1
|
Thudium CS, Rasmussen S, Karsdal MA, Bay-Jensen AC. Association between type III collagen degradation and local tissue damage of a single joint. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100527. [PMID: 39502930 PMCID: PMC11535998 DOI: 10.1016/j.ocarto.2024.100527] [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: 04/05/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
Objective The development of disease-modifying drugs is limited by OA's heterogeneity and the challenge of defining clinical endpoints. Serological biomarkers are considered potential surrogate endpoints, but their contribution from single joints to systemic levels in OA patients is unclear. In this exploratory study we longitudinally assessed systemic biomarker levels' response to tissue damage and healing before and after surgery in patients undergoing knee or hip joint replacement revision for aseptic failure. Patients with chronic pain associated with a prior hip or knee arthroplasty, but not receiving revision surgery were included as control. Method The serological biomarker of MMP mediated type III collagen degradation C3M, associated with synovial tissue degradation, was measured at baseline before revision surgery, after revision surgery and at a 6-month follow-up in 48 patients with aseptic loosening of a knee or hip prosthesis and in 18 patients with chronic pain from a hip or knee prosthesis. Longitudinal changes in biomarkers were modeled using linear mixed models. Results No differences between the aseptic loosening and chronic pain groups were observed at baseline. Revision surgery in the aseptic loosening group led to a swift increase in C3M, which normalized within 2-3 months. No changes in biomarker level were observed in chronic pain patients over three months. Conclusion These findings suggest that tissue damage in a single joint significantly impacts systemic biomarker levels and underscores the relevance of systemic biomarkers in assessing local tissue remodeling.
Collapse
Affiliation(s)
| | - Sten Rasmussen
- Department of Clinical Medicine, Aalborg University, Denmark
| | | | | |
Collapse
|
2
|
Eckstein F, Maschek S, Wirth W, Ladel C, Bihlet AR, Knight C, Somberg K, Zhao L. Unbiased analysis of knee cartilage thickness change over three years after sprifermin vs. placebo treatment - A post-hoc analysis from the phase 2B FORWARD study. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100513. [PMID: 39286575 PMCID: PMC11403365 DOI: 10.1016/j.ocarto.2024.100513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Objective Post-treatment cartilage morphometry in the FORWARD study was performed without blinding to MRI acquisition order, involving potential reader bias. Here we obtained unbiased estimates of cartilage change post-treatment, reading year (Y)2 and Y5 MRIs with blinding to time point. We studied whether post-treatment cartilage thickness change differed between sprifermin- and placebo-treated knees. Methods FORWARD was a 5-year randomized control trial in 549 knee osteoarthritis patients. Here, Y2/Y5 images were analyzed with blinding to relative temporal order and treatment group. Cartilage change during Y2→Y5 was obtained in 337 participants: n = 57 treated with placebo intra-articular injections every 6 months (q6M); n = 69 with 30 μg sprifermin every 12 months (q12 M), n = 67 with 30 μg q6M, n = 73 with 100 μg q12 M, and n = 71 with 100 μg q6M between baseline (BL) and 18 M. Total femorotibial joint (TFTJ) cartilage thickness was the primary analytic focus. Results TFTJ cartilage thickness change during Y2→Y5 was -26μm (SD64; 95%CI -32,-19) across the cohort; no statistically significant difference (p = 0.80) was observed between Sprifermin treated or placebo arms (one-way ANOVA). All groups lost cartilage, but the treatment-related difference in cartilage thickness in Sprifermin arms relative to placebo at Y2 was maintained until Y5. Annualized cartilage change in placebo participants was -8.2 μm (SD21; 95%CI -14,-2.5) during Y2→Y5 vs. -5.4 μm (SD27; 95%CI -13,1.8) during BL→Y2; no significant difference was identified (t-test). Conclusion FORWARD is the first study evaluating post-treatment benefits of a potential disease modifying osteoarthritis drug. Cartilage thickness gained with 100 μg sprifermin at Y2 is maintained to Y5 and thus appears viable and sustainable.This is a post-hoc analysis of the FORWARD trial: ClinicalTrials.gov Identifier: NCT01919164.
Collapse
Affiliation(s)
- Felix Eckstein
- Research Program for Musculoskeletal Imaging, Center for Anatomy & Cell Biology, Paracelsus Medical University (PMU), Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University (PMU), Salzburg, Austria
- Chondrometrics GmbH, Freilassing, Germany
| | - Susanne Maschek
- Research Program for Musculoskeletal Imaging, Center for Anatomy & Cell Biology, Paracelsus Medical University (PMU), Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University (PMU), Salzburg, Austria
- Chondrometrics GmbH, Freilassing, Germany
| | - Wolfgang Wirth
- Research Program for Musculoskeletal Imaging, Center for Anatomy & Cell Biology, Paracelsus Medical University (PMU), Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University (PMU), Salzburg, Austria
- Chondrometrics GmbH, Freilassing, Germany
| | | | | | | | | | | |
Collapse
|
3
|
Roemer FW, Jansen MP, Maschek S, Mastbergen SC, Marijnissen AK, Wisser A, Heiss R, Weinans HH, Blanco FJ, Berenbaum F, Kloppenburg M, Haugen IK, Eckstein F, Hunter DJ, Guermazi A, Wirth W. Fluctuation of Bone Marrow Lesions and Inflammatory MRI Markers over 2 Years and Concurrent Associations with Quantitative Cartilage Loss. Cartilage 2024:19476035241287694. [PMID: 39460605 DOI: 10.1177/19476035241287694] [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] [Indexed: 10/28/2024] Open
Abstract
OBJECTIVE To assess whether change of semiquantitatively magnetic resonance imaging (MRI)-defined bone marrow lesions (BMLs) and inflammatory markers is associated with change in quantitatively-assessed cartilage loss in the femorotibial joint (FTJ) in knees with radiographic osteoarthritis (OA) over 24 months. DESIGN Participants were included from the IMI-APPROACH and the Osteoarthritis Initiative FNIH studies. Semiquantitative MRI assessment was performed for BMLs, Hoffa- and effusion-synovitis. Quantitative cartilage thickness measurements were performed manually. Definitions of change included number of subregions with BMLs, change in sum and change in maximum increase in size. Change in Hoffa-synovitis and effusion-synovitis was categorized in addition. Between-group comparisons regarding cartilage loss in the FTJ, medial and lateral compartments were performed using analysis of variance (ANOVA). RESULTS A total of 629 participants were included. Knees without any BMLs at baseline (BL) and follow-up (FU) had significantly less cartilage loss compared to the other subgroups. Change in both directions in the sum score of BMLs was associated with increased rates of cartilage loss. Maximum increase in size of BMLs was associated with increased rates of cartilage loss (FTJ increase by 2 grades -0.183 mm, 95% CI [-0.335, -0.031], by 3 grades -0.306 mm, [-0.511, -0.101]). Worsening of Hoffa-synovitis was associated with increased rates of cartilage loss. CONCLUSION Knees without BMLs at BL and FU showed lowest rates of cartilage loss. Knees with an increase in BML size showed increased rates of concurrent cartilage loss. Approaches with the aim to inhibit BML development, avoidance of increase in size and avoidance of Hoffa-synovitis worsening may have beneficial effects on cartilage loss.
Collapse
Affiliation(s)
- Frank W Roemer
- Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
- Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | | | | | | | - Anna Wisser
- Chondrometrics GmbH, Freilassing, Germany
- Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
| | - Rafael Heiss
- Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | | | - Francis Berenbaum
- AP-HP Saint- Antoine Hospital, Paris, France
- Sorbonne University, Paris, France
| | | | | | - Felix Eckstein
- Chondrometrics GmbH, Freilassing, Germany
- Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
| | - David J Hunter
- Royal North Shore Hospital and Sydney Musculoskeletal Health, Kolling Institute, University of Sydney, St. Leonards, NSW, Australia
| | - Ali Guermazi
- Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
- VA Boston Healthcare System, West Roxbury, MA, USA
| | - Wolfgang Wirth
- Chondrometrics GmbH, Freilassing, Germany
- Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
| |
Collapse
|
4
|
Eckstein F, Wisser A, Maschek S, Wirth W, Ladel C, Bihlet AR, Knight C, Somberg K, Zhao L. Is detection of disease-modifying osteoarthritis drug treatment more effective when performing cartilage morphometry without blinding to MR image acquisition order? Osteoarthritis Cartilage 2024; 32:1346-1351. [PMID: 38844160 DOI: 10.1016/j.joca.2024.05.012] [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: 01/19/2024] [Revised: 05/02/2024] [Accepted: 05/28/2024] [Indexed: 06/16/2024]
Abstract
OBJECTIVE We here explore whether observed treatment effects of a putative disease-modifying osteoarthritis drug (DMOAD) are greater when cartilage morphometry is performed with rather than without knowledge of magnetic resonance imaging (MRI) acquisition order (unblinded/blinded to time point). METHODS In the FORWARD (FGF-18 Osteoarthritis Randomized Controlled Trial with Administration of Repeated Doses) randomized controlled trial, 549 knee osteoarthritis patients were randomized 1:1:1:1:1 to three once-weekly intra-articular injections of placebo, 30 µg sprifermin every 6 or 12 months (M), or 100 µg every 6/12 M. After year 2, cartilage segmentation of BL through 24 M MRIs was performed, with blinding to acquisition order. After year 5, 24 and 60 M MRIs were analyzed together, with unknown relative order, but with segmented BL images as reference (24 M unblinded vs. BL), by the same operators. Total femorotibial joint cartilage thickness (TFTJ_ThC) change was obtained for 352 participants analyzed under both conditions. RESULTS Twenty-four-month data read unblinded to order revealed a -35 ± 44 µm lower TFTJ_ThC than blinded analysis (all groups: lower/upper bounds -120/+51 µm; correlation r2 = 97%). With unblinded analysis, the placebo group lost -46 ± 57 µm TFTJ_ThC over 24 M, whereas 100 µg/every 6 M lost -2.2 ± 73 µm (difference =44 µm [95% CI: 22, 66]). With blinded analysis, placebo lost -11 ± 53 µm, whereas 100 µg/every 6 M gained 30 ± 62 µm (difference = 40 µm [95% CI: 21, 60]). 100 µg sprifermin injected every 6 M showed statistically significant (p < 0.001) treatment effects on TFTJ_ThC, with Cohen D = -0.66 for unblinded and D = -0.69 for blinded analysis. CONCLUSIONS These results do not reveal that detection of proposed DMOAD treatment is enhanced with MRIs read unblinded to order; rather, the sensitivity is similar to blinded analysis. Choices on blinded vs. unblinded analysis may thus be based on other criteria.
Collapse
Affiliation(s)
- Felix Eckstein
- Research Program for Musculoskeletal Imaging, Center for Anatomy & Cell Biology, Paracelsus Medical University (PMU), Salzburg, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University (PMU), Salzburg, Austria; Chondrometrics GmbH, Freilassing, Germany.
| | - Anna Wisser
- Research Program for Musculoskeletal Imaging, Center for Anatomy & Cell Biology, Paracelsus Medical University (PMU), Salzburg, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University (PMU), Salzburg, Austria; Chondrometrics GmbH, Freilassing, Germany
| | - Susanne Maschek
- Research Program for Musculoskeletal Imaging, Center for Anatomy & Cell Biology, Paracelsus Medical University (PMU), Salzburg, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University (PMU), Salzburg, Austria; Chondrometrics GmbH, Freilassing, Germany
| | - Wolfgang Wirth
- Research Program for Musculoskeletal Imaging, Center for Anatomy & Cell Biology, Paracelsus Medical University (PMU), Salzburg, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University (PMU), Salzburg, Austria; Chondrometrics GmbH, Freilassing, Germany
| | | | | | | | | | | |
Collapse
|
5
|
Pande S, Pati F, Chakraborty P. Harnessing Peptide-Based Hydrogels for Enhanced Cartilage Tissue Engineering. ACS APPLIED BIO MATERIALS 2024; 7:5885-5905. [PMID: 39159490 DOI: 10.1021/acsabm.4c00879] [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: 08/21/2024]
Abstract
Cartilage tissue engineering remains a formidable challenge due to its complex, avascular structure and limited regenerative capacity. Traditional approaches, such as microfracture, autografts, and stem cell delivery, often fail to restore functional tissue adequately. Recently, there has been a surge in the exploration of new materials that mimic the extracellular microenvironment necessary to guide tissue regeneration. This review investigates the potential of peptide-based hydrogels as an innovative solution for cartilage regeneration. These hydrogels, formed via supramolecular self-assembly, exhibit excellent properties, including biocompatibility, ECM mimicry, and controlled biodegradation, making them highly suitable for cartilage tissue engineering. This review explains the structure of cartilage and the principles of supramolecular and peptide hydrogels. It also delves into their specific properties relevant to cartilage regeneration. Additionally, this review presents recent examples and a comparative analysis of various peptide-based hydrogels used for cartilage regeneration. The review also addresses the translational challenges of these materials, highlighting regulatory hurdles and the complexities of clinical application. This comprehensive investigation provides valuable insights for biomedical researchers, tissue engineers, and clinical professionals aiming to enhance cartilage repair methodologies.
Collapse
Affiliation(s)
- Shreya Pande
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | - Priyadarshi Chakraborty
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| |
Collapse
|
6
|
Eckstein F, Wirth W, Putz R. Sexual dimorphism in articular tissue anatomy - Key to understanding sex differences in osteoarthritis? Osteoarthritis Cartilage 2024; 32:1019-1031. [PMID: 38871022 DOI: 10.1016/j.joca.2024.05.014] [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: 11/15/2023] [Revised: 05/06/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVE Osteoarthritis (OA) prevalence and incidence varies between women and men, but it is unknown whether this follows sex-specific differences in systemic factors (e.g. hormones) and/or differences in pre-morbid joint anatomy. We recognize that classifications of sex within humans cannot be reduced to female/male, but given the lack of literature on non-binary individuals, this review is limited to the sexual dimorphism of articular morphotypes. METHODS Based on a Pubmed search using relevant terms, and input from experts, we selected articles based on the authors' judgment of their relevance, interest, originality, and scientific quality; no "hard" bibliometric measures were used to evaluate their quality or importance. Focus was on clinical rather than pre-clinical studies, with most (imaging) data being available for the knee joint. RESULTS After introducing "sexual dimorphism", the specific literature on articular morphotypes is reviewed, structured by: radiographic joint space width (JSW), meniscus, ligaments, articular cartilage morphology, articular cartilage composition and deformation, and articular tissue response to treatment. CONCLUSIONS Sex-specific differences were clearly observed for JSW, meniscus damage, ligament size, and cartilage morphometry (volume, thickness, and surface areas) but not for cartilage composition. Ligament and cartilage measures were smaller in women even after matching for confounders. Taken together, the findings indicate that female (knee) joints may be structurally more vulnerable and at greater risk of OA. The "one size/sex fits all" approach must be abandoned in OA research, and all observational and interventional studies should report their results for sex-specific strata, at least in pre-specified secondary or post-hoc analyses.
Collapse
MESH Headings
- Humans
- Female
- Male
- Cartilage, Articular/pathology
- Cartilage, Articular/diagnostic imaging
- Sex Characteristics
- Osteoarthritis, Knee/diagnostic imaging
- Osteoarthritis, Knee/pathology
- Knee Joint/diagnostic imaging
- Knee Joint/pathology
- Sex Factors
- Osteoarthritis/diagnostic imaging
- Osteoarthritis/pathology
- Menisci, Tibial/diagnostic imaging
- Menisci, Tibial/pathology
- Menisci, Tibial/anatomy & histology
- Radiography
- Ligaments, Articular/anatomy & histology
- Ligaments, Articular/pathology
- Ligaments, Articular/diagnostic imaging
Collapse
Affiliation(s)
- Felix Eckstein
- Research Program for Musculoskeletal Imaging, Center for Anatomy and Cell Biology, Paracelsus Medical University, Salzburg, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University, Salzburg, Austria; Chondrometrics GmbH, Freilassing, Germany.
| | - Wolfgang Wirth
- Research Program for Musculoskeletal Imaging, Center for Anatomy and Cell Biology, Paracelsus Medical University, Salzburg, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation (LBIAR), Paracelsus Medical University, Salzburg, Austria; Chondrometrics GmbH, Freilassing, Germany
| | - Reinhard Putz
- Anatomische Anstalt, Ludwig Maximilians Universität München, Munich, Germany
| |
Collapse
|
7
|
Luo P, Lu L, Xu R, Jiang L, Li G. Gaining Insight into Updated MR Imaging for Quantitative Assessment of Cartilage Injury in Knee Osteoarthritis. Curr Rheumatol Rep 2024; 26:311-320. [PMID: 38809506 DOI: 10.1007/s11926-024-01152-x] [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] [Accepted: 05/24/2024] [Indexed: 05/30/2024]
Abstract
PURPOSE OF THE REVIEW Knee Osteoarthritis (KOA) entails progressive cartilage degradation, reviewed via MRI for morphology, biochemical composition, and microtissue alterations, discussing clinical advantages, limitations, and research applicability. RECENT FINDINGS Compositional MRI, like T2/T2* mapping, T1rho mapping, gagCEST, dGEMRIC, sodium imaging, diffusion-weighted imaging, and diffusion-tensor imaging, provide insights into cartilage injury in KOA. These methods quantitatively measure collagen, glycosaminoglycans, and water content, revealing important information about biochemical compositional and microstructural alterations. Innovative techniques like hybrid multi-dimensional MRI and diffusion-relaxation correlation spectrum imaging show potential in depicting initial cartilage changes at a sub-voxel level. Integration of automated image analysis tools addressed limitations in manual cartilage segmentation, ensuring robust and reproducible assessments of KOA cartilage. Compositional MRI techniques reveal microstructural changes in cartilage. Multi-dimensional MR imaging assesses biochemical alterations in KOA-afflicted cartilage, aiding early degeneration identification. Integrating artificial intelligence enhances cartilage analysis, optimal diagnostic accuracy for early KOA detection and monitoring.
Collapse
Affiliation(s)
- Peng Luo
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai, 200437, China
| | - Li Lu
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai, 200437, China
| | - Run Xu
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai, 200437, China
| | - Lei Jiang
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai, 200437, China
| | - Guanwu Li
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai, 200437, China.
| |
Collapse
|
8
|
Chalian M, Pooyan A, Alipour E, Roemer FW, Guermazi A. What is New in Osteoarthritis Imaging? Radiol Clin North Am 2024; 62:739-753. [PMID: 39059969 DOI: 10.1016/j.rcl.2024.02.006] [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: 07/28/2024]
Abstract
Osteoarthritis (OA) is the leading joint disorder globally, affecting a significant proportion of the population. Recent studies have changed our understanding of OA, viewing it as a complex pathology of the whole joint with a multifaceted etiology, encompassing genetic, biological, and biomechanical elements. This review highlights the role of imaging in diagnosing and monitoring OA. Today's role of radiography is discussed, while also elaborating on the advances in computed tomography and magnetic resonance imaging, discussing semiquantitative methods, quantitative morphologic and compositional techniques, and giving an outlook on the potential role of artificial intelligence in OA research.
Collapse
Affiliation(s)
- Majid Chalian
- Department of Radiology, University of Washington, Seattle, USA; Musculoskeletal Imaging and Intervention, University of Washington, UW Radiology, Roosevelt Clinic, 4245 Roosevelt Way, NE Box 354755, Seattle, WA 98105, USA
| | - Atefe Pooyan
- Department of Radiology, University of Washington, Seattle, USA; Musculoskeletal Imaging and Intervention, University of Washington, UW Radiology, Roosevelt Clinic, 4245 Roosevelt Way, NE Box 354755, Seattle, WA 98105, USA
| | - Ehsan Alipour
- Department of Radiology, University of Washington, Seattle, USA; Musculoskeletal Imaging and Intervention, University of Washington, UW Radiology, Roosevelt Clinic, 4245 Roosevelt Way, NE Box 354755, Seattle, WA 98105, USA
| | - Frank W Roemer
- Department of Radiology, Friedrich-Alexander University Erlangen-Nürnberg; Universitätsklinikum Erlangen, Erlangen, Germany; Department of Radiology, Quantitative Imaging Center, Boston University School of Medicine
| | - Ali Guermazi
- Department of Radiology, Quantitative Imaging Center, Boston University School of Medicine; Department of Radiology, VA Boston Healthcare System, Boston, MA, USA.
| |
Collapse
|
9
|
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
|
10
|
Huang K, Liu X, Qin H, Li Y, Zhu J, Yin B, Zheng Q, Zuo C, Cao H, Tong Z, Sun Z. FGF18 encoding circular mRNA-LNP based on glycerolipid engineering of mesenchymal stem cells for efficient amelioration of osteoarthritis. Biomater Sci 2024; 12:4427-4439. [PMID: 39037353 DOI: 10.1039/d4bm00668b] [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: 07/23/2024]
Abstract
Mesenchymal stem cells (MSCs) exhibit substantial potential for osteoarthritis (OA) therapy through cartilage regeneration, yet the realization of optimal therapeutic outcomes is hampered by their limited intrinsic reparative capacities. Herein, MSCs are engineered with circular mRNA (cmRNA) encoding fibroblast growth factor 18 (FGF18) encapsulated within lipid nanoparticles (LNP) derived from a glycerolipid to facilitate OA healing. A proprietary biodegradable and ionizable glycerolipid, TG6A, with branched tails and five ester bonds, forms LNP exhibiting above 9-fold and 41-fold higher EGFP protein expression in MSCs than commercial LNP from DLin-MC3-DMA and ALC-0315, respectively. The introduction of FGF18 not only augmented the proliferative capacity of MSCs but also upregulated the expression of chondrogenic genes and glycosaminoglycan (GAG) content. Additionally, FGF18 enhanced the production of proteoglycans and type II collagen in chondrocyte pellet cultures in a three-dimensional culture. In an OA rat model, transplantation with FGF18-engineered MSCs remarkably preserved cartilage integrity and facilitated functional repair of cartilage lesions, as evidenced by thicker cartilage layers, reduced histopathological scores, maintenance of zone structure, and incremental type II collagen and extracellular matrix (ECM) deposition. Taken together, our findings suggest that TG6A-based LNP loading with cmRNA for engineering MSCs present an innovative strategy to overcome the current limitations in OA treatment.
Collapse
Affiliation(s)
- Ke Huang
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
- Suzhou Industrial Park Monash Research Institute of Science and Technology, Suzhou, 215000, China
| | - Xiaoyun Liu
- Jiangsu Purecell Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Haitang Qin
- Jiangsu Purecell Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Yingwen Li
- Suzhou CureMed Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Jiafeng Zhu
- Suzhou CureMed Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Bo Yin
- National University of Singapore (Suzhou) Research Institute, Suzhou, 215123, China.
| | - Qijun Zheng
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
- Suzhou Industrial Park Monash Research Institute of Science and Technology, Suzhou, 215000, China
| | - Chijian Zuo
- Suzhou CureMed Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Hui Cao
- Jiangsu Purecell Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Zhenbo Tong
- Southeast University-Monash University Joint Research Institute, Suzhou 215125, China
| | - Zhenhua Sun
- Suzhou CureMed Biopharma Technology Co., Ltd, Suzhou 215125, China.
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Mohamed KE, Larsen AT, Melander S, Andersen F, Kerrn EB, Karsdal MA, Henriksen K. The dual amylin and calcitonin receptor agonist KBP-336 elicits a unique combination of weight loss, antinociception and bone protection - a novel disease-modifying osteoarthritis drug. Arthritis Res Ther 2024; 26:129. [PMID: 38997785 PMCID: PMC11241783 DOI: 10.1186/s13075-024-03361-2] [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: 02/05/2024] [Accepted: 06/30/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Despite the extensive research to provide a disease-modifying osteoarthritis drug (DMOAD), there is still no approved DMOAD. Dual amylin and calcitonin receptor agonists (DACRA) can provide metabolic benefits along with antinociceptive and potential structural preserving effects. In these studies, we tested a DACRA named KBP-336 on a metabolic model of OA in meniscectomised (MNX) rats. METHODS We evaluated KBP-336's effect on pain-like symptoms in Sprague Dawley (SD) rats on high-fat diet (HFD) that underwent meniscectomy using the von Frey test to measure the 50% paw withdrawal threshold (PWT) and analyzed using one-way ANOVA. Short in vivo studies and in vitro cell receptor expression systems were used to illustrate receptor pharmacology. RESULTS After 30 weeks on HFD, including an 8-week treatment, female MNX animals receiving KBP-336 4.5 nmol/Kg/72 h had lower body weight and smaller adipose tissues than their vehicle-treated counterparts. After 20 weeks on HFD, including an 8-week treatment, male rats receiving KBP-336 had lower body weight than the vehicle group. In both the female and male rats, the MNX groups on KBP-336 treatment had a higher PWT than the vehicle-treated MNX group. Aiming to identify the receptor influencing pain alleviation, KBP-336 was compared to the long-acting human calcitonin (hCTA). Single-dose studies on 12-week-old male rats showed that hCTA lowers CTX-I without affecting food intake, confirming its calcitonin receptor selectivity. On the metabolic OA model with 18 weeks of HFD, including 6-week treatment, hCTA at 100 nmol/Kg/24 h and KBP-336 at 0.5, 1.5, and 4.5 nmol/Kg/72 h produced significantly higher PWT in MNX animals compared to MNX animals on vehicle treatment. hCTA and KBP-336 at 0.5 nmol/Kg did not affect body weight and fat tissues. CONCLUSION Overall, KBP-336 improved the pain observed in the metabolic OA model. Calcitonin receptor activation proved to be essential in this antinociceptive effect.
Collapse
Affiliation(s)
- Khaled Elhady Mohamed
- Nordic Bioscience Biomarkers and Research, Herlev Hovedgade 207, Herlev, DK-2730, Denmark.
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| | - Anna Thorsø Larsen
- Nordic Bioscience Biomarkers and Research, Herlev Hovedgade 207, Herlev, DK-2730, Denmark
| | - Simone Melander
- Nordic Bioscience Biomarkers and Research, Herlev Hovedgade 207, Herlev, DK-2730, Denmark
| | - Frederik Andersen
- Nordic Bioscience Biomarkers and Research, Herlev Hovedgade 207, Herlev, DK-2730, Denmark
| | - Ellen Barendorff Kerrn
- Nordic Bioscience Biomarkers and Research, Herlev Hovedgade 207, Herlev, DK-2730, Denmark
| | - Morten Asser Karsdal
- Nordic Bioscience Biomarkers and Research, Herlev Hovedgade 207, Herlev, DK-2730, Denmark
- KeyBioscience AG, Stans, Switzerland
| | - Kim Henriksen
- Nordic Bioscience Biomarkers and Research, Herlev Hovedgade 207, Herlev, DK-2730, Denmark
- KeyBioscience AG, Stans, Switzerland
- Department of Molecular and Medical Biology, Roskilde University Center, Roskilde, Denmark
| |
Collapse
|
14
|
Li HZ, Zhang JL, Yuan DL, Xie WQ, Ladel CH, Mobasheri A, Li YS. Role of signaling pathways in age-related orthopedic diseases: focus on the fibroblast growth factor family. Mil Med Res 2024; 11:40. [PMID: 38902808 PMCID: PMC11191355 DOI: 10.1186/s40779-024-00544-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Fibroblast growth factor (FGF) signaling encompasses a multitude of functions, including regulation of cell proliferation, differentiation, morphogenesis, and patterning. FGFs and their receptors (FGFR) are crucial for adult tissue repair processes. Aberrant FGF signal transduction is associated with various pathological conditions such as cartilage damage, bone loss, muscle reduction, and other core pathological changes observed in orthopedic degenerative diseases like osteoarthritis (OA), intervertebral disc degeneration (IVDD), osteoporosis (OP), and sarcopenia. In OA and IVDD pathologies specifically, FGF1, FGF2, FGF8, FGF9, FGF18, FGF21, and FGF23 regulate the synthesis, catabolism, and ossification of cartilage tissue. Additionally, the dysregulation of FGFR expression (FGFR1 and FGFR3) promotes the pathological process of cartilage degradation. In OP and sarcopenia, endocrine-derived FGFs (FGF19, FGF21, and FGF23) modulate bone mineral synthesis and decomposition as well as muscle tissues. FGF2 and other FGFs also exert regulatory roles. A growing body of research has focused on understanding the implications of FGF signaling in orthopedic degeneration. Moreover, an increasing number of potential targets within the FGF signaling have been identified, such as FGF9, FGF18, and FGF23. However, it should be noted that most of these discoveries are still in the experimental stage, and further studies are needed before clinical application can be considered. Presently, this review aims to document the association between the FGF signaling pathway and the development and progression of orthopedic diseases. Besides, current therapeutic strategies targeting the FGF signaling pathway to prevent and treat orthopedic degeneration will be evaluated.
Collapse
Affiliation(s)
- Heng-Zhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing-Lve Zhang
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Dong-Liang Yuan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Wen-Qing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | | | - Ali Mobasheri
- Faculty of Medicine, Research Unit of Health Sciences and Technology, University of Oulu, 90014, Oulu, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406, Vilnius, Lithuania.
- Department of Rheumatology and Clinical Immunology, Universitair Medisch Centrum Utrecht, Utrecht, 3508, GA, the Netherlands.
- Department of Joint Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, B-4000, Liège, Belgium.
| | - Yu-Sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
15
|
Tong MJ, Song MX, Liu Z, Yu W, Wang CZ, Cai CD, Zhang YK, Zhang YQ, Wang LP, Zhu ZZ, Yin XF, Yan ZQ. A Bionic Thermosensitive Sustainable Delivery System for Reversing the Progression of Osteoarthritis by Remodeling the Joint Homeostasis. Adv Healthc Mater 2024; 13:e2303792. [PMID: 38394066 DOI: 10.1002/adhm.202303792] [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: 10/31/2023] [Revised: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Although the pathogenesis of osteoarthritis (OA) is unclear, inflammatory cytokines are related to its occurrence. However, few studies focused on the therapeutic strategies of regulating joint homeostasis by simultaneously remodeling the anti-inflammatory and immunomodulatory microenvironments. Fibroblast growth factor 18 (FGF18) is the only disease-modifying OA drug (DMOAD) with a potent ability and high efficiency in maintaining the phenotype of chondrocytes within cell culture models. However, its potential role in the immune microenvironment remains unknown. Besides, information on an optimal carrier, whose interface and chondral-biomimetic microenvironment mimic the native articular tissue, is still lacking, which substantially limits the clinical efficacy of FGF18. Herein, to simulate the cartilage matrix, chondroitin sulfate (ChS)-based nanoparticles (NPs) are integrated into poly(D, L-lactide)-poly(ethylene glycol)-poly(D, L-lactide) (PLEL) hydrogels to develop a bionic thermosensitive sustainable delivery system. Electrostatically self-assembled ChS and ε-poly-l-lysine (EPL) NPs are prepared for the bioencapsulation of FGF18. This bionic delivery system suppressed the inflammatory response in interleukin-1β (IL-1β)-mediated chondrocytes, promoted macrophage M2 polarization, and inhibited M1 polarization, thereby ameliorating cartilage degeneration and synovitis in OA. Thus, the ChS-based hydrogel system offers a potential strategy to regulate the chondrocyte-macrophage crosstalk, thus re-establishing the anti-inflammatory and immunomodulatory microenvironment for OA therapy.
Collapse
Affiliation(s)
- Min-Ji Tong
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Meng-Xiong Song
- Department of Orthopedic Surgery, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Zhe Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Wei Yu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chen-Zhong Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chuan-Dong Cai
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ying-Kai Zhang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yue-Qi Zhang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Li-Peng Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhen-Zhong Zhu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiao-Fan Yin
- Department of Orthopedic Surgery, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Zuo-Qin Yan
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| |
Collapse
|
16
|
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
|
17
|
Xiao-Feng L, Jin-Shan Z, Yong-Qiang Z, Ze-Feng W, Yong-Quan X, Yang-Zhen F, Zhen-Yu L, Liang L, Hong-Peng Z, Xiao-Peng H. Early cartilage lesion and 5-year incident joint surgery in knee osteoarthritis patients: a retrospective cohort study. BMC Musculoskelet Disord 2024; 25:398. [PMID: 38773475 PMCID: PMC11106971 DOI: 10.1186/s12891-024-07225-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/23/2024] [Indexed: 05/23/2024] Open
Abstract
OBJECTIVE to investigate the association between cartilage lesion-related features observed in knee osteoarthritis (OA) patients' first MRI examination and incident knee surgery within 5 years. Additionally, to assess the predictive value of these features for the incident knee surgery. METHODS We identified patients diagnosed with knee OA and treated at our institution between January 2015 and January 2018, and retrieved their baseline clinical data and first MRI examination films from the information system. Next, we proceeded to determine joint space narrowing grade, cartilage lesion size grade, cartilage full-thickness loss grade and cartilage lesion sum score for the medial and lateral compartments, respectively. Generalized linear regression models examined the association of these features with 5-year incident knee surgery. Positive and negative predictive values (PPVs and NPVs) were determined referring to 5-year incident knee surgery. RESULTS Totally, 878 participants (knees) were found eligible to form the study population. Within the 5 years, surgery was performed on 61 knees. None of the cartilage-related features had been found significantly associated with incident surgery. The results were similar for medial and lateral compartments. The PPVs were low for all the features. CONCLUSIONS Among symptomatic clinically diagnosed OA knees, cartilage lesions observed in the first MRI examinations were not found to be associated with the occurrence of joint surgery within a 5-year period. All these cartilage-related features appear to have no additional value in predicting 5-year incident joint surgery.
Collapse
Affiliation(s)
- Liu Xiao-Feng
- Department of Orthopedics, Jinjiang Municipal Hospital, Fujian, China
- Clinical Research Center for Orthopaedic Trauma and Reconstruction of Fujian Province, Jinjiang Municipal Hospital, Fujian, China
| | - Zhang Jin-Shan
- Department of Orthopedics, Jinjiang Municipal Hospital, Fujian, China.
- Clinical Research Center for Orthopaedic Trauma and Reconstruction of Fujian Province, Jinjiang Municipal Hospital, Fujian, China.
| | - Zheng Yong-Qiang
- Department of Orthopedics, Jinjiang Municipal Hospital, Fujian, China
- Clinical Research Center for Orthopaedic Trauma and Reconstruction of Fujian Province, Jinjiang Municipal Hospital, Fujian, China
| | - Wang Ze-Feng
- Department of Orthopedics, Jinjiang Municipal Hospital, Fujian, China
- Clinical Research Center for Orthopaedic Trauma and Reconstruction of Fujian Province, Jinjiang Municipal Hospital, Fujian, China
| | - Xu Yong-Quan
- Department of Orthopedics, Jinjiang Municipal Hospital, Fujian, China
- Clinical Research Center for Orthopaedic Trauma and Reconstruction of Fujian Province, Jinjiang Municipal Hospital, Fujian, China
| | - Fang Yang-Zhen
- Department of Orthopedics, Jinjiang Municipal Hospital, Fujian, China
- Clinical Research Center for Orthopaedic Trauma and Reconstruction of Fujian Province, Jinjiang Municipal Hospital, Fujian, China
| | - Lin Zhen-Yu
- Department of Orthopedics, Jinjiang Municipal Hospital, Fujian, China
- Clinical Research Center for Orthopaedic Trauma and Reconstruction of Fujian Province, Jinjiang Municipal Hospital, Fujian, China
| | - Lin Liang
- Department of Orthopedics, Jinjiang Municipal Hospital, Fujian, China
- Clinical Research Center for Orthopaedic Trauma and Reconstruction of Fujian Province, Jinjiang Municipal Hospital, Fujian, China
| | - Zhang Hong-Peng
- Department of Orthopedics, Jinjiang Municipal Hospital, Fujian, China
- Clinical Research Center for Orthopaedic Trauma and Reconstruction of Fujian Province, Jinjiang Municipal Hospital, Fujian, China
| | - Huang Xiao-Peng
- Department of Orthopedics, Jinjiang Municipal Hospital, Fujian, China
- Clinical Research Center for Orthopaedic Trauma and Reconstruction of Fujian Province, Jinjiang Municipal Hospital, Fujian, China
| |
Collapse
|
18
|
Grol MW. The evolving landscape of gene therapy strategies for the treatment of osteoarthritis. Osteoarthritis Cartilage 2024; 32:372-384. [PMID: 38199296 DOI: 10.1016/j.joca.2023.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/05/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
OBJECTIVES Significant advances have been made in our understanding of osteoarthritis (OA) pathogenesis; however, no disease-modifying therapies have been identified. This review will summarize the gene therapy landscape, its initial successes for OA, and possible challenges using recent studies and examples of gene therapies in clinical trials. DESIGN This narrative review has three major sections: 1) vector systems for OA gene therapy, 2) current and emerging targets for OA gene therapy, and 3) considerations and future directions. RESULTS Gene therapy is the strategy by which nucleic acids are delivered to treat and reverse disease progression. Specificity and prolonged expression of these nucleic acids are achieved by manipulating promoters, genes, and vector systems. Certain vector systems also allow for the development of combinatorial nucleic acid strategies that can be delivered in a single intraarticular injection - an approach likely required to treat the complexity of OA pathogenesis. Several viral and non-viral vector-based gene therapies are in clinical trials for OA, and many more are being evaluated in the preclinical arena. CONCLUSIONS In a post-coronavirus disease 2019 (COVID-19) era, the future of gene therapy for OA is certainly promising; however, the majority of preclinical validation continues to focus heavily on post-traumatic models and changes in only cartilage and subchondral bone. To ensure successful translation, new candidates in the preclinical arena should be examined against all joint tissues as well as pain using diverse models of injury-, obesity-, and age-induced disease. Lastly, consideration must be given to strategies for repeat administration and the cost of treatment owing to the chronic nature of OA.
Collapse
Affiliation(s)
- Matthew W Grol
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
19
|
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
|
20
|
Chen M, Lu Y, Liu Y, Liu Q, Deng S, Liu Y, Cui X, Liang J, Zhang X, Fan Y, Wang Q. Injectable Microgels with Hybrid Exosomes of Chondrocyte-Targeted FGF18 Gene-Editing and Self-Renewable Lubrication for Osteoarthritis Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312559. [PMID: 38266145 DOI: 10.1002/adma.202312559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/14/2024] [Indexed: 01/26/2024]
Abstract
Abnormal silencing of fibroblast growth factor (FGF) signaling significantly contributes to joint dysplasia and osteoarthritis (OA); However, the clinical translation of FGF18-based protein drugs is hindered by their short half-life, low delivery efficiency and the need for repeated articular injections. This study proposes a CRISPR/Cas9-based approach to effectively activate the FGF18 gene of OA chondrocytes at the genome level in vivo, using chondrocyte-affinity peptide (CAP) incorporated hybrid exosomes (CAP/FGF18-hyEXO) loaded with an FGF18-targeted gene-editing tool. Furthermore, CAP/FGF18-hyEXO are encapsulated in methacrylic anhydride-modified hyaluronic (HAMA) hydrogel microspheres via microfluidics and photopolymerization to create an injectable microgel system (CAP/FGF18-hyEXO@HMs) with self-renewable hydration layers to provide persistent lubrication in response to frictional wear. Together, the injectable CAP/FGF18-hyEXO@HMs, combined with in vivo FGF18 gene editing and continuous lubrication, have demonstrated their capacity to synergistically promote cartilage regeneration, decrease inflammation, and prevent ECM degradation both in vitro and in vivo, holding great potential for clinical translation.
Collapse
Affiliation(s)
- Manyu Chen
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Yan Lu
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Yuhan Liu
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, P. R. China
| | - Quanying Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Siyan Deng
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Yuan Liu
- Orthopedics Research Institute, Department of Orthopedics, West China Hospital Sichuan University, Chengdu, 610041, P. R. China
| | - Xiaolin Cui
- School of medicine the Chinese University of Hong Kong, Shenzhen, 518172, P. R. China
- Department of Orthopedic Surgery & Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine University of Otago, Christchurch, 8140, New Zealand
| | - Jie Liang
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- Sichuan Testing Center for Biomaterials and Medical Devices Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| |
Collapse
|
21
|
Roemer FW, Jarraya M, Hayashi D, Crema MD, Haugen IK, Hunter DJ, Guermazi A. A perspective on the evolution of semi-quantitative MRI assessment of osteoarthritis: Past, present and future. Osteoarthritis Cartilage 2024; 32:460-472. [PMID: 38211810 DOI: 10.1016/j.joca.2024.01.001] [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: 09/21/2023] [Revised: 12/15/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
OBJECTIVE This perspective describes the evolution of semi-quantitative (SQ) magnetic resonance imaging (MRI) in characterizing structural tissue pathologies in osteoarthritis (OA) imaging research over the last 30 years. METHODS Authors selected representative articles from a PubMed search to illustrate key steps in SQ MRI development, validation, and application. Topics include main scoring systems, reading techniques, responsiveness, reliability, technical considerations, and potential impact of artificial intelligence (AI). RESULTS Based on original research published between 1993 and 2023, this article introduces available scoring systems, including but not limited to Whole-Organ Magnetic Resonance Imaging Score (WORMS) as the first system for whole-organ assessment of the knee and the now commonly used MRI Osteoarthritis Knee Score (MOAKS) instrument. Specific systems for distinct OA subtypes or applications have been developed as well as MRI scoring instruments for other joints such as the hip, the fingers or thumb base. SQ assessment has proven to be valid, reliable, and responsive, aiding OA investigators in understanding the natural history of the disease and helping to detect response to treatment. AI may aid phenotypic characterization in the future. SQ MRI assessment's role is increasing in eligibility and safety evaluation in knee OA clinical trials. CONCLUSIONS Evidence supports the validity, reliability, and responsiveness of SQ MRI assessment in understanding structural aspects of disease onset and progression. SQ scoring has helped explain associations between structural tissue damage and clinical manifestations, as well as disease progression. While AI may support human readers to more efficiently perform SQ assessment in the future, its current application in clinical trials still requires validation and regulatory approval.
Collapse
Affiliation(s)
- Frank W Roemer
- Universitätsklinikum Erlangen & Friedrich Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA.
| | - Mohamed Jarraya
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daichi Hayashi
- Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA; Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Michel D Crema
- Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA; Institute of Sports Imaging, French National Institute of Sports (INSEP), Paris, France
| | - Ida K Haugen
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - David J Hunter
- Department of Rheumatology, Royal North Shore Hospital and Sydney Musculoskeletal Health, Kolling Institute, University of Sydney, St. Leonards, NSW, Australia
| | - Ali Guermazi
- Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA; Boston VA Healthcare System, West Roxbury, MA, USA
| |
Collapse
|
22
|
Winthrop KL, Mease P, Kerschbaumer A, Voll RE, Breedveld FC, Smolen JS, Gottenberg JE, Baraliakos X, Kiener HP, Aletaha D, Isaacs JD, Buch MH, Crow MK, Kay J, Crofford L, van Vollenhoven RF, Ospelt C, Siebert S, Kloppenburg M, McInnes IB, Huizinga TW, Gravallese EM. Unmet need in rheumatology: reports from the Advances in Targeted Therapies meeting, 2023. Ann Rheum Dis 2024; 83:409-416. [PMID: 38123338 DOI: 10.1136/ard-2023-224916] [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: 08/24/2023] [Accepted: 10/25/2023] [Indexed: 12/23/2023]
Abstract
The Advances in Targeted Therapies meets annually, convening experts in the field of rheumatology to both provide scientific updates and identify existing scientific gaps within the field. To review the major unmet scientific needs in rheumatology. The 23rd annual Advances in Targeted Therapies meeting convened with more than 100 international basic scientists and clinical researchers in rheumatology, immunology, infectious diseases, epidemiology, molecular biology and other specialties relating to all aspects of immune-mediated inflammatory diseases. We held breakout sessions in five rheumatological disease-specific groups including: rheumatoid arthritis (RA), psoriatic arthritis (PsA), axial spondyloarthritis (axSpa), systemic lupus erythematosus (SLE), systemic sclerosis (SSc) and vasculitis, and osteoarthritis (OA). In each group, experts were asked to identify and prioritise current unmet needs in clinical and translational research. An overarching theme across all disease states is the continued need for clinical trial design innovation with regard to therapeutics, endpoint and disease endotypes. Within RA, unmet needs comprise molecular classification of disease pathogenesis and activity, pre-/early RA strategies, more refined pain profiling and innovative trials designs to deliver on precision medicine. Continued scientific questions within PsA include evaluating the genetic, immunophenotypic, clinical signatures that predict development of PsA in patients with psoriasis, and the evaluation of combination therapies for difficult-to-treat disease. For axSpA, there continues to be the need to understand the role of interleukin-23 (IL-23) in pathogenesis and the genetic relationship of the IL-23-receptor polymorphism with other related systemic inflammatory diseases (eg, inflammatory bowel disease). A major unmet need in the OA field remains the need to develop the ability to reliably phenotype and stratify patients for inclusion in clinical trials. SLE experts identified a number of unmet needs within clinical trial design including the need for allowing endpoints that reflect pharmacodynamic/functional outcomes (eg, inhibition of type I interferon pathway activation; changes in urine biomarkers). Lastly, within SSc and vasculitis, there is a lack of biomarkers that predict response or disease progression, and that allow patients to be stratified for therapies. There remains a strong need to innovate clinical trial design, to identify systemic and tissue-level biomarkers that predict progression or response to therapy, endotype disease, and to continue developing therapies and therapeutic strategies for those with treatment-refractory disease. This document, based on expert consensus, should provide a roadmap for prioritising scientific endeavour in the field of rheumatology.
Collapse
Affiliation(s)
- Kevin L Winthrop
- Department of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Philip Mease
- Department of Rheumatology, University of Washington, Seattle, Washington, USA
- Department of Rheumatology, Medical University of Vienna, Wien, Austria
| | | | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, University of Freiburg, Freiburg im Breisgau, Germany
| | | | - Josef S Smolen
- Department of Rheumatology, Medical University of Vienna, Wien, Austria
| | | | | | - Hans P Kiener
- Department of Rheumatology, Medical University of Vienna, Wien, Austria
| | - Daniel Aletaha
- Department of Rheumatology, Medical University of Vienna, Wien, Austria
| | - John D Isaacs
- Newcastle University and Newcastle upon Tyne Hospitals NHS Foundation Trust, Institute of Cellular Medicine, Newcastle upon Tyne, UK
| | - Maya H Buch
- Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
- Department of Rheumatology, University of Manchester, Manchester, UK
| | - Mary K Crow
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, New York, NY, New York, USA
| | - Jonathan Kay
- Medicine, UMass Memorial Medical Center, Worcester, Massachusetts, USA
- Medicine, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Leslie Crofford
- Department of Rheumatology, Vanderbilt University, Nashville, Tennessee, USA
| | - Ronald F van Vollenhoven
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam, The Netherlands
| | - Caroline Ospelt
- Department of Rheumatology, Center of Experimental Rheumatology, Zurich, Switzerland
| | - Stefan Siebert
- Institute of Infection, Immunity & Inflammation, Glasgow University, Glasgow, UK
| | | | - Iain B McInnes
- MVLS College Office, University of Glasgow, Glasgow, UK
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Tom Wj Huizinga
- Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ellen M Gravallese
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Deng R, Zhao R, Zhang Z, Chen Y, Yang M, Lin Y, Ye J, Li N, Qin H, Yan X, Shi J, Yuan F, Song S, Xu Z, Song Y, Fu J, Xu B, Nie G, Yu JK. Chondrocyte membrane-coated nanoparticles promote drug retention and halt cartilage damage in rat and canine osteoarthritis. Sci Transl Med 2024; 16:eadh9751. [PMID: 38381849 DOI: 10.1126/scitranslmed.adh9751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 01/30/2024] [Indexed: 02/23/2024]
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by progressive degeneration of articular cartilage. A challenge in the development of disease-modifying drugs is effective delivery to chondrocytes. The unique structure of the joint promotes rapid clearance of drugs through synovial fluid, and the dense and avascular cartilage extracellular matrix (ECM) limits drug penetration. Here, we show that poly(lactide-co-glycolic acid) nanoparticles coated in chondrocyte membranes (CM-NPs) were preferentially taken up by rat chondrocytes ex vivo compared with uncoated nanoparticles. Internalization of the CM-NPs was mediated primarily by E-cadherin, clathrin-mediated endocytosis, and micropinocytosis. These CM-NPs adhered to the cartilage ECM in rat knee joints in vivo and penetrated deeply into the cartilage matrix with a residence time of more than 34 days. Simulated synovial fluid clearance studies showed that CM-NPs loaded with a Wnt pathway inhibitor, adavivint (CM-NPs-Ada), delayed the catabolic metabolism of rat and human chondrocytes and cartilage explants under inflammatory conditions. In a surgical model of rat OA, drug-loaded CM-NPs effectively restored gait, attenuated periarticular bone remodeling, and provided chondroprotection against cartilage degeneration. OA progression was also mitigated by CM-NPs-Ada in a canine model of anterior cruciate ligament transection. These results demonstrate the feasibility of using chondrocyte membrane-coated nanoparticles to improve the pharmacokinetics and efficacy of anti-OA drugs.
Collapse
Affiliation(s)
- Ronghui Deng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zining Zhang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yang Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Meng Yang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yixuan Lin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jing Ye
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Nan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hao Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xin Yan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Jian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Fuzhen Yuan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Shitang Song
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Zijie Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yifan Song
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Jiangnan Fu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Bingbing Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jia-Kuo Yu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
- Orthopedic Sports Medicine Center, Beijing Tsinghua Changgung Hospital, Affiliated Hospital of Tsinghua University, Beijing 102218, P. R. China
| |
Collapse
|
24
|
Ansari M, Darvishi A, Sabzevari A. A review of advanced hydrogels for cartilage tissue engineering. Front Bioeng Biotechnol 2024; 12:1340893. [PMID: 38390359 PMCID: PMC10881834 DOI: 10.3389/fbioe.2024.1340893] [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: 11/19/2023] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
With the increase in weight and age of the population, the consumption of tobacco, inappropriate foods, and the reduction of sports activities in recent years, bone and joint diseases such as osteoarthritis (OA) have become more common in the world. From the past until now, various treatment strategies (e.g., microfracture treatment, Autologous Chondrocyte Implantation (ACI), and Mosaicplasty) have been investigated and studied for the prevention and treatment of this disease. However, these methods face problems such as being invasive, not fully repairing the tissue, and damaging the surrounding tissues. Tissue engineering, including cartilage tissue engineering, is one of the minimally invasive, innovative, and effective methods for the treatment and regeneration of damaged cartilage, which has attracted the attention of scientists in the fields of medicine and biomaterials engineering in the past several years. Hydrogels of different types with diverse properties have become desirable candidates for engineering and treating cartilage tissue. They can cover most of the shortcomings of other treatment methods and cause the least secondary damage to the patient. Besides using hydrogels as an ideal strategy, new drug delivery and treatment methods, such as targeted drug delivery and treatment through mechanical signaling, have been studied as interesting strategies. In this study, we review and discuss various types of hydrogels, biomaterials used for hydrogel manufacturing, cartilage-targeting drug delivery, and mechanosignaling as modern strategies for cartilage treatment.
Collapse
Affiliation(s)
- Mojtaba Ansari
- Department of Biomedical Engineering, Meybod University, Meybod, Iran
| | - Ahmad Darvishi
- Department of Biomedical Engineering, Meybod University, Meybod, Iran
| | - Alireza Sabzevari
- Department of Biomedical Engineering, Meybod University, Meybod, Iran
| |
Collapse
|
25
|
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
|
26
|
Hannani MT, Thudium CS, Karsdal MA, Ladel C, Mobasheri A, Uebelhoer M, Larkin J, Bacardit J, Struglics A, Bay-Jensen AC. From biochemical markers to molecular endotypes of osteoarthritis: a review on validated biomarkers. Expert Rev Mol Diagn 2024; 24:23-38. [PMID: 38353446 DOI: 10.1080/14737159.2024.2315282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/02/2024] [Indexed: 02/22/2024]
Abstract
INTRODUCTION Osteoarthritis (OA) affects over 500 million people worldwide. OA patients are symptomatically treated, and current therapies exhibit marginal efficacy and frequently carry safety-risks associated with chronic use. No disease-modifying therapies have been approved to date leaving surgical joint replacement as a last resort. To enable effective patient care and successful drug development there is an urgent need to uncover the pathobiological drivers of OA and how these translate into disease endotypes. Endotypes provide a more precise and mechanistic definition of disease subgroups than observable phenotypes, and a panel of tissue- and pathology-specific biochemical markers may uncover treatable endotypes of OA. AREAS COVERED We have searched PubMed for full-text articles written in English to provide an in-depth narrative review of a panel of validated biochemical markers utilized for endotyping of OA and their association to key OA pathologies. EXPERT OPINION As utilized in IMI-APPROACH and validated in OAI-FNIH, a panel of biochemical markers may uncover disease subgroups and facilitate the enrichment of treatable molecular endotypes for recruitment in therapeutic clinical trials. Understanding the link between biochemical markers and patient-reported outcomes and treatable endotypes that may respond to given therapies will pave the way for new drug development in OA.
Collapse
Affiliation(s)
- Monica T Hannani
- ImmunoScience, Nordic Bioscience A/S, Herlev, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Ali 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, Liège, Belgium
| | | | - Jonathan Larkin
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- SynOA Therapeutics, Philadelphia, PA, USA
| | - Jaume Bacardit
- School of Computing, Newcastle University, Newcastle upon Tyne, UK
| | - André Struglics
- Department of Clinical Sciences, Orthopaedics, Lund University, Lund, Sweden
| | | |
Collapse
|
27
|
Yao Y, Zhong J, Zhang L, Khan S, Chen W. CartiMorph: A framework for automated knee articular cartilage morphometrics. Med Image Anal 2024; 91:103035. [PMID: 37992496 DOI: 10.1016/j.media.2023.103035] [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: 10/13/2022] [Revised: 08/25/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023]
Abstract
We introduce CartiMorph, a framework for automated knee articular cartilage morphometrics. It takes an image as input and generates quantitative metrics for cartilage subregions, including the percentage of full-thickness cartilage loss (FCL), mean thickness, surface area, and volume. CartiMorph leverages the power of deep learning models for hierarchical image feature representation. Deep learning models were trained and validated for tissue segmentation, template construction, and template-to-image registration. We established methods for surface-normal-based cartilage thickness mapping, FCL estimation, and rule-based cartilage parcellation. Our cartilage thickness map showed less error in thin and peripheral regions. We evaluated the effectiveness of the adopted segmentation model by comparing the quantitative metrics obtained from model segmentation and those from manual segmentation. The root-mean-squared deviation of the FCL measurements was less than 8%, and strong correlations were observed for the mean thickness (Pearson's correlation coefficient ρ∈[0.82,0.97]), surface area (ρ∈[0.82,0.98]) and volume (ρ∈[0.89,0.98]) measurements. We compared our FCL measurements with those from a previous study and found that our measurements deviated less from the ground truths. We observed superior performance of the proposed rule-based cartilage parcellation method compared with the atlas-based approach. CartiMorph has the potential to promote imaging biomarkers discovery for knee osteoarthritis.
Collapse
Affiliation(s)
- Yongcheng Yao
- CU Lab of AI in Radiology (CLAIR), Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong, China.
| | - Junru Zhong
- CU Lab of AI in Radiology (CLAIR), Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong, China
| | - Liping Zhang
- CU Lab of AI in Radiology (CLAIR), Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong, China
| | - Sheheryar Khan
- School of Professional Education and Executive Development, The Hong Kong Polytechnic University, Hong Kong, China
| | - Weitian Chen
- CU Lab of AI in Radiology (CLAIR), Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
28
|
Patnaik R, Riaz S, Sivani BM, Faisal S, Naidoo N, Rizzo M, Banerjee Y. Evaluating the potential of Vitamin D and curcumin to alleviate inflammation and mitigate the progression of osteoarthritis through their effects on human chondrocytes: A proof-of-concept investigation. PLoS One 2023; 18:e0290739. [PMID: 38157375 PMCID: PMC10756552 DOI: 10.1371/journal.pone.0290739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/14/2023] [Indexed: 01/03/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disorder primarily affecting the elderly, characterized by a prominent inflammatory component. The long-term side effects associated with current therapeutic approaches necessitate the development of safer and more efficacious alternatives. Nutraceuticals, such as Vitamin D and curcumin, present promising therapeutic potentials due to their safety, efficacy, and cost-effectiveness. In this study, we utilized a proinflammatory human chondrocyte model of OA to assess the anti-inflammatory properties of Vitamin D and curcumin, with a particular focus on the Protease-Activated Receptor-2 (PAR-2) mediated inflammatory pathway. Employing a robust siRNA approach, we effectively modulated the expression of PAR-2 to understand its role in the inflammatory process. Our results reveal that both Vitamin D and curcumin attenuate the expression of PAR-2, leading to a reduction in the downstream proinflammatory cytokines, such as Tumor Necrosis Factor-alpha (TNF-α), Interleukin 6 (IL-6), and Interleukin 8 (IL-8), implicated in the OA pathogenesis. Concurrently, these compounds suppressed the expression of Receptor Activator of Nuclear Factor kappa-Β Ligand (RANKL) and its receptor RANK, which are associated with PAR-2 mediated TNF-α stimulation. Additionally, Vitamin D and curcumin downregulated the expression of Interferon gamma (IFN-γ), known to elevate RANKL levels, underscoring their potential therapeutic implications in OA. This study, for the first time, provides evidence of the mitigating effect of Vitamin D and curcumin on PAR-2 mediated inflammation, employing an siRNA approach in OA. Thus, our findings pave the way for future research and the development of novel, safer, and more effective therapeutic strategies for managing OA.
Collapse
Affiliation(s)
- Rajashree Patnaik
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Sumbal Riaz
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Bala Mohan Sivani
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Shemima Faisal
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Nerissa Naidoo
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties (Promise), University of Palermo, Palermo, Italy
| | - Yajnavalka Banerjee
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
- Centre for Medical Education, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
29
|
Li M, Xia Z, Li X, lan L, Mo X, Xie L, Zhan Y, Li W. Difference in quantitative MRI measurements of cartilage between Wiberg type III patella and stable patella based on a 3.0-T synthetic MRI sequence. Eur J Radiol Open 2023; 11:100526. [PMID: 37953964 PMCID: PMC10632675 DOI: 10.1016/j.ejro.2023.100526] [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: 07/23/2023] [Revised: 09/17/2023] [Accepted: 09/30/2023] [Indexed: 11/14/2023] Open
Abstract
Purpose The purpose of this study was to investigate the difference between the quantitative MRI values of Wiberg type III and stable patellar cartilage, and to improve the accuracy of MRI quantification in early patellar cartilage damage. Methods The knee joints of 94 healthy volunteers were scanned by a GE Signa Pioneer 3.0-T synthetic MRI machine. According to the Wiberg classification, the patella was divided into types I-III. Types I-II made up the stable patella group, and type III made up the unstable patella group. Two radiologists independently measured patellar cartilage thickness and quantitative synthetic MRI values (T1, T2, PD) in both groups. Interobserver agreement for quantitative variables was assessed using the Bland-Altman method. A third radiologist assessed differences in measurements. Results The medial T2 and T1 value of Wiberg III patella did not show a normal distribution (all P > 0.05). Compared with the stable group, the Wiberg type III group had thinner cartilage of the medial surface of the patella (P < 0.05), lower cartilage T2 and PD values (P < 0.05), but a similar cartilage T1 value (P > 0.05). There was no significant difference in the cartilage thickness, T1, T2, or PD value of the lateral patella between the Wiberg type III and the stable group (P > 0.05). Conclusion There were certain differences in the cartilage thickness of the medial surface of the patella and the quantitative value of synthetic MRI in Wiberg type III patellas. Quantitative studies of patellar cartilage MRI measurements need to consider the influence of patellar morphology.
Collapse
Affiliation(s)
- Min Li
- The Second Affiliated Hospital of Guangxi Medical University, Department of Radiology, Nanning, Guangxi 530007, China
| | - Zhenyuan Xia
- The Second Affiliated Hospital of Guangxi Medical University, Department of Radiology, Nanning, Guangxi 530007, China
| | - Xiaohua Li
- The Second Affiliated Hospital of Guangxi Medical University, Department of Radiology, Nanning, Guangxi 530007, China
| | - Lan lan
- The Second Affiliated Hospital of Guangxi Medical University, Department of Radiology, Nanning, Guangxi 530007, China
| | - Xinxin Mo
- The Second Affiliated Hospital of Guangxi Medical University, Department of Radiology, Nanning, Guangxi 530007, China
| | - La Xie
- The Second Affiliated Hospital of Guangxi Medical University, Department of Radiology, Nanning, Guangxi 530007, China
| | - Yu Zhan
- The Second Affiliated Hospital of Guangxi Medical University, Department of Radiology, Nanning, Guangxi 530007, China
| | - Weixiong Li
- The Second Affiliated Hospital of Guangxi Medical University, Department of Radiology, Nanning, Guangxi 530007, China
| |
Collapse
|
30
|
Kong K, Jin M, Zhao C, Qiao H, Chen X, Li B, Rong K, Zhang P, Shan Y, Xu Z, Chang Y, Li H, Zhai Z. Mechanical overloading leads to chondrocyte degeneration and senescence via Zmpste24-mediated nuclear membrane instability. iScience 2023; 26:108119. [PMID: 37965144 PMCID: PMC10641493 DOI: 10.1016/j.isci.2023.108119] [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: 07/22/2023] [Revised: 08/28/2023] [Accepted: 09/29/2023] [Indexed: 11/16/2023] Open
Abstract
Patients with OA and varus knees are subject to abnormal mechanical environment and objective of this study was to investigate the molecular mechanisms underlying chondrocyte senescence caused by mechanical overloading and the role of Zmpste24-mediated nuclear membrane instability in varus knees. Finite element analysis showed that anteromedial region of tibial plateau experienced the most mechanical stress in an osteoarthritis patient with a varus knee. Immunohistochemistry exhibited lower Zmpste24 expression and higher expression of senescence marker p21 in the anteromedial region. Animal experiments and cell-stretch models also demonstrated an inverse relationship between Zmpste24 and mechanically induced senescence. Zmpste24 overexpression rescued cartilage degeneration and senescence in vitro by scavenging ROS. In conclusion, anteromedial tibial plateau is exposed to abnormal stress in varus knees, downregulation of Zmpste24, and nuclear membrane stability may explain increased senescence in this region. Zmpste24 and nuclear membrane stability are potential targets for treating osteoarthritis caused by abnormal alignment.
Collapse
Affiliation(s)
- Keyu Kong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Minghao Jin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chen Zhao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hua Qiao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuzhuo Chen
- Shanghai Key Laboratory of Stomatology, Department of Oral Surgery, College of Stomatology, Ninth People’s Hospital, Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baixing Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kewei Rong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Pu Zhang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu Shan
- Suzhou Ninth People’s Hospital, Department of Orthopedics, Suzhou Ninth Hospital affiliated to Soochow University, Suzhou, China
| | - Zhengquan Xu
- Suzhou Municipal Hospital, Department of Orthopedics, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Yongyun Chang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huiwu Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zanjing Zhai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
Roemer FW, Jarraya M, Collins JE, Kwoh CK, Hayashi D, Hunter DJ, Guermazi A. Structural phenotypes of knee osteoarthritis: potential clinical and research relevance. Skeletal Radiol 2023; 52:2021-2030. [PMID: 36161341 PMCID: PMC10509066 DOI: 10.1007/s00256-022-04191-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 02/02/2023]
Abstract
A joint contains many different tissues that can exhibit pathological changes, providing many potential targets for treatment. Researchers are increasingly suggesting that osteoarthritis (OA) comprises several phenotypes or subpopulations. Consequently, a treatment for OA that targets only one pathophysiologic abnormality is unlikely to be similarly efficacious in preventing or delaying the progression of all the different phenotypes of structural OA. Five structural phenotypes have been proposed, namely the inflammatory, meniscus-cartilage, subchondral bone, and atrophic and hypertrophic phenotypes. The inflammatory phenotype is characterized by marked synovitis and/or joint effusion, while the meniscus-cartilage phenotype exhibits severe meniscal and cartilage damage. Large bone marrow lesions characterize the subchondral bone phenotype. The hypertrophic and atrophic OA phenotype are defined based on the presence large osteophytes or absence of any osteophytes, respectively, in the presence of concomitant cartilage damage. Limitations of the concept of structural phenotyping are that they are not mutually exclusive and that more than one phenotype may be present. It must be acknowledged that a wide range of views exist on how best to operationalize the concept of structural OA phenotypes and that the concept of structural phenotypic characterization is still in its infancy. Structural phenotypic stratification, however, may result in more targeted trial populations with successful outcomes and practitioners need to be aware of the heterogeneity of the disease to personalize their treatment recommendations for an individual patient. Radiologists should be able to define a joint at risk for progression based on the predominant phenotype present at different disease stages.
Collapse
Affiliation(s)
- Frank W Roemer
- Quantitative Imaging Center, Department of Radiology, Boston University School of Medicine, 820 Harrison Avenue, FGH Building, 4th floor, Boston, MA, 02118, USA.
- Department of Radiology, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054, Erlangen, Germany.
| | - Mohamed Jarraya
- Department of Radiology, Massachusetts General Hospital, Harvard University, 55 Fruit St, Boston, MA, 02114, USA
| | - Jamie E Collins
- Orthopaedics and Arthritis Center of Outcomes Research, Brigham and Women's Hospital, Harvard Medical, School, 75 Francis Street, BTM Suite 5016, Boston, MA, 02115, USA
| | - C Kent Kwoh
- University of Arizona Arthritis Center, The University of Arizona College of Medicine, 1501 N. Campbell Avenue, Suite, Tucson, AZ, 8303, USA
| | - Daichi Hayashi
- Department of Radiology, Stony Brook University Renaissance School of Medicine, State University of New York, 101 Nicolls Rd, HSc Level 4, Room 120, Stony Brook, NY, 11794-8460, USA
| | - David J Hunter
- Department of Rheumatology, Royal North Shore Hospital and Institute of Bone and Joint Research, Kolling Institute, University of Sydney, Reserve Rd, St. Leonards, 2065, NSW, Australia
| | - Ali Guermazi
- Quantitative Imaging Center, Department of Radiology, Boston University School of Medicine, 820 Harrison Avenue, FGH Building, 4th floor, Boston, MA, 02118, USA
- Department of Radiology, VA Boston Healthcare System, 1400 VFW Parkway, Suite 1B105, West Roxbury, MA, 02132, USA
| |
Collapse
|
32
|
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
|
33
|
Ng JQ, Jafarov TH, Little CB, Wang T, Ali AM, Ma Y, Radford GA, Vrbanac L, Ichinose M, Whittle S, Hunter DJ, Lannagan TRM, Suzuki N, Goyne JM, Kobayashi H, Wang TC, Haynes DR, Menicanin D, Gronthos S, Worthley DL, Woods SL, Mukherjee S. Loss of Grem1-lineage chondrogenic progenitor cells causes osteoarthritis. Nat Commun 2023; 14:6909. [PMID: 37907525 PMCID: PMC10618187 DOI: 10.1038/s41467-023-42199-1] [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] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 10/03/2023] [Indexed: 11/02/2023] Open
Abstract
Osteoarthritis (OA) is characterised by an irreversible degeneration of articular cartilage. Here we show that the BMP-antagonist Gremlin 1 (Grem1) marks a bipotent chondrogenic and osteogenic progenitor cell population within the articular surface. Notably, these progenitors are depleted by injury-induced OA and increasing age. OA is also caused by ablation of Grem1 cells in mice. Transcriptomic and functional analysis in mice found that articular surface Grem1-lineage cells are dependent on Foxo1 and ablation of Foxo1 in Grem1-lineage cells caused OA. FGFR3 signalling was confirmed as a promising therapeutic pathway by administration of pathway activator, FGF18, resulting in Grem1-lineage chondrocyte progenitor cell proliferation, increased cartilage thickness and reduced OA. These findings suggest that OA, in part, is caused by mechanical, developmental or age-related attrition of Grem1 expressing articular cartilage progenitor cells. These cells, and the FGFR3 signalling pathway that sustains them, may be effective future targets for biological management of OA.
Collapse
Affiliation(s)
- Jia Q Ng
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Toghrul H Jafarov
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Christopher B Little
- Raymond Purves Bone & Joint Research Laboratories, Kolling Institute, University of Sydney Faculty of Medicine and Health, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Tongtong Wang
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Abdullah M Ali
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Yan Ma
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Georgette A Radford
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Laura Vrbanac
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Mari Ichinose
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Samuel Whittle
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Rheumatology Unit, The Queen Elizabeth Hospital, Woodville South, SA, Australia
| | - David J Hunter
- Northern Clinical School, University of Sydney, St. Leonards, Sydney, NSW, Australia
| | - Tamsin R M Lannagan
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Nobumi Suzuki
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Jarrad M Goyne
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Hiroki Kobayashi
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Timothy C Wang
- Department of Medicine and Irving Cancer Research Center, Columbia University, New York, NY, USA
| | - David R Haynes
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Danijela Menicanin
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Stan Gronthos
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Daniel L Worthley
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
- Colonoscopy Clinic, Brisbane, QLD, Australia.
| | - Susan L Woods
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| | | |
Collapse
|
34
|
Jan Vilim, Ghazalova T, Petulova E, Horackova A, Stepankova V, Chaloupkova R, Bednar D, Damborsky J, Prokop Z. Computer-assisted stabilization of fibroblast growth factor FGF-18. Comput Struct Biotechnol J 2023; 21:5144-5152. [PMID: 37920818 PMCID: PMC10618113 DOI: 10.1016/j.csbj.2023.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/04/2023] Open
Abstract
The fibroblast growth factors (FGF) family holds significant potential for addressing chronic diseases. Specifically, recombinant FGF18 shows promise in treating osteoarthritis by stimulating cartilage formation. However, recent phase 2 clinical trial results of sprifermin (recombinant FGF18) indicate insufficient efficacy. Leveraging our expertise in rational protein engineering, we conducted a study to enhance the stability of FGF18. As a result, we obtained a stabilized variant called FGF18-E4, which exhibited improved stability with 16 °C higher melting temperature, resistance to trypsin and a 2.5-fold increase in production yields. Moreover, the FGF18-E4 maintained mitogenic activity after 1-week incubation at 37 °C and 1-day at 50 °C. Additionally, the inserted mutations did not affect its binding to the fibroblast growth factor receptors, making FGF18-E4 a promising candidate for advancing FGF-based osteoarthritis treatment.
Collapse
Affiliation(s)
- Jan Vilim
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
- Enantis Ltd., INBIT, Kamenice 34, 625 00 Brno, Czech Republic
| | | | - Eliska Petulova
- Enantis Ltd., INBIT, Kamenice 34, 625 00 Brno, Czech Republic
| | - Aneta Horackova
- Enantis Ltd., INBIT, Kamenice 34, 625 00 Brno, Czech Republic
| | | | | | - David Bednar
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Jiri Damborsky
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Zbynek Prokop
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| |
Collapse
|
35
|
Gan L, Deng Z, Wei Y, Li H, Zhao L. Decreased expression of GEM in osteoarthritis cartilage regulates chondrogenic differentiation via Wnt/β-catenin signaling. J Orthop Surg Res 2023; 18:751. [PMID: 37794464 PMCID: PMC10548561 DOI: 10.1186/s13018-023-04236-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND GEM (GTP-binding protein overexpressed in skeletal muscle) is one of the atypical small GTPase subfamily members recently identified as a regulator of cell differentiation. Abnormal chondrogenesis coupled with an imbalance in the turnover of cartilaginous matrix formation is highly relevant to the onset and progression of osteoarthritis (OA). However, how GEM regulates chondrogenic differentiation remains unexplored. METHODS Cartilage tissues were obtained from OA patients and graded according to the ORASI and ICRS grading systems. The expression alteration of GEM was detected in the Grade 4 cartilage compared to Grade 0 and verified in OA mimic culture systems. Next, to investigate the specific function of GEM during these processes, we generated a Gem knockdown (Gem-Kd) system by transfecting siRNA targeting Gem into ATDC5 cells. Acan, Col2a1, Sox9, and Wnt target genes of Gem-Kd ATDC5 cells were detected during induction. The transcriptomic sequencing analysis was performed to investigate the mechanism of GEM regulation. Wnt signaling pathways were verified by real-time PCR and immunoblot analysis. Finally, a rescue model generated by treating Gem-KD ATDC5 cells with a Wnt signaling agonist was established to validate the mechanism identified by RNA sequencing analysis. RESULTS A decreased expression of GEM in OA patients' cartilage tissues and OA mimic chondrocytes was observed. While during chondrogenesis differentiation and cartilage matrix formation, the expression of GEM was increased. Gem silencing suppressed chondrogenic differentiation and the expressions of Acan, Col2a1, and Sox9. RNA sequencing analysis revealed that Wnt signaling was downregulated in Gem-Kd cells. Decreased expression of Wnt signaling associated genes and the total β-CATENIN in the nucleus and cytoplasm were observed. The exogenous Wnt activation exhibited reversed effect on Gem loss-of-function cells. CONCLUSION These findings collectively validated that GEM functions as a novel regulator mediating chondrogenic differentiation and cartilage matrix formation through Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Lu Gan
- Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhonghao Deng
- Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yiran Wei
- Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | | | - Liang Zhao
- Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
36
|
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
|
37
|
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
|
38
|
Gong J, Nhan J, St-Pierre JP, Gillies ER. Designing polymers for cartilage uptake: effects of architecture and molar mass. J Mater Chem B 2023; 11:8804-8816. [PMID: 37668597 DOI: 10.1039/d3tb01417g] [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] [Indexed: 09/06/2023]
Abstract
Osteoarthritis (OA) is a progressive disease, involving the progressive breakdown of cartilage, as well as changes to the synovium and bone. There are currently no disease-modifying treatments available clinically. An increasing understanding of the disease pathophysiology is leading to new potential therapeutics, but improved approaches are needed to deliver these drugs, particularly to cartilage tissue, which is avascular and contains a dense matrix of collagens and negatively charged aggrecan proteoglycans. Cationic delivery vehicles have been shown to effectively penetrate cartilage, but these studies have thus far largely focused on proteins or nanoparticles, and the effects of macromolecular architectures have not yet been explored. Described here is the synthesis of a small library of polycations composed of N-(2-hydroxypropyl)methacrylamide (HPMA) and N-(3-aminopropyl)methacrylamide (APMA) with linear, 4-arm, or 8-arm structures and varying degrees of polymerization (DP) by reversible addition fragmentation chain-transfer (RAFT) polymerization. Uptake and retention of the polycations in bovine articular cartilage was assessed. While all polycations penetrated cartilage, uptake and retention generally increased with DP before decreasing for the highest DP. In addition, uptake and retention were higher for the linear polycations compared to the 4-arm and 8-arm polycations. In general, the polycations were well tolerated by bovine chondrocytes, but the highest DP polycations imparted greater cytotoxicity. Overall, this study reveals that linear polymer architectures may be more favorable for binding to the cartilage matrix and that the DP can be tuned to maximize uptake while minimizing cytotoxicity.
Collapse
Affiliation(s)
- Jue Gong
- Department of Chemistry, The University of Western Ontario, 1151 Richmond St., London, Ontario, N6A 5B7, Canada.
| | - Jordan Nhan
- Department of Chemical and Biological Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada.
| | - Jean-Philippe St-Pierre
- Department of Chemical and Biological Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada.
| | - Elizabeth R Gillies
- Department of Chemistry, The University of Western Ontario, 1151 Richmond St., London, Ontario, N6A 5B7, Canada.
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, 1151 Richmond St., London, Ontario, N6A 5B9, Canada
| |
Collapse
|
39
|
Bay-Jensen AC, Attur M, Samuels J, Thudium CS, Abramson SB, Karsdal MA. Pathological tissue formation and degradation biomarkers correlate with patient reported pain outcomes: an explorative study. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100379. [PMID: 37342785 PMCID: PMC10277584 DOI: 10.1016/j.ocarto.2023.100379] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/30/2023] [Indexed: 06/23/2023] Open
Abstract
Background The lack of disease modifying drugs in Osteoarthritis (OA) may be attributed to the difficulty in robust response based on patient-reported outcomes (PROs) linked to drug mechanism of action. Joint tissue turnover biomarkers are associated with disease progression. A subset of patients has elevated serum levels of CRP metabolite (CRPM). This explorative study investigates the associations between PROs and joint tissue turnover markers in patients with high or low CRPM. Methods Serum of 146 knee OA patients of the New York Inflammation cohort and 21 healthy donors were assessed for biomarkers of collagen degradation (C1M, C2M, C3M, C4M), formation (PRO-C1, PRO-C2, PRO-C3, PRO-C4), and CRPM. Mean (SD) age was 62.5 (10.1); BMI, 26.6 (3.6); 62% women; and, 67.6% had symptomatic OA. WOMAC pain, stiffness, function, and total were recorded at baseline and at two-year follow-up. Associations were adjusted for race, sex, age, BMI, and NSAID. Results There was no difference in markers between donors and patients. C2M correlated with the WOMAC scores in all CRPM groups. Significant correlations were observed between PROs and PRO-C4, C1M, and C3M in the CRPMhigh group. The best predictive models for improvement were found for function and total with AUCs of 0.74 (p < 0.01) and 0.78 (p < 0.01). The best predictive models for worsening were found for function and total with AUCs of 0.84 (p < 0.01) and 0.80 (p < 0.05). Conclusion We hypothesize that collagen markers are prognostic tools for segregating patient populations in clinical trials.
Collapse
Affiliation(s)
| | - Mukundan Attur
- Division of Rheumatology, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY-10003, USA
| | - Jonathan Samuels
- Division of Rheumatology, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY-10003, USA
| | | | - Steven B. Abramson
- Division of Rheumatology, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY-10003, USA
| | | |
Collapse
|
40
|
Eckstein F, Maschek S, Culvenor A, Sharma L, Roemer F, Duda G, Wirth W. Which risk factors determine cartilage thickness and composition change in radiographically normal knees? - Data from the Osteoarthritis Initiative. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100365. [PMID: 37207279 PMCID: PMC10188628 DOI: 10.1016/j.ocarto.2023.100365] [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: 01/26/2023] [Accepted: 04/24/2023] [Indexed: 05/21/2023] Open
Abstract
Objective Therapy for osteoarthritis ideally aims at preserving structure before radiographic change occurs. This study tests: a) whether longitudinal deterioration in cartilage thickness and composition (transverse relaxation-time T2) are greater in radiographically normal knees "at risk" of incident osteoarthritis than in those without risk factors; and b) which risk factors may be associated with these deteriorations. Design 755 knees from the Osteoarthritis Initiative were studied; all were bilaterally Kellgren Lawrence grade [KLG] 0 initially, and had magnetic resonance images available at 12- and 48-month follow-up. 678 knees were "at risk", whereas 77 were not (i.e., non-exposed reference). Cartilage thickness and composition change was determined in 16 femorotibial subregions, with deep and superficial T2 being analyzed in a subset (n = 59/52). Subregion values were used to compute location-independent change scores. Results In KLG0 knees "at risk", the femorotibial cartilage thinning score (-634 ± 516 μm) over 3 years exceeded the thickening score by approximately 20%, and was 27% greater (p < 0.01; Cohen D -0.27) than the thinning score in "non-exposed" knees (-501 ± 319 μm). Superficial and deep cartilage T2 change, however, did not differ significantly between both groups (p ≥ 0.38). Age, sex, body mass index, knee trauma/surgery history, family history of joint replacement, presence of Heberden's nodes, repetitive knee bending were not significantly associated with cartilage thinning (r2<1%), with only knee pain reaching statistical significance. Conclusions Knees "at risk" of incident knee OA displayed greater cartilage thinning scores than those "non-exposed". Except for knee pain, the greater cartilage loss was not significantly associated with demographic or clinical risk factors.
Collapse
Affiliation(s)
- F. Eckstein
- Department of Imaging and Functional Musculoskeletal Research, Institute of Anatomy and Cell Biology & Ludwig Boltzmann Intitute of Arthritis & Rehabilitation (LBIAR), Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
- Chondrometrics GmbH, Ainring, Germany
- Corresponding author. Institute of Anatomy & Cell Biology, Paracelsus Medical University, Strubergasse 21, A-5020 Salzburg, Austria.
| | - S. Maschek
- Department of Imaging and Functional Musculoskeletal Research, Institute of Anatomy and Cell Biology & Ludwig Boltzmann Intitute of Arthritis & Rehabilitation (LBIAR), Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
- Chondrometrics GmbH, Ainring, Germany
| | - A. Culvenor
- Department of Imaging and Functional Musculoskeletal Research, Institute of Anatomy and Cell Biology & Ludwig Boltzmann Intitute of Arthritis & Rehabilitation (LBIAR), Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
- La Trobe Sport and Exercise Medicine Research Centre, School of Allied Health La Trobe University, Bundoora, Australia
| | - L. Sharma
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago IL, USA
| | - F.W. Roemer
- Department of Radiology, Friedrich-Alexander University Erlangen-Nürnberg & Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Radiology, Boston University School of Medicine, Boston, MA, USA
| | - G.N. Duda
- Julius Wolff Institute, Berlin-Brandenburg Institute of Health at Charité – Universitätsmedizin Berlin, Germany
| | - W. Wirth
- Department of Imaging and Functional Musculoskeletal Research, Institute of Anatomy and Cell Biology & Ludwig Boltzmann Intitute of Arthritis & Rehabilitation (LBIAR), Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
- Chondrometrics GmbH, Ainring, Germany
| |
Collapse
|
41
|
Vincent TL, Conaghan PG. Are pro-regenerative therapies the future of osteoarthritis disease modification? Osteoarthritis Cartilage 2023; 31:1152-1153. [PMID: 37196977 DOI: 10.1016/j.joca.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/02/2023] [Indexed: 05/19/2023]
Affiliation(s)
- Tonia L Vincent
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| | - Philip G Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, and NIHR Leeds Biomedical Research Centre, Leeds, UK.
| |
Collapse
|
42
|
Jansen MP, Roemer FW, Marijnissen AKCA, Kloppenburg M, Blanco FJ, Haugen IK, Berenbaum F, Lafeber FPJG, Welsing PMJ, Mastbergen SC, Wirth W. Exploring the differences between radiographic joint space width and MRI cartilage thickness changes using data from the IMI-APPROACH cohort. Skeletal Radiol 2023; 52:1339-1348. [PMID: 36607356 DOI: 10.1007/s00256-022-04259-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Longitudinal weight-bearing radiographic joint space width (JSW) and non-weight-bearing MRI-based cartilage thickness changes often show weak correlations. The current objective was to investigate these correlations, and to explore the influence of different factors that could contribute to longitudinal differences between the two methods. METHODS The current study included 178 participants with medial osteoarthritis (OA) out of the 297 knee OA participants enrolled in the IMI-APPROACH cohort. Changes over 2 years in medial JSW (ΔJSWmed), minimum JSW (ΔJSWmin), and medial femorotibial cartilage thickness (ΔMFTC) were assessed using linear regression, using measurements from radiographs and MRI acquired at baseline, 6 months, and 1 and 2 years. Pearson R correlations were calculated. The influence of cartilage quality (T2 mapping), meniscal extrusion (MOAKS scoring), potential pain-induced unloading (difference in knee-specific pain scores), and increased loading (BMI) on the correlations was analyzed by dividing participants in groups based on each factor separately, and comparing correlations (slope and strength) between groups using linear regression models. RESULT Correlations between ΔMFTC and ΔJSWmed and ΔJSWmin were statistically significant (p < 0.004) but weak (R < 0.35). Correlations were significantly different between groups based on cartilage quality and on meniscal extrusion: only patients with the lowest T2 values and with meniscal extrusion showed significant moderate correlations. Pain-induced unloading or BMI-induced loading did not influence correlations. CONCLUSIONS While the amount of loading does not seem to make a difference, weight-bearing radiographic JSW changes are a better reflection of non-weight-bearing MRI cartilage thickness changes in knees with higher quality cartilage and with meniscal extrusion.
Collapse
Affiliation(s)
- Mylène P Jansen
- Department of Rheumatology & Clinical Immunology, HP G02.228 Heidelberglaan 100 3584CX, Utrecht, The Netherlands.
| | - Frank W 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
| | - Anne Karien C A Marijnissen
- Department of Rheumatology & Clinical Immunology, HP G02.228 Heidelberglaan 100 3584CX, Utrecht, The Netherlands
| | - Margreet Kloppenburg
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
- Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Francisco J Blanco
- Departamento de Fisioterapia Y Medicina, Grupo de Investigación de Reumatología (GIR), INIBIC - Complejo Hospitalario Universitario de A Coruña, SERGAS. Centro de Investigación CICA, Universidad de A Coruña, A Coruña, Spain. Servicio de Reumatologia, INIBIC- Universidade de A Coruña, A Coruña, Spain
| | - Ida K Haugen
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Francis Berenbaum
- Department of Rheumatology, AP-HP Saint-Antoine Hospital, Paris, France
- INSERM, Sorbonne University, Paris, France
| | - Floris P J G Lafeber
- Department of Rheumatology & Clinical Immunology, HP G02.228 Heidelberglaan 100 3584CX, Utrecht, The Netherlands
| | - Paco M J Welsing
- Department of Rheumatology & Clinical Immunology, HP G02.228 Heidelberglaan 100 3584CX, Utrecht, The Netherlands
| | - Simon C Mastbergen
- Department of Rheumatology & Clinical Immunology, HP G02.228 Heidelberglaan 100 3584CX, Utrecht, The Netherlands
| | - Wolfgang Wirth
- Department of Imaging & Functional Musculoskeletal Research, Institute of Anatomy & Cell Biology, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
- Ludwig Boltzmann Inst. for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
- Chondrometrics GmbH, Freilassing, Germany
| |
Collapse
|
43
|
Englund M, Turkiewicz A. Pain in clinical trials for knee osteoarthritis: estimation of regression to the mean. THE LANCET. RHEUMATOLOGY 2023; 5:e309-e311. [PMID: 38251596 DOI: 10.1016/s2665-9913(23)00090-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/15/2023] [Accepted: 03/15/2023] [Indexed: 01/23/2024]
Affiliation(s)
- Martin Englund
- Clinical Epidemiology Unit, Orthopaedics, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, SE-221 00 Lund, Sweden.
| | - Aleksandra Turkiewicz
- Clinical Epidemiology Unit, Orthopaedics, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, SE-221 00 Lund, Sweden
| |
Collapse
|
44
|
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
|
45
|
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
|
46
|
Assi R, Quintiens J, Monteagudo S, Lories RJ. Innovation in Targeted Intra-articular Therapies for Osteoarthritis. Drugs 2023; 83:649-663. [PMID: 37067759 DOI: 10.1007/s40265-023-01863-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2023] [Indexed: 04/18/2023]
Abstract
Osteoarthritis is the most common chronic joint disease characterized by progressive damage to the joints, leading to pain and loss of function. There is currently no cure or disease-modifying therapy for osteoarthritis. Hence, the increasing disease prevalence linked with ageing and obesity represents a substantial socio-economic burden. Intra-articular therapy by injection of drugs into affected joints can optimize local drug bioavailability, while reducing risks of systemic toxicity, a concern in an ageing patient population. In this review, we investigate the current landscape of intra-articular drug therapies for osteoarthritis, including established approaches and those in clinical development. We performed a literature review using PubMed, complemented with a search for clinical trials using the ClinicalTrials.gov repository. Additionally, conference abstracts and presentations were identified and systematic snowballing was applied. Identified drugs were divided into several groups by main mechanism of action, and include drugs that reduce inflammation (anti-inflammatory), drugs aiming to prevent or reverse structural damage (structure modifying), drugs that aim to reduce the pain, and other drugs with a specific target. Most studies have been performed for osteoarthritis of the knee, a joint that is easily accessible for intra-articular treatments. Optimal therapy would provide symptomatic relief, while preventing further damage to the joint. The field of intra-articular drug therapies for osteoarthritis is rapidly evolving with clear challenges identified: definition of relevant outcome measures, optimization of clinical trial set-ups, and dealing with placebo responses. While many uncertainties persist, it appears that the innovation in drug development and improved clinical trial set-up may finally deliver successful therapies for this important disease.
Collapse
Affiliation(s)
- Reem Assi
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium
| | - Jolien Quintiens
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium
- Division of Rheumatology, University Hospitals Leuven, 3000, Leuven, Belgium
| | - Silvia Monteagudo
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium
| | - Rik J Lories
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium.
- Division of Rheumatology, University Hospitals Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
47
|
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
|
48
|
Householder NA, Raghuram A, Agyare K, Thipaphay S, Zumwalt M. A Review of Recent Innovations in Cartilage Regeneration Strategies for the Treatment of Primary Osteoarthritis of the Knee: Intra-articular Injections. Orthop J Sports Med 2023; 11:23259671231155950. [PMID: 37138944 PMCID: PMC10150434 DOI: 10.1177/23259671231155950] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/09/2022] [Indexed: 05/05/2023] Open
Abstract
Background The pathology of primary osteoarthritis (OA) begins with structural cartilage damage, which initiates a self-propagating inflammatory pathway that further exacerbates cartilage deterioration. Current standard of care for knee primary OA involves treating the inflammatory symptoms to manage pain, which includes intra-articular (IA) injections of cortisone, an anti-inflammatory steroid, followed by a series of joint-cushioning hyaluronic acid gel injections. However, these injections do not delay the progression of primary OA. More focus on the underlying cellular pathology of OA has prompted researchers to develop treatments targeting the biochemical mechanisms of cartilage degradation. Purpose Researchers have yet to develop a United States Food and Drug Administration (FDA)-approved injection that has been demonstrated to significantly regenerate damaged articular cartilage. This paper reviews the current research on experimental injections aimed at achieving cellular restoration of the hyaline cartilage tissue of the knee joint. Study Design Narrative review. Methods The authors conducted a narrative literature review examining studies on primary OA pathogenesis and a systematic review of non-FDA-approved IA injections for the treatment of primary OA of the knee, described as "disease-modifying osteoarthritis drugs" in phase 1, 2, and 3 clinical trials. Conclusion New treatment approaches for primary OA investigate the potential of genetic therapies to restore native cartilage. It is clear that the most promising IA injections that could improve treatment of primary OA are bioengineered advanced-delivery steroid-hydrogel preparations, ex vivo expanded allogeneic stem cell injections, genetically engineered chondrocyte injections, recombinant fibroblast growth factor therapy, injections of selective proteinase inhibitors, senolytic therapy via injections, injectable antioxidant therapies, injections of Wnt pathway inhibitors, injections of nuclear factor-kappa β inhibitors, injections of modified human angiopoietin-like-3, various potential viral vector-based genetic therapy approaches, and RNA genetic technology administered via injections.
Collapse
Affiliation(s)
| | - Akshay Raghuram
- School of Medicine, Texas Tech University
Health Sciences Center, Lubbock, Texas, USA
| | - Kofi Agyare
- School of Medicine, Texas Tech University
Health Sciences Center, Lubbock, Texas, USA
| | - Skyler Thipaphay
- School of Medicine, Texas Tech University
Health Sciences Center, Lubbock, Texas, USA
| | - Mimi Zumwalt
- School of Medicine, Texas Tech University
Health Sciences Center, Lubbock, Texas, USA
- Mimi Zumwalt, MD, Orthopaedics
Department, Texas Tech University Health Sciences Center, 3601 4th Street, Stop 9436,
Lubbock, TX 79430-9436, USA ()
| |
Collapse
|
49
|
Ng JQ, Jafarov TH, Little CB, Wang T, Ali A, Ma Y, Radford GA, Vrbanac L, Ichinose M, Whittle S, Hunter D, Lannagan TRM, Suzuki N, Goyne JM, Kobayashi H, Wang TC, Haynes D, Menicanin D, Gronthos S, Worthley DL, Woods SL, Mukherjee S. Loss of Grem1-articular cartilage progenitor cells causes osteoarthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.29.534651. [PMID: 37034712 PMCID: PMC10081168 DOI: 10.1101/2023.03.29.534651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Osteoarthritis (OA), which carries an enormous disease burden across the world, is characterised by irreversible degeneration of articular cartilage (AC), and subsequently bone. The cellular cause of OA is unknown. Here, using lineage tracing in mice, we show that the BMP-antagonist Gremlin 1 (Grem1) marks a novel chondrogenic progenitor (CP) cell population in the articular surface that generates joint cartilage and subchondral bone during development and adulthood. Notably, this CP population is depleted in injury-induced OA, and with age. OA is also induced by toxin-mediated ablation of Grem1 CP cells in young mice. Transcriptomic analysis and functional modelling in mice revealed articular surface Grem1-lineage cells are dependent on Foxo1; ablation of Foxo1 in Grem1-lineage cells led to early OA. This analysis identified FGFR3 signalling as a therapeutic target, and injection of its activator, FGF18, caused proliferation of Grem1-lineage CP cells, increased cartilage thickness, and reduced OA pathology. We propose that OA arises from the loss of CP cells at the articular surface secondary to an imbalance in progenitor cell homeostasis and present a new progenitor population as a locus for OA therapy.
Collapse
Affiliation(s)
- Jia Q. Ng
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
- These authors contributed equally
| | - Toghrul H. Jafarov
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- These authors contributed equally
| | - Christopher B. Little
- Raymond Purves Bone & Joint Research Laboratories, Kolling Institute, University of Sydney Faculty of Medicine and Health, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Tongtong Wang
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Abdullah Ali
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Yan Ma
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Georgette A Radford
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Laura Vrbanac
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Mari Ichinose
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Samuel Whittle
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
- Rheumatology Unit, The Queen Elizabeth Hospital, Woodville South, SA, Australia
| | - David Hunter
- Northern Clinical School, University of Sydney, St. Leonards, Sydney, NSW, Australia
| | - Tamsin RM Lannagan
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Nobumi Suzuki
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Jarrad M. Goyne
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Hiroki Kobayashi
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Timothy C. Wang
- Department of Medicine and Irving Cancer Research Center, Columbia University, New York, NY USA
| | - David Haynes
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Danijela Menicanin
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
| | - Stan Gronthos
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
- School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Daniel L. Worthley
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Colonoscopy Clinic, Brisbane, Qld, Australia
- These authors contributed equally, corresponding authors
| | - Susan L. Woods
- Adelaide Medical School, Faculty of Health and Medical Sciences University of Adelaide, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- These authors contributed equally, corresponding authors
| | - Siddhartha Mukherjee
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- These authors contributed equally, corresponding authors
| |
Collapse
|
50
|
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
|