha-1 antitrypsin (AAT) are involved in the chronic degenerative joint process. This study aimed to determine the potential involvement of cathepsin K, cathepsin D, and AAT in patients with osteoarthritis (OA). METHODS This study was performed on 31 patients with knee OA and 29 age- and sex-matched healthy subjects (both with Fars ethnicity from Iran). American College of Rheumatology (ACR) criteria were used to diagnose OA patients. The clinical status of the patients was scored by Western Ontario McMaster Universities Osteoarthritis (WOMAC), and pain intensity was measured by the Visual Analog Scale (VAS). The serum level of AAT was measured using high-resolution cellulose acetate electrophoresis. Additionally, serum levels of cathepsin D and cathepsin K were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS The findings showed that the serum level of cathepsin K was significantly increased in OA patients compared to healthy subjects (P = 0.01), while there was no significant difference between serum level of cathepsin D in study groups (P = 0.2). In addition, the serum concentration of AAT was significantly decreased in OA patients compared to healthy subjects (P = 0.003). There was a significant correlation between WOMAC score and age (r = 0.644, P = 0.0001) and VAS (r = 0.866, P < 0.0001) in OA patients. CONCLUSIONS The decreased level of AAT in OA patients and a rise in serum level of cathepsin K are involved in the pathogenesis of OA via stimulation of bone resorption and cartilage degradation.
Collapse
Affiliation(s)
- Alireza Khoshdel
- Nervous System Stem Cells Research Center , Semnan University of Medical Sciences, Semnan, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mohammad Forootan
- Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mehdi Afsharinasab
- Department of Clinical Biochemistry, Schoolof Medicine, Tehran University of Medical Sciences, Rafsanjan, Iran
| | - Mohsen Rezaian
- Epidemiology and Biostatistics Department, Occupational Environmental Research Center, Rafsanjan Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mitra Abbasifard
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
40
|
Joint Cartilage in Long-Duration Spaceflight. Biomedicines 2022; 10:biomedicines10061356. [PMID: 35740378 PMCID: PMC9220015 DOI: 10.3390/biomedicines10061356] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/05/2022] [Accepted: 06/06/2022] [Indexed: 12/14/2022] Open
Abstract
This review summarizes the current literature available on joint cartilage alterations in long-duration spaceflight. Evidence from spaceflight participants is currently limited to serum biomarker data in only a few astronauts. Findings from analogue model research, such as bed rest studies, as well as data from animal and cell research in real microgravity indicate that unloading and radiation exposure are associated with joint degeneration in terms of cartilage thinning and changes in cartilage composition. It is currently unknown how much the individual cartilage regions in the different joints of the human body will be affected on long-term missions beyond the Low Earth Orbit. Given the fact that, apart from total joint replacement or joint resurfacing, currently no treatment exists for late-stage osteoarthritis, countermeasures might be needed to avoid cartilage damage during long-duration missions. To plan countermeasures, it is important to know if and how joint cartilage and the adjacent structures, such as the subchondral bone, are affected by long-term unloading, reloading, and radiation. The use of countermeasures that put either load and shear, or other stimuli on the joints, shields them from radiation or helps by supporting cartilage physiology, or by removing oxidative stress possibly help to avoid OA in later life following long-duration space missions. There is a high demand for research on the efficacy of such countermeasures to judge their suitability for their implementation in long-duration missions.
Collapse
|
41
|
Oo WM, Hunter DJ. Repurposed and investigational disease-modifying drugs in osteoarthritis (DMOADs). Ther Adv Musculoskelet Dis 2022; 14:1759720X221090297. [PMID: 35619876 PMCID: PMC9128067 DOI: 10.1177/1759720x221090297] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/10/2022] [Indexed: 12/12/2022] Open
Abstract
In spite of a major public health burden with increasing prevalence, current osteoarthritis (OA) management is largely palliative with an unmet need for effective treatment. Both industry and academic researchers have invested a vast amount of time and financial expense to discover the first diseasing-modifying osteoarthritis drugs (DMOADs), with no regulatory success so far. In this narrative review, we discuss repurposed drugs as well as investigational agents which have progressed into phase II and III clinical trials based on three principal endotypes: bone-driven, synovitis-driven and cartilage-driven. Then, we will briefly describe the recent failures and lessons learned, promising findings from predefined post hoc analyses and insights gained, novel methodologies to enhance future success and steps underway to overcome regulatory hurdles.
Collapse
Affiliation(s)
- Win Min Oo
- Rheumatology Department, Royal North Shore Hospital, and Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Physical Medicine and Rehabilitation, Mandalay General Hospital, University of Medicine, Mandalay, Mandalay, Myanmar
| | - David J. Hunter
- Rheumatology Department, Royal North Shore Hospital, and Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2065, Australia
| |
Collapse
|
42
|
Vincent TL, Alliston T, Kapoor M, Loeser RF, Troeberg L, Little CB. Osteoarthritis Pathophysiology: Therapeutic Target Discovery may Require a Multifaceted Approach. Clin Geriatr Med 2022; 38:193-219. [PMID: 35410676 PMCID: PMC9107912 DOI: 10.1016/j.cger.2021.11.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Molecular understanding of osteoarthritis (OA) has greatly increased through careful analysis of tissue samples, preclinical models, and large-scale agnostic "-omic" studies. There is broad acceptance that systemic and biomechanical signals affect multiple tissues of the joint, each of which could potentially be targeted to improve patient outcomes. In this review six experts in different aspects of OA pathogenesis provide their independent view on what they believe to be good tractable approaches to OA target discovery. We conclude that molecular discovery has been high but future transformative studies require a multidisciplinary holistic approach to develop therapeutic strategies with high clinical efficacy.
Collapse
Affiliation(s)
- Tonia L Vincent
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Mohit Kapoor
- Department of Surgery and Laboratory Medicine and Pathobiology, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, University of Toronto, Toronto, Canada
| | - Richard F Loeser
- Department of Medicine, Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Linda Troeberg
- University of East Anglia, Norwich Medical School, Norwich NR4 7UQ, UK
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute University of Sydney Faculty of Medicine and Health at Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia.
| |
Collapse
|
43
|
|
44
|
|
45
|
Connection between Mesenchymal Stem Cells Therapy and Osteoclasts in Osteoarthritis. Int J Mol Sci 2022; 23:ijms23094693. [PMID: 35563083 PMCID: PMC9102843 DOI: 10.3390/ijms23094693] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
The use of mesenchymal stem cells constitutes a promising therapeutic approach, as it has shown beneficial effects in different pathologies. Numerous in vitro, pre-clinical, and, to a lesser extent, clinical trials have been published for osteoarthritis. Osteoarthritis is a type of arthritis that affects diarthritic joints in which the most common and studied effect is cartilage degradation. Nowadays, it is known that osteoarthritis is a disease with a very powerful inflammatory component that affects the subchondral bone and the rest of the tissues that make up the joint. This inflammatory component may induce the differentiation of osteoclasts, the bone-resorbing cells. Subchondral bone degradation has been suggested as a key process in the pathogenesis of osteoarthritis. However, very few published studies directly focus on the activity of mesenchymal stem cells on osteoclasts, contrary to what happens with other cell types of the joint, such as chondrocytes, synoviocytes, and osteoblasts. In this review, we try to gather the published bibliography in relation to the effects of mesenchymal stem cells on osteoclastogenesis. Although we find promising results, we point out the need for further studies that can support mesenchymal stem cells as a therapeutic tool for osteoclasts and their consequences on the osteoarthritic joint.
Collapse
|
46
|
Astrike-Davis EM, Coryell P, Loeser RF. Targeting cellular senescence as a novel treatment for osteoarthritis. Curr Opin Pharmacol 2022; 64:102213. [PMID: 35447516 DOI: 10.1016/j.coph.2022.102213] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/12/2022] [Indexed: 12/12/2022]
Abstract
Cellular senescence is associated with normal development and wound healing, but has also been implicated in the pathogenesis of numerous aging-related diseases including osteoarthritis (OA). Treatment strategies for OA are being developed that target senescent cells and the paracrine and autocrine secretions of the senescence-associated secretory phenotype (SASP). The field of potential therapies continues to expand as new mechanistic targets of cell senescence and the SASP are identified. Ongoing pre-clinical and clinical studies of drugs targeting cellular senescence yield significant promise, but have yet to demonstrate long-term efficacy. Therapeutic targeting of senescence is challenged by the diverse phenotypes of senescent cells, which can vary depending on age, species, tissue source, and type of physiologic stressor. Accordingly, there remains considerable demand for more studies to further develop and assess senotherapeutics as disease-modifying treatments for OA.
Collapse
Affiliation(s)
- Emma M Astrike-Davis
- Division of Rheumatology, Allergy, and Immunology, The Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Philip Coryell
- Division of Rheumatology, Allergy, and Immunology, The Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Richard F Loeser
- Division of Rheumatology, Allergy, and Immunology, The Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
47
|
New Directions in the Development of Pharmacotherapy for Osteoarthrosis Based on Modern Concepts of the Disease Pathogenesis (A Review). Pharm Chem J 2022. [DOI: 10.1007/s11094-022-02572-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
48
|
Carpintero-Fernández P, Varela-Eirín M, García-Yuste A, López-Díaz I, Caeiro JR, Mayán MD. Osteoarthritis: Mechanistic Insights, Senescence, and Novel Therapeutic Opportunities. Bioelectricity 2022; 4:39-47. [PMID: 39355566 PMCID: PMC11441363 DOI: 10.1089/bioe.2021.0039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disease. In the last years, the research community has focused on understanding the molecular mechanisms that led to the pathogenesis of the disease, trying to identify different molecular and clinical phenotypes along with the discovery of new therapeutic opportunities. Different types of cell-to-cell communication mechanisms have been proposed to contribute to OA progression, including mechanisms mediated by connexin43 (Cx43) channels or by small extracellular vesicles. Furthermore, changes in the chondrocyte phenotype such as cellular senescence have been proposed as new contributors of the OA progression, changing the paradigm of the disease. The use of different drugs able to restore chondrocyte phenotype, to reduce cellular senescence and senescence-associated secretory phenotype components, and to modulate ion channel activity or Cx43 appears to be promising therapeutic strategies for the different types of OA. In this review, we aim to summarize the current knowledge in OA phenotypes related with aging and tissue damage and the new therapeutic opportunities currently available.
Collapse
Affiliation(s)
- Paula Carpintero-Fernández
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - Marta Varela-Eirín
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Alejandro García-Yuste
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - Iñaki López-Díaz
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - José Ramón Caeiro
- Department of Orthopaedic Surgery and Traumatology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - María D Mayán
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| |
Collapse
|
49
|
Henrotin Y. Osteoarthritis in year 2021: biochemical markers. Osteoarthritis Cartilage 2022; 30:237-248. [PMID: 34798278 DOI: 10.1016/j.joca.2021.11.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/30/2021] [Accepted: 11/01/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To summarize recent scientific advances in protein-derived soluble biomarkers of osteoarthritis. DESIGN A systematic search on the PubMed electronic database of clinical studies on protein-derived soluble biochemical markers of osteoarthritis in humans that were published between January 1st 2020 and March 31th 2021. The studies were selected on the basis of objective criteria and summarized in a table. Then they were described in a narrative review. RESULTS Out of 1971 publications, 48 fulfilled all selection criteria and 16 were selected by the author for the narrative review. The papers were classified according their clinical significance as defined in the BIPEDS classification. Two papers investigated the "burden of disease", two were dedicated to "investigative biomarkers", four papers question the "prognosis", three the "efficacy of treatment" and five the "diagnosis and phenotyping" value of protein-derived biomarkers. CONCLUSIONS Currently, biomarkers research is focused on their use as tools to identify molecular endotypes and clinical phenotypes and to facilitate patient screening and monitoring in clinical trials. This approach should allow a more targeted management of patients suffering from osteoarthritis.
Collapse
Affiliation(s)
- Y Henrotin
- musculoSKeletal Innovative research Lab (mSKIL), Institute of Pathology, Level 5, CHU Sart-Tilman, Center for Interdisciplinary Research on Medicines (CIRM), Department of Motricity Sciences, University of Liège, Belgium; Department of Physical Therapy and Rehabilitation, Princess Paola Hospital, Vivalia, Marche-en-Famenne, Belgium.
| |
Collapse
|
50
|
Jiang Y. Osteoarthritis year in review 2021: biology. Osteoarthritis Cartilage 2022; 30:207-215. [PMID: 34801671 DOI: 10.1016/j.joca.2021.11.009] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 02/02/2023]
Abstract
This year in review on osteoarthritis biology summarizes a series of research articles published between the 2020 and 2021 Osteoarthritis Research Society International (OARSI) World Congress. Research hightlights were selected and discussed based on the new discoveries of OA's cellular molecular mechanism, anatomical signatures, potential therapeutic targets, and regenerative therapy. The recently developed potential therapeutic targets are summarized, and the research focuses on TGFβ and WNT signaling in joint tissue homeostasis, joint aging and the dynamic of synolytics in OA joint, and the roles of TRP2, LDHA, OSCAR in cartilage homeostasis and OA joints are highlighted. Subsquencially, new anatomical structures and OA features are introduced, such as synovitis-induced venous portal circulation, horiozontal fissures between cartilage and subchondral bone, the cellular derivation of osteophytes formation, OA subtypes, and subchondral remodeling and pain biology. Then, research on the possibility of tissue regeneration in OA joints are discussed; skeletal stem cells in OA cartilage regeneration, and preclinical results of regenerative therapy for meniscus tear and osteochondral tissue morphoghesis are included. At last, the clinical evidence of the importance of delivery site of bone marrow stem cells for OA treatment is discussed. These findings represent advances in our understanding of OA pathophysiology.
Collapse
Affiliation(s)
- Y Jiang
- Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|