101
|
Osteoarthritis year in review 2020: imaging. Osteoarthritis Cartilage 2021; 29:170-179. [PMID: 33418028 DOI: 10.1016/j.joca.2020.12.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/23/2020] [Accepted: 12/17/2020] [Indexed: 02/02/2023]
Abstract
This narrative "Year in Review" highlights a selection of articles published between January 2019 and April 2020, to be presented at the OARSI World Congress 2020 within the field of osteoarthritis (OA) imaging. Articles were obtained from a PubMed search covering the above period, utilizing a variety of relevant search terms. We then selected original and review studies on OA-related imaging in humans, particularly those with direct clinical relevance, with a focus on the knee. Topics selected encompassed clinically relevant models of early OA, particularly imaging applications on cruciate ligament rupture, as these are of direct clinical interest and provide potential opportunity to evaluate preventive therapy. Further, imaging applications on structural modification of articular tissues in patients with established OA, by non-pharmacological, pharmacological and surgical interventions are summarized. Finally, novel deep learning approaches to imaging are reviewed, as these facilitate implementation and scaling of quantitative imaging application in clinical trials and clinical practice. Methodological or observational studies outside these key focus areas were not included. Studies focused on biology, biomechanics, biomarkers, genetics and epigenetics, and clinical studies that did not contain an imaging component are covered in other articles within the OARSI "Year in Review" series. In conclusion, exciting progress has been made in clinically validating human models of early OA, and the field of automated articular tissue segmentation. Most importantly though, it has been shown that structure modification of articular cartilage is possible, and future research should focus on the translation of these structural findings to clinical benefit.
Collapse
|
102
|
Reed KSM, Ulici V, Kim C, Chubinskaya S, Loeser RF, Phanstiel DH. Transcriptional response of human articular chondrocytes treated with fibronectin fragments: an in vitro model of the osteoarthritis phenotype. Osteoarthritis Cartilage 2021; 29:235-247. [PMID: 33248223 PMCID: PMC7870543 DOI: 10.1016/j.joca.2020.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/19/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Fibronectin is a matrix protein that is fragmented during cartilage degradation in osteoarthritis (OA). Treatment of chondrocytes with fibronectin fragments (FN-f) has been used to model OA in vitro, but the system has not been fully characterized. This study sought to define the transcriptional response of chondrocytes to FN-f, and directly compare it to responses traditionally observed in OA. DESIGN Normal human femoral chondrocytes isolated from tissue donors were treated with either FN-f or PBS (control) for 3, 6, or 18 h. RNA-seq libraries were compared between time-matched FN-f and control samples in order to identify changes in gene expression over time. Differentially expressed genes were compared to a published OA gene set and used for pathway, transcription factor motif, and kinome analysis. RESULTS FN-f treatment resulted in 3,914 differentially expressed genes over the time course. Genes that are up- or downregulated in OA were significantly up- (P < 0.00001) or downregulated (P < 0.0004) in response to FN-f. Early response genes were involved in proinflammatory pathways, whereas many late response genes were involved in ferroptosis. The promoters of upregulated genes were enriched for NF-κB, AP-1, and IRF motifs. Highly upregulated kinases included CAMK1G, IRAK2, and the uncharacterized kinase DYRK3, while growth factor receptors TGFBR2 and FGFR2 were downregulated. CONCLUSIONS FN-f treatment of normal human articular chondrocytes recapitulated many key aspects of the OA chondrocyte phenotype. This in vitro model is promising for future OA studies, especially considering its compatibility with genomics and genome-editing techniques.
Collapse
Affiliation(s)
- K S M Reed
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - V Ulici
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA; Division of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| | - C Kim
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA; Division of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| | - S Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA.
| | - R F Loeser
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA; Division of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| | - D H Phanstiel
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, 27599, USA; Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
103
|
Neuroscience and Neuroimmunology Solutions for Osteoarthritis Pain: Biological Drugs, Growth Factors, Peptides and Monoclonal Antibodies Targeting Peripheral Nerves. NEUROSCI 2021. [DOI: 10.3390/neurosci2010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Neuroscience is a vast discipline that deals with the anatomy, biochemistry, molecular biology, physiology and pathophysiology of central and peripheral nerves. Advances made through basic, translational, and clinical research in the field of neuroscience have great potential for long-lasting and beneficial impacts on human and animal health. The emerging field of biological therapy is intersecting with the disciplines of neuroscience, orthopaedics and rheumatology, creating new horizons for interdisciplinary and applied research. Biological drugs, growth factors, therapeutic peptides and monoclonal antibodies are being developed and tested for the treatment of painful arthritic and rheumatic diseases. This concise communication focuses on the solutions provided by the fields of neuroscience and neuroimmunology for real-world clinical problems in the field of orthopaedics and rheumatology, focusing on synovial joint pain and the emerging biological treatments that specifically target pathways implicated in osteoarthritis pain in peripheral nerves.
Collapse
|
104
|
Peat G, Thomas MJ. Osteoarthritis year in review 2020: epidemiology & therapy. Osteoarthritis Cartilage 2021; 29:180-189. [PMID: 33242603 DOI: 10.1016/j.joca.2020.10.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/29/2020] [Accepted: 10/29/2020] [Indexed: 02/02/2023]
Abstract
This personal choice of research themes and highlights from within the past year (1 May 2019 to 14 April 2020) spans descriptive, analytical-observational, and intervention studies. Descriptive estimates of the burden of osteoarthritis continue to underscore its position as a leading cause of disability worldwide, but whose burden is often felt greatest among disadvantaged and marginalised communities. Many of the major drivers of that burden are known but epidemiological studies continue the important work of elaborating on their timing, dose, specificity, and reversibility and placing them within an appropriate multi-level framework. A similar process of elaboration is seen also in studies (re-)estimating the relative benefits and risks of existing interventions, in some cases helping to identify low-value care, unwarranted variation, and initiating processes of deprescribing and decommissioning. Such research need not engender therapeutic nihilism. Our review closes by highlighting some emerging evidence on the efficacy and safety of novel therapeutic interventions and with a selective roll-call of methodological and meta-research in OA illustrating the continued commitment to improving research quality.
Collapse
Affiliation(s)
- G Peat
- Primary Care Centre Versus Arthritis, School of Medicine, Faculty of Medicine & Health Sciences, Keele University, Keele, UK.
| | - M J Thomas
- Primary Care Centre Versus Arthritis, School of Medicine, Faculty of Medicine & Health Sciences, Keele University, Keele, UK; Haywood Academic Rheumatology Centre, Midlands Partnership NHS Foundation Trust, Haywood Hospital, Staffordshire, UK
| |
Collapse
|
105
|
Wiegertjes R, van de Loo FAJ, Blaney Davidson EN. A roadmap to target interleukin-6 in osteoarthritis. Rheumatology (Oxford) 2021; 59:2681-2694. [PMID: 32691066 PMCID: PMC7516110 DOI: 10.1093/rheumatology/keaa248] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/26/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023] Open
Abstract
Joint inflammation is present in the majority of OA patients and pro-inflammatory mediators, such as IL-6, are actively involved in disease progression. Increased levels of IL-6 in serum or synovial fluid from OA patients correlate with disease incidence and severity, with IL-6 playing a pivotal role in the development of cartilage pathology, e.g. via induction of matrix-degrading enzymes. However, IL-6 also increases expression of anti-catabolic factors, suggesting a protective role. Until now, this dual role of IL-6 is incompletely understood and may be caused by differential effects of IL-6 classic vs trans-signalling. Here, we review current evidence regarding the role of IL-6 classic- and trans-signalling in local joint pathology of cartilage, synovium and bone. Furthermore, we discuss targeting of IL-6 in experimental OA models and provide future perspective for OA treatment by evaluating currently available IL-6 targeting strategies.
Collapse
Affiliation(s)
- Renske Wiegertjes
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fons A J van de Loo
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Esmeralda N Blaney Davidson
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
106
|
Logashina YA, Palikova YA, Palikov VA, Kazakov VA, Smolskaya SV, Dyachenko IA, Tarasova NV, Andreev YA. Anti-Inflammatory and Analgesic Effects of TRPV1 Polypeptide Modulator APHC3 in Models of Osteo- and Rheumatoid Arthritis. Mar Drugs 2021; 19:md19010039. [PMID: 33477357 PMCID: PMC7830295 DOI: 10.3390/md19010039] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/07/2021] [Accepted: 01/13/2021] [Indexed: 12/26/2022] Open
Abstract
Arthritis is a widespread inflammatory disease associated with progressive articular surface degradation, ongoing pain, and hyperalgesia causing the development of functional limitations and disability. TRPV1 channel is one of the high-potential targets for the treatment of inflammatory diseases. Polypeptide APHC3 from sea anemone Heteractis crispa is a mode-selective TRPV1 antagonist that causes mild hypothermia and shows significant anti-inflammatory and analgesic activity in different models of pain. We evaluated the anti-inflammatory properties of APHC3 in models of monosodium iodoacetate (MIA)-induced osteoarthritis and complete Freund’s adjuvant (CFA)-induced rheumatoid monoarthritis in comparison with commonly used non-steroidal anti-inflammatory drugs (NSAIDs) such as diclofenac, ibuprofen, and meloxicam. Subcutaneous administration of APHC3 (0.1 mg/kg) significantly reversed joint swelling, disability, grip strength impairment, and thermal and mechanical hypersensitivity. The effect of APHC3 was equal to or better than that of reference NSAIDs. Protracted treatment with APHC3 decreased IL-1b concentration in synovial fluid, reduced inflammatory changes in joints, and prevented the progression of cartilage degradation. Therefore, polypeptide APHC3 has the potential to be an analgesic and anti-inflammatory substance for the alleviation of arthritis symptoms.
Collapse
Affiliation(s)
- Yulia A. Logashina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia;
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya str. 8, bld. 2, 119991 Moscow, Russia; (S.V.S.); (N.V.T.)
| | - Yulia A. Palikova
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, 142290 Pushchino, Russia; (Y.A.P.); (V.A.P.); (V.A.K.); (I.A.D.)
| | - Viktor A. Palikov
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, 142290 Pushchino, Russia; (Y.A.P.); (V.A.P.); (V.A.K.); (I.A.D.)
| | - Vitaly A. Kazakov
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, 142290 Pushchino, Russia; (Y.A.P.); (V.A.P.); (V.A.K.); (I.A.D.)
| | - Sviatlana V. Smolskaya
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya str. 8, bld. 2, 119991 Moscow, Russia; (S.V.S.); (N.V.T.)
| | - Igor A. Dyachenko
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, 142290 Pushchino, Russia; (Y.A.P.); (V.A.P.); (V.A.K.); (I.A.D.)
| | - Nadezhda V. Tarasova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya str. 8, bld. 2, 119991 Moscow, Russia; (S.V.S.); (N.V.T.)
| | - Yaroslav A. Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia;
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya str. 8, bld. 2, 119991 Moscow, Russia; (S.V.S.); (N.V.T.)
- Correspondence:
| |
Collapse
|
107
|
Coryell PR, Diekman BO, Loeser RF. Mechanisms and therapeutic implications of cellular senescence in osteoarthritis. Nat Rev Rheumatol 2021; 17:47-57. [PMID: 33208917 PMCID: PMC8035495 DOI: 10.1038/s41584-020-00533-7] [Citation(s) in RCA: 324] [Impact Index Per Article: 108.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 12/13/2022]
Abstract
The development of osteoarthritis (OA) correlates with a rise in the number of senescent cells in joint tissues, and the senescence-associated secretory phenotype (SASP) has been implicated in cartilage degradation and OA. Age-related mitochondrial dysfunction and associated oxidative stress might induce senescence in joint tissue cells. However, senescence is not the only driver of OA, and the mechanisms by which senescent cells contribute to disease progression are not fully understood. Furthermore, it remains uncertain which joint cells and SASP-factors contribute to the OA phenotype. Research in the field has looked at developing therapeutics (namely senolytics and senomorphics) that eliminate or alter senescent cells to stop disease progression and pathogenesis. A better understanding of how senescence contributes to joint dysfunction may enhance the effectiveness of these approaches and provide relief for patients with OA.
Collapse
Affiliation(s)
- Philip R Coryell
- Division of Rheumatology, Allergy, and Immunology, Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Brian O Diekman
- Division of Rheumatology, Allergy, and Immunology, Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | - Richard F Loeser
- Division of Rheumatology, Allergy, and Immunology, Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
108
|
Au M, Liu Z, Rong L, Zheng Y, Wen C. Endothelin-1 induces chondrocyte senescence and cartilage damage via endothelin receptor type B in a post-traumatic osteoarthritis mouse model. Osteoarthritis Cartilage 2020; 28:1559-1571. [PMID: 32858189 DOI: 10.1016/j.joca.2020.08.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 08/08/2020] [Accepted: 08/14/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVES This study aimed to investigate the role of endothelin-1 (ET-1), originally known as the potent vasoconstrictor, and its receptors in chondrocyte senescence and osteoarthritis (OA) development. METHOD Temporal changes of ET-1 and its receptors with OA development were characterized in a posttraumatic OA (PTOA) mouse model at time zero, 1-month and 4-month after surgical induction via destabilization of medial meniscus (DMM). A transgenic ET-1 overexpression (TET-1) mouse model was deployed to assess the impact of upregulated ET-1 on chondrocyte senescence and cartilage degradation. Effects of endothelin receptor blockade on chondrocyte senescence and OA development were further examined both in vitro and in vivo. RESULTS Local expression of ET-1 in subchondral bone and synovium upregulated after DMM with an increase of plasma ET-1 level from 3.18 ± 0.21 pg/ml at time zero to 6.47 ± 0.34 pg/ml at 4-month post-surgery. Meanwhile, endothelin type B receptor (ETBR) (53.31 ± 2.42% to 83.8 ± 2.65%) and p16INK4a (10.91 ± 1.07% to 28.2 ± 1.0%) positve chondrocytes accumulated in articular cartilage since 1-month prior to cartilage loss at 4-month post-surgery. Overexpressed ET-1 promoted p16INK4a-positive senescent chondrocytes accumulation and cartilage degradation in TET-1 mice. Selective blockade of ETBR, but not ETAR, lowered the expression of p16INK4a in ET-1 or H2O2-induced chondrocyte senescence model, and mitigated the severity of murine PTOA. Intriguingly, reactive oxygen species (ROS) scavenger, Vitamin C, could rescue ET-1-induced chondrocyte senescence in vitro associated with restoration of mitochondrial dynamics. CONCLUSION ET-1 could induce chondrocytes senescence and cartilage damages via ETBR in PTOA.
Collapse
Affiliation(s)
- M Au
- Department of Biomedical Engineering, Faculty of Engineering, Hong Kong Polytechnic University, Kowloon, Hong Kong.
| | - Z Liu
- Department of Biomedical Engineering, Faculty of Engineering, Hong Kong Polytechnic University, Kowloon, Hong Kong; Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, China.
| | - L Rong
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, China.
| | - Y Zheng
- Department of Biomedical Engineering, Faculty of Engineering, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - C Wen
- Department of Biomedical Engineering, Faculty of Engineering, Hong Kong Polytechnic University, Kowloon, Hong Kong.
| |
Collapse
|
109
|
|
110
|
Is Lutikizumab, an Anti-Interleukin-1 α/ β Dual Variable Domain Immunoglobulin, efficacious for Osteoarthritis? Results from a bayesian network meta-analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9013283. [PMID: 33204726 PMCID: PMC7661137 DOI: 10.1155/2020/9013283] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/20/2020] [Accepted: 10/09/2020] [Indexed: 12/17/2022]
Abstract
Objective Most guidelines recommend the use of nonsteroidal anti-inflammatory drugs (NSAIDs), duloxetine, and tramadol for the nonoperative treatment of osteoarthritis (OA), but the use of them is limited by the tolerability and safety concerns. Lutikizumab is a novel anti–IL-1α/β dual variable domain immunoglobulin that can simultaneously bind and inhibit IL-1α and IL-1β to relieve the pain and dysfunction symptoms. We conducted this network meta-analysis to comprehensively compare the clinical efficacy and safety of lutikizumab with other drugs recommended by guidelines. Methods We conducted a Bayesian network and conventional meta-analyses to compare the efficacy and safety of lutikizumab with other traditional drugs. All eligible randomized clinical trials, in PubMed, CNKI, EMBASE, and Web of Science databases, from January 2000 to January 2020, were included. The Cochrane risk of the bias assessment tool was used for quality assessment. Pain relief, function improvement, and risk of adverse effects (AEs) were compared in this study. Results 24 articles with 11858 patients were included. Duloxetine (DUL) had the largest effect for pain relief (4.76, 95% CI [2.35 to 7.17]), and selective cox-2 inhibitors (SCI) were the most efficacious treatment for physical function improvement (SMD 3.94, 95% CI [2.48 to 5.40]). Lutikizumab showed no benefit compared with placebo for both pain relief (SMD 1.11, 95% CI [-2.29 to 4.52]) and function improvement (SMD 0.992, 95% CI [-0.433 to 4.25]). Lutikizumab and all other drugs are of favorable tolerance for patients in the treatment of OA compared with placebo. Conclusions Lutikizumab, the new anti–Interleukin-1α/β dual variable domain immunoglobulin, showed no improvement in pain or function when compared with placebo. Selective cox-2 inhibitors and duloxetine remain the most effective and safest treatment for OA. More high-quality trials are still needed to reconfirm the findings of this study.
Collapse
|
111
|
Abstract
The prevalence of osteoarthritis (OA) and the burden associated with the disease are steadily increasing worldwide, representing a major public health challenge for the coming decades. The lack of specific treatments for OA has led to it being recognized as a serious disease that has an unmet medical need. Advances in the understanding of OA pathophysiology have enabled the identification of a variety of potential therapeutic targets involved in the structural progression of OA, some of which are promising and under clinical investigation in randomized controlled trials. Emerging therapies include those targeting matrix-degrading proteases or senescent chondrocytes, promoting cartilage repair or limiting bone remodelling, local low-grade inflammation or Wnt signalling. In addition to these potentially disease-modifying OA drugs (DMOADs), several targets are being explored for the treatment of OA-related pain, such as nerve growth factor inhibitors. The results of these studies are expected to considerably reshape the landscape of OA management over the next few years. This Review describes the pathophysiological processes targeted by emerging therapies for OA, along with relevant clinical data and discussion of the main challenges for the further development of these therapies, to provide context for the latest advances in the field of pharmaceutical therapies for OA.
Collapse
|
112
|
Tryfonidou MA, de Vries G, Hennink WE, Creemers LB. "Old Drugs, New Tricks" - Local controlled drug release systems for treatment of degenerative joint disease. Adv Drug Deliv Rev 2020; 160:170-185. [PMID: 33122086 DOI: 10.1016/j.addr.2020.10.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022]
Abstract
Osteoarthritis (OA) and chronic low back pain (CLBP) caused by intervertebral disc (IVD) degeneration are joint diseases that have become major causes for loss of quality of life worldwide. Despite the unmet need, effective treatments other than invasive, and often ineffective, surgery are lacking. Systemic administration of drugs entails suboptimal local drug exposure in the articular joint and IVD. This review provides an overview of the potency of biomaterial-based drug delivery systems as novel treatment modality, with a focus on the biological effects of drug release systems that have reached translation at the level of in vivo models and relevant ex vivo models. These studies have shown encouraging results of biomaterial-based local delivery of several types of drugs, mostly inhibitors of inflammatory cytokines or other degenerative factors. Prevention of inflammation and degeneration and pain relief was achieved, although mainly in small animal models, with interventions applied at an early disease stage. Less convincing data were obtained with the delivery of regenerative factors. Multidisciplinary efforts towards tackling the discord between in vitro and in vivo release, combined with adaptations in the regulatory landscape may be needed to enhance safe and expeditious introduction of more and more effective controlled release-based treatments with the OA and CLBP patients.
Collapse
|
113
|
Schieker M, Conaghan PG, Mindeholm L, Praestgaard J, Solomon DH, Scotti C, Gram H, Thuren T, Roubenoff R, Ridker PM. Effects of Interleukin-1β Inhibition on Incident Hip and Knee Replacement : Exploratory Analyses From a Randomized, Double-Blind, Placebo-Controlled Trial. Ann Intern Med 2020; 173:509-515. [PMID: 32744862 PMCID: PMC8503784 DOI: 10.7326/m20-0527] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Osteoarthritis is a common inflammatory disorder with no disease-modifying therapies. Whether inhibition of interleukin-1β (IL-1β) can reduce the consequences of large joint osteoarthritis is unclear. OBJECTIVE To determine whether IL-1β inhibition with canakinumab reduces incident total hip or knee replacement (THR/TKR). DESIGN Exploratory analysis of a randomized trial. (ClinicalTrials.gov: NCT01327846). SETTING 1091 clinical sites in 39 countries. PARTICIPANTS 10 061 CANTOS (Canakinumab Anti-inflammatory Thrombosis Outcomes Study) participants. INTERVENTION Random allocation to placebo or canakinumab (50, 150, or 300 mg) subcutaneously once every 3 months. MEASUREMENTS The primary and secondary outcomes were time to first incident THR/TKR and time to first occurrence of an osteoarthritis-related adverse event (AE). Data were obtained through blinded ascertainment of trial clinical and safety databases. RESULTS Median follow-up was 3.7 years. For the individual canakinumab dose groups, compared with placebo, hazard ratios (HRs) for incident THR/TKR during follow-up were 0.60 (95% CI, 0.38 to 0.95) for the 50-mg group, 0.53 (CI, 0.33 to 0.84) for the 150-mg group, and 0.60 (CI, 0.38 to 0.93) for the 300-mg group. Thus, in the pooled canakinumab groups, compared with the placebo group, incidence rates for THR/TKR were 0.31 and 0.54 events per 100 person-years (HR, 0.58 [CI, 0.42 to 0.80]; P = 0.001), respectively. The HR for the secondary end point of osteoarthritis-related AEs was 0.73 (CI, 0.61 to 0.87). Similar findings were observed in analyses restricted to participants with a history of osteoarthritis. LIMITATION Because the parent trial was not designed to examine the efficacy of IL-1β inhibitors in osteoarthritis, information on structural joint outcomes was not collected. CONCLUSION Findings from this exploratory analysis of a randomized controlled trial support further investigation of IL-1β inhibition for treatment of large joint osteoarthritis. PRIMARY FUNDING SOURCE Novartis Pharmaceuticals.
Collapse
Affiliation(s)
- Matthias Schieker
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, and Basel, Switzerland, and Ludwig Maximilian University of Munich, Munich, Germany (M.S.)
| | - Philip G Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, and National Institute for Health Research Leeds BiomedicalResearch Centre, Leeds, United Kingdom (P.G.C.)
| | - Linda Mindeholm
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, and Basel, Switzerland (L.M., J.P., C.S., H.G., R.R.)
| | - Jens Praestgaard
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, and Basel, Switzerland (L.M., J.P., C.S., H.G., R.R.)
| | - Daniel H Solomon
- Brigham and Women's Hospital, Boston, Massachusetts (D.H.S., P.M.R.)
| | - Celeste Scotti
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, and Basel, Switzerland (L.M., J.P., C.S., H.G., R.R.)
| | - Herman Gram
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, and Basel, Switzerland (L.M., J.P., C.S., H.G., R.R.)
| | - Tom Thuren
- Novartis Pharma AG, Basel, Switzerland (T.T.)
| | - Ronenn Roubenoff
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, and Basel, Switzerland (L.M., J.P., C.S., H.G., R.R.)
| | - Paul M Ridker
- Brigham and Women's Hospital, Boston, Massachusetts (D.H.S., P.M.R.)
| |
Collapse
|
114
|
Affiliation(s)
- Nancy Lane
- UC Davis Health, Sacramento, California (N.L.)
| | - David Felson
- Boston University School of Medicine, Boston, Massachusetts (D.F.)
| |
Collapse
|
115
|
Vincent TL. Of mice and men: converging on a common molecular understanding of osteoarthritis. THE LANCET. RHEUMATOLOGY 2020; 2:e633-e645. [PMID: 32989436 PMCID: PMC7511206 DOI: 10.1016/s2665-9913(20)30279-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Despite an increasing burden of osteoarthritis in developed societies, target discovery has been slow and there are currently no approved disease-modifying osteoarthritis drugs. This lack of progress is due in part to a series of misconceptions over the years: that osteoarthritis is an inevitable consequence of ageing, that damaged articular cartilage cannot heal itself, and that osteoarthritis is driven by synovial inflammation similar to that seen in rheumatoid arthritis. Molecular interrogation of disease through ex-vivo tissue analysis, in-vitro studies, and preclinical models have radically reshaped the knowledge landscape. Inflammation in osteoarthritis appears to be distinct from that seen in rheumatoid arthritis. Recent randomised controlled trials, using treatments repurposed from rheumatoid arthritis, have largely been unsuccessful. Genome-wide studies point to defects in repair pathways, which accords well with recent promise using growth factor therapies or Wnt pathway antagonism. Nerve growth factor has emerged as a robust target in osteoarthritis pain in phase 2-3 trials. These studies, both positive and negative, align well with those in preclinical surgical models of osteoarthritis, indicating that pathogenic mechanisms identified in mice can lead researchers to valid human targets. Several novel candidate pathways are emerging from preclinical studies that offer hope of future translational impact. Enhancing trust between industry, basic, and clinical scientists will optimise our collective chance of success.
Collapse
Affiliation(s)
- Tonia L Vincent
- Centre for Osteoarthritis Pathogenesis, Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
116
|
Sandomenico A, Sivaccumar JP, Ruvo M. Evolution of Escherichia coli Expression System in Producing Antibody Recombinant Fragments. Int J Mol Sci 2020; 21:ijms21176324. [PMID: 32878291 PMCID: PMC7504322 DOI: 10.3390/ijms21176324] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/12/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
Antibodies and antibody-derived molecules are continuously developed as both therapeutic agents and key reagents for advanced diagnostic investigations. Their application in these fields has indeed greatly expanded the demand of these molecules and the need for their production in high yield and purity. While full-length antibodies require mammalian expression systems due to the occurrence of functionally and structurally important glycosylations, most antibody fragments and antibody-like molecules are non-glycosylated and can be more conveniently prepared in E. coli-based expression platforms. We propose here an updated survey of the most effective and appropriate methods of preparation of antibody fragments that exploit E. coli as an expression background and review the pros and cons of the different platforms available today. Around 250 references accompany and complete the review together with some lists of the most important new antibody-like molecules that are on the market or are being developed as new biotherapeutics or diagnostic agents.
Collapse
|
117
|
Future of Chondroprotectors in the Treatment of Degenerative Processes of Connective Tissue. Pharmaceuticals (Basel) 2020; 13:ph13090220. [PMID: 32872387 PMCID: PMC7557725 DOI: 10.3390/ph13090220] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/19/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis is one of the most common diseases of the connective tissue of the elderly. It was found that most epidemiological studies used the Kellgren and Lawrence system for classification of osteoarthritis, which indicates one of the 5 degrees (0-4) of osteoarthritis in various joints according to the radiographic atlas. It has been proven that chondroprotectors are represented by the following active substances: chondroitin sulfate, glucosamine sulfate or hydrochloride, hyaluronic acid, glycosaminoglycans, extraction preparations from animal or plant raw materials. The sources of raw materials for the manufacture of combined chondroprotectors are known, methods for their preparation and use are described. The main drugs on the chondroprotective market are presented. The effectiveness of their use for the treatment of osteoarthritis has been proven. It was found that preparations containing chondroitin sulfate have anti-inflammatory activity, affecting mainly the cellular component of inflammation, stimulate the synthesis of hyaluronic acid and proteoglycans. Methods of treating osteoarthritis using cell therapy (the use of readily available, highly proliferative, and multipotent mesenchymal stromal cells) are presented.
Collapse
|
118
|
Drug delivery in intervertebral disc degeneration and osteoarthritis: Selecting the optimal platform for the delivery of disease-modifying agents. J Control Release 2020; 328:985-999. [PMID: 32860929 DOI: 10.1016/j.jconrel.2020.08.041] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/16/2022]
Abstract
Osteoarthritis (OA) and intervertebral disc degeneration (IVDD) as major cause of chronic low back pain represent the most common degenerative joint pathologies and are leading causes of pain and disability in adults. Articular cartilage (AC) and intervertebral discs are cartilaginous tissues with a similar biochemical composition and pathophysiological aspects of degeneration. Although treatments directed at reversing these conditions are yet to be developed, many promising disease-modifying drug candidates are currently under investigation. Given the localized nature of these chronic diseases, drug delivery systems have the potential to enhance therapeutic outcomes by providing controlled and targeted release of bioactives, minimizing the number of injections needed and increasing drug concentration in the affected areas. This review provides a comprehensive overview of the currently most promising disease-modifying drugs as well as potential drug delivery systems for OA and IVDD therapy.
Collapse
|
119
|
Chadha S, Behl T, Bungau S, Kumar A, Arora R, Gupta A, Uddin MS, Zengin G, Aleya L, Setia D, Arora S. Mechanistic insights into the role of pyroptosis in rheumatoid arthritis. Curr Res Transl Med 2020; 68:151-158. [PMID: 32830085 DOI: 10.1016/j.retram.2020.07.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/09/2020] [Accepted: 07/28/2020] [Indexed: 12/30/2022]
Abstract
Cell death is ascribed as an essential biological process that is fundamental for the development of an organism along with its survival. The procedure comprises of apoptosis and pyroptosis. Pyroptosis is a programmed procedure for cell death which is inflammatory in nature and this pathway gets activated via human caspase-4, human caspase-11 and human caspase-5. The activation of this process leads to release of pro-inflammatory mediators including cytokines, alarmins, IL-18 and IL-1β. The pro-inflammatory mediators released via interaction of intracellular kinases direct the development of Rheumatoid arthritis. Rheumatoid arthritis is characterized as disorder/disease that is auto-immune and chronic in nature. It involves erosions in marginal bone along with articular cartilage which is responsible for joint destruction. The cytokine along with its complex network is responsible for inflammation. The process of pyroptosis is linked with the destruction of plasma membrane, that releases these mediators and excessive release of these mediators is linked with rheumatoid arthritis.
Collapse
Affiliation(s)
- Swati Chadha
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Rashmi Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Amit Gupta
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk Uniersity Campus, Konya, Turkey
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, France
| | - Dhruv Setia
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
120
|
Therapeutic Manipulation of Macrophages Using Nanotechnological Approaches for the Treatment of Osteoarthritis. NANOMATERIALS 2020; 10:nano10081562. [PMID: 32784839 PMCID: PMC7466380 DOI: 10.3390/nano10081562] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/31/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is the most common joint pathology causing severe pain and disability. Macrophages play a central role in the pathogenesis of OA. In the joint microenvironment, macrophages with an M1-like pro-inflammatory phenotype induce chronic inflammation and joint destruction, and they have been correlated with the development and progression of the disease, while the M2-like anti-inflammatory macrophages support the recovery of the disease, promoting tissue repair and the resolution of inflammation. Nowadays, the treatment of OA in the clinic relies on systemic and/or intra-articular administration of anti-inflammatory and pain relief drugs, as well as surgical interventions for the severe cases (i.e., meniscectomy). The disadvantages of the pharmacological therapy are related to the chronic nature of the disease, requiring prolonged treatments, and to the particular location of the pathology in joint tissues, which are separated anatomical compartments with difficult access for the drugs. To overcome these challenges, nanotechnological approaches have been investigated to improve the delivery of drugs toward macrophages into the diseased joint. This strategy may offer advantages by reducing off-target toxicities and improving long-term therapeutic efficacy. In this review, we describe the nanomaterial-based approaches designed so far to directly or indirectly manipulate macrophages for the treatment of osteoarthritis.
Collapse
|
121
|
Ragni E, Mangiavini L, Viganò M, Brini AT, Peretti GM, Banfi G, de Girolamo L. Management of Osteoarthritis During the COVID-19 Pandemic. Clin Pharmacol Ther 2020; 108:719-729. [PMID: 32438454 PMCID: PMC7280639 DOI: 10.1002/cpt.1910] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022]
Abstract
The pandemic spread of the new coronavirus disease 2019 (COVID‐19) infection in China first, and all over the world at present, has become a global health emergency due to the rapidly increasing number of affected patients. Currently, a clear relationship between COVID‐19 infection incidence and/or complications due to chronic or occasional treatments for other pathologies is still not clear, albeit the COVID‐19 pandemic may condition the treatment strategy of complex disorders, such as osteoarthritis (OA). Importantly, OA is the most common age‐related joint disease, affecting more than 80% of people older than the age of 55, an age burden also shared with the highest severity in COVID‐19 patients. OA patients often show a large array of concomitant pathologies, such as diabetes, inflammation, and cardiovascular diseases that are again shared with COVID‐19 patients and may therefore increase complications. Moreover, different OA treatments, such as NSAIDs, paracetamol, corticosteroids, opioids, or other molecules have a wide array of iatrogenic effects, potentially increasing COVID‐19 secondary infection incidence or complications. In this review we critically analyze the evidence on either negative or positive effects of drugs commonly used to manage OA in this particular scenario. This would provide orthopedic surgeons in particular, and physicians, pharmacologists, and clinicians in general, a comprehensive description about the safety of the current pharmacological approaches and a decision‐making tool to treat their OA patients as the coronavirus pandemic continues.
Collapse
Affiliation(s)
- Enrico Ragni
- IRCCS Istituto Ortopedico Galeazzi, Laboratorio di Biotecnologie Applicate all'Ortopedia, Milan, Italy
| | - Laura Mangiavini
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Marco Viganò
- IRCCS Istituto Ortopedico Galeazzi, Laboratorio di Biotecnologie Applicate all'Ortopedia, Milan, Italy
| | - Anna Teresa Brini
- IRCCS Istituto Ortopedico Galeazzi, Laboratorio di Applicazioni Biotecnologiche, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Giuseppe Michele Peretti
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Giuseppe Banfi
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Laura de Girolamo
- IRCCS Istituto Ortopedico Galeazzi, Laboratorio di Biotecnologie Applicate all'Ortopedia, Milan, Italy
| |
Collapse
|
122
|
The association of plasma IL-1Ra and related cytokines with radiographic severity of early knee osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2020; 2:100046. [DOI: 10.1016/j.ocarto.2020.100046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 02/13/2020] [Indexed: 12/28/2022] Open
|
123
|
Abstract
Osteoarthritis (OA) is one of the most debilitating diseases and is associated with a high personal and socioeconomic burden. So far, there is no therapy available that effectively arrests structural deterioration of cartilage and bone or is able to successfully reverse any of the existing structural defects. Efforts to identify more tailored treatment options led to the development of strategies that enabled the classification of patient subgroups from the pool of heterogeneous phenotypes that display distinct common characteristics. To this end, the classification differentiates the structural endotypes into cartilage and bone subtypes, which are predominantly driven by structure-related degenerative events. In addition, further classifications have highlighted individuals with an increased inflammatory contribution (inflammatory phenotype) and pain-driven phenotypes as well as senescence and metabolic syndrome phenotypes. Most probably, it will not be possible to classify individuals by a single definite subtype, but it might help to identify groups of patients with a predominant pathology that would more likely benefit from a specific drug or cell-based therapy. Current clinical trials addressed mainly regeneration/repair of cartilage and bone defects or targeted pro-inflammatory mediators by intra-articular injections of drugs and antibodies. Pain was treated mostly by antagonizing nerve growth factor (NGF) activity and its receptor tropomyosin-related kinase A (TrkA). Therapies targeting metabolic disorders such as diabetes mellitus and senescence/aging-related pathologies are not specifically addressing OA. However, none of these therapies has been proven to modify disease progression significantly or successfully prevent final joint replacement in the advanced disease stage. Within this review, we discuss the recent advances in phenotype-specific treatment options and evaluate their applicability for use in personalized OA therapy.
Collapse
Affiliation(s)
- Susanne Grässel
- Department of Orthopedic Surgery, Exp. Orthopedics, ZMB/Biopark 1, Am Biopark 9, University of Regensburg, Regensburg, 93053, Germany
| | - Dominique Muschter
- Department of Orthopedic Surgery, Exp. Orthopedics, ZMB/Biopark 1, Am Biopark 9, University of Regensburg, Regensburg, 93053, Germany
| |
Collapse
|
124
|
Aslam F, England BR, Cannella A, Sharp V, Kao L, Arnason J, Albayda J, Bakewell C, Sanghvi S, Fairchild R, Torralba KD, Evangelisto A, DeMarco PJ, Bethina N, Kissin EY. Ultrasound Doppler and tenosynovial fluid analysis in tenosynovitis. Ann Rheum Dis 2020; 79:908-913. [PMID: 32213497 DOI: 10.1136/annrheumdis-2020-216927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 03/09/2020] [Accepted: 03/13/2020] [Indexed: 01/26/2023]
Abstract
OBJECTIVE To assess Doppler ultrasound (US) and tenosynovial fluid (TSF) characteristics in tenosynovitis within common rheumatic conditions, as well as their diagnostic utility. METHODS Subjects with tenosynovitis underwent Doppler US and US-guided TSF aspiration for white cell count (WCC) and crystal analysis. Tenosynovial Doppler scores (DS) were semiquantitatively graded. TSF WCC and DS were compared using Kruskal-Wallis tests and logistic regression between non-inflammatory conditions (NIC), inflammatory conditions (IC) and crystal-related conditions (CRC). Receiver operating curves, sensitivity and specificity assessed the ability of WCC and DS to discriminate IC from NIC. RESULTS We analysed 100 subjects from 14 sites. The mean age was 62 years, 65% were female, and the mean TSF volume was 1.2 mL. Doppler signal was present in 93.7% of the IC group and was more frequent in IC than in NIC group (OR 6.82, 95% CI 1.41 to 32.97). The TSF median WCC per 109/L was significantly higher in the IC (2.58, p<0.001) and CRC (1.07, p<0.01) groups versus the NIC group (0.38). A TSF cut-off of ≥0.67 WCC per 109/L optimally discriminated IC versus NIC with a sensitivity and specificity each of 81.3%. In the IC group, 20 of 48 (41.7%) subjects had a TSF WCC <2.00 per 109/L. CONCLUSIONS A negative DS helps rule out IC in tenosynovitis, but a positive DS is non-specific and merits TSF testing. Unlike synovial fluid, a lower TSF WCC better discriminates IC from NIC. US guidance facilitates aspiration of minute TSF volume, which is critical for diagnosing tenosynovial CRC.
Collapse
Affiliation(s)
- Fawad Aslam
- Rheumatology, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Bryant R England
- Rheumatology, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Rheumatology, VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Amy Cannella
- Division of Rheumatology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Veronika Sharp
- Division of Rheumatology, Department of Medicine, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Lily Kao
- Division of Rheumatology, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Jon Arnason
- Division of Rheumatology, University of Wisconsin Hospital and Clinics, Madison, Wisconsin, USA
| | - Jemima Albayda
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Catherine Bakewell
- Division of Rheumatology, Intermountain Health Care, Salt Lake City, Utah, USA
| | - Shruti Sanghvi
- Division of Rheumatology, Intermountain Health Care, Salt Lake City, Utah, USA
| | - Robert Fairchild
- Division of Rheumatology, Stanford University, Stanford, California, USA
| | - Karina D Torralba
- Division of Rheumatology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Amy Evangelisto
- Division of Rheumatology, Arthritis, Rheumatic & Back Disease Associates, Voorhees, New Jersey, USA
| | - Paul J DeMarco
- Center for Rheumatology and Bone Research, Arthritis and Rheumatism Associates, Wheaton, Maryland, USA.,Rheumatology, Georgetown University School of Medicine, Washington, DC, USA
| | - Narandra Bethina
- Division of Rheumatology, University of Vermont, Burlington, Vermont, USA
| | - Eugene Y Kissin
- Rheumatology, Boston University Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
125
|
Kloppenburg M, Berenbaum F. Osteoarthritis year in review 2019: epidemiology and therapy. Osteoarthritis Cartilage 2020; 28:242-248. [PMID: 31945457 DOI: 10.1016/j.joca.2020.01.002] [Citation(s) in RCA: 285] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/03/2019] [Accepted: 01/08/2020] [Indexed: 02/02/2023]
Abstract
Over the past year many studies and clinical trials have been published in the osteoarthritis (OA) field. This review is based on systematic literature review covering the period May 1st, 2018 to April 19th, 2019; the final selection of articles was subjective. Specifically those articles considered to be presenting novel insights and of potential importance for clinical practice, are discussed. Further evidence has emerged that OA is a serious disease with increasing impact worldwide. Our understanding of development of pain in OA has increased. Detailed studies investigating widely used pharmacological treatments have shown the benefits to be limited, whereas the risks seem higher than expected, suggesting further studies and reconsideration of currently used guidelines. Promising new pharmacological treatments have been developed and published, however subsequent studies are warranted. While waiting for new treatment modalities to appear joint replacement is an effective alternative; new data have become available on how long they might last.
Collapse
Affiliation(s)
- M Kloppenburg
- Departments of Rheumatology and Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands.
| | - F Berenbaum
- Sorbonne Université, INSERM CRSA, Department of Rheumatology, AP-HP Saint-Antoine Hospital, Paris, France
| |
Collapse
|
126
|
New Molecular Targets in the Therapy of Arthrosis Patients. ARS MEDICA TOMITANA 2020. [DOI: 10.2478/arsm-2019-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Introduction: Arthrosis represents the progressive degeneration of the joint cartilage, accompanied by the narrowing of the articular space and inflammation, which affects 70% of the population after the age of 60. Research purpose: This paper reviews the opportunity of using proinflammatory cytokine inhibitors as a means of stopping the progress of arthrosis. Material and method: As a result to a research into various clinical trial registers (Arthritis Clinical Trials, Clinical Research and Drug Information) and on specialized e-platforms, 5 randomized, multicentric double-blind clinical studies have been identified, which monitored the efficiency of various biological molecules in the treatment of arthrosis (etanercept, adalimumab, litikizumab, fasinumab and tanezumab). Results: The current pharmacological interventions consist mainly in the prescription of analgesics (acetaminophen, opioid analgesics), non-steroidal and chondroprotective anti-inflammatories. The proinflammatory cytokine inhibitors are already widely used in the inflammatory joint diseases, such as the rheumatoid polyarthritis. Their introduction into the treatment of arthrosis blocks the disease’s etiopathogenic mechanisms. Discussions: Arthrosis physiopathology involves a series of systemic, biological, biochemical factors, molecular and enzymatic processes that generate minimum inflammation. IL-1b and TNF-α are two major cytokines produced by the synovial cells and chondrocytes, which are involved in the destruction of the cartilage matrix by stimulating the production of proteolytic enzymes (MMP and aggrecanase). Conclusions: The utilisation of proinflammatory cytokine inhibitors in arthrosis represents a therapeutic option that requires studies in order to establish whether the introduction of proinflammatory cytokine inhibitors in arthrosis therapy might slow down the disease’s etiopathogenic mechanisms.
Collapse
|
127
|
Affiliation(s)
- Xavier Chevalier
- Department of Rheumatology, Hospital Henri Mondor, UPEC Paris XII, Créteil, France.
| | - Florent Eymard
- Department of Rheumatology, Hospital Henri Mondor, UPEC Paris XII, Créteil, France
| |
Collapse
|
128
|
Spel L, Martinon F. Inflammasomes contributing to inflammation in arthritis. Immunol Rev 2020; 294:48-62. [DOI: 10.1111/imr.12839] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Lotte Spel
- Departement of Biochemistry University of Lausanne Epalinges Switzerland
| | - Fabio Martinon
- Departement of Biochemistry University of Lausanne Epalinges Switzerland
| |
Collapse
|
129
|
Kahlenberg JM, Kang I. Advances in Disease Mechanisms and Translational Technologies: Clinicopathologic Significance of Inflammasome Activation in Autoimmune Diseases. Arthritis Rheumatol 2020; 72:386-395. [PMID: 31562704 DOI: 10.1002/art.41127] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/24/2019] [Indexed: 12/14/2022]
Abstract
Autoimmune diseases are characterized by dysregulated immune tolerance to self and inflammatory damage to tissues and organs. The development of inflammation involves multiple innate and adaptive immune pathways. Inflammasomes are multimeric cytosolic protein complexes that form to mediate host immune responses upon recognizing pathogen- or damage-associated molecular patterns via pattern-recognition receptors (PRRs). The accelerating pace of inflammasome research has demonstrated important roles for inflammasome activation in many pathologic conditions, including infectious, metabolic, autoinflammatory, and autoimmune diseases. The inflammasome generally comprises a PRR, procaspase 1, and an adaptor molecule connecting the PRR and procaspase 1. Upon inflammasome activation, procaspase 1 becomes active caspase 1 that converts pro-interleukin-1β (proIL-1β) and proIL-18 into mature and active IL-1β and IL-18, respectively. The cytokines IL-1β and IL-18 have multipotent effects on immune and nonimmune cells and induce and promote systemic and local inflammatory responses. Human studies have shown increased levels of these cytokines, altered activation of inflammasome-related molecules, and/or the presence of inflammasome activators in rheumatic diseases, including systemic lupus erythematosus, rheumatoid arthritis, crystal-induced arthropathies, and Sjögren's syndrome. Such changes are found in the primary target organs, such as the kidneys, joints, and salivary glands, as well as in the cardiovascular system. In animal models of rheumatic diseases, inflammation and tissue damage improve upon genetic or pharmacologic targeting of the inflammasome, supporting its pathogenic role. Herein, we review the clinicopathologic significance and therapeutic targeting of inflammasome activation in rheumatic diseases and related conditions based on recent findings.
Collapse
|
130
|
Bispecific Antibodies for Autoimmune and Inflammatory Diseases: Clinical Progress to Date. BioDrugs 2020; 34:111-119. [DOI: 10.1007/s40259-019-00400-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
131
|
Mobasheri A, Saarakkala S, Finnilä M, Karsdal MA, Bay-Jensen AC, van Spil WE. Recent advances in understanding the phenotypes of osteoarthritis. F1000Res 2019; 8. [PMID: 31885861 PMCID: PMC6913225 DOI: 10.12688/f1000research.20575.1] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/05/2019] [Indexed: 01/15/2023] Open
Abstract
Recent research in the field of osteoarthritis (OA) has focused on understanding the underlying molecular and clinical phenotypes of the disease. This narrative review article focuses on recent advances in our understanding of the phenotypes of OA and proposes that the disease represents a diversity of clinical phenotypes that are underpinned by a number of molecular mechanisms, which may be shared by several phenotypes and targeted more specifically for therapeutic purposes. The clinical phenotypes of OA supposedly have different underlying etiologies and pathogenic pathways and they progress at different rates. Large OA population cohorts consist of a majority of patients whose disease progresses slowly and a minority of individuals whose disease may progress faster. The ability to identify the people with relatively rapidly progressing OA can transform clinical trials and enhance their efficiency. The identification, characterization, and classification of molecular phenotypes of rapidly progressing OA, which represent patients who may benefit most from intervention, could potentially serve as the basis for precision medicine for this disabling condition. Imaging and biochemical markers (biomarkers) are important diagnostic and research tools that can assist with this challenge.
Collapse
Affiliation(s)
- Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, 08661, Lithuania.,Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, FI-90014, Finland.,Centre for Sport, Exercise and Osteoarthritis Research Versus Arthritis, Queen's Medical Centre, Nottingham, UK.,ImmunoScience, Nordic Bioscience Biomarkers and Research, Herlev, DK-2730, Denmark
| | - Simo Saarakkala
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, FI-90014, Finland
| | - Mikko Finnilä
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, FI-90014, Finland
| | - Morten A Karsdal
- ImmunoScience, Nordic Bioscience Biomarkers and Research, Herlev, DK-2730, Denmark
| | | | - Willem Evert van Spil
- Division of Internal Medicine & Dermatology, Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.,Rheumatology, Dijklander Hospital, 1620 AR Hoorn, The Netherlands
| |
Collapse
|
132
|
The IL-1 family of cytokines and receptors in rheumatic diseases. Nat Rev Rheumatol 2019; 15:612-632. [DOI: 10.1038/s41584-019-0277-8] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2019] [Indexed: 02/07/2023]
|
133
|
Abstract
Advances in the treatment of rheumatoid arthritis (RA) are attributed to several aspects such as new classification criteria enabling early diagnosis and intensive treatment with the application of treat-to-target principles as well as better understanding of the pathogenesis of RA contributing to the development of targeted therapies. However, reaching remission is still not achieved in most patients with RA, which is one of the driving forces behind the continuous development of novel therapies and the optimization of therapeutic strategies. This review will outline several new therapeutic antibodies modulating anti-inflammatory cytokines interleukin (IL)-2 and IL-10 and pro-inflammatory mediators granulocyte-macrophage colony-stimulating factor, fractalkine, and IL-6 that are in various stages of clinical development as well as the progress in manufacturing biotechnologies contributing to the next generation of antibodies and their potential to expand the therapeutic armamentarium for RA. In addition, the fate of unsuccessful therapies including agents targeting IL-15, the IL-20 family, IL-21, chemokine CXCL10, B-cell activating factor (BAFF), and regulatory T (Treg) cells or a novel concept targeting synovial fibroblasts via cadherin-11 will be discussed.
Collapse
Affiliation(s)
- Ladislav Senolt
- Department of Rheumatology, First Faculty of Medicine, Charles University, Institute of Rheumatology, Prague, Czech Republic, 128 50, Czech Republic
| |
Collapse
|
134
|
Kloppenburg M, Levesque M. Response to: 'Can IL-1 be used as a target for osteoarthritis?' by Cheng et al. Ann Rheum Dis 2019; 79:e89. [PMID: 31248854 DOI: 10.1136/annrheumdis-2019-215612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 06/15/2019] [Indexed: 11/04/2022]
Affiliation(s)
| | - Marc Levesque
- Immunology Development, AbbVie, North Chicago, Illinois, USA
| |
Collapse
|
135
|
Abstract
The concept of interleukin-1 (IL-1) as a target in osteoarthritis (OA) has been an attractive one for many years. It is a highly potent inducer of cartilage degradation, causing the induction of mRNA and controlling the bioavailability of disease-relevant proteases such as ADAMTS5 and MMP13. It drives synovitis and can induce other disease-relevant genes such as nerve growth factor, a key pain sensitiser in OA. However, the quality of evidence for its involvement in disease is modest. Descriptive studies have demonstrated expression of IL-1α and β in OA cartilage and elevated levels in the synovial fluid of some patients. Agnostic transcriptomic and genomic analyses do not identify IL-1 as a key pathway.
In vivo models show a conflicting role for this molecule; early studies using therapeutic approaches in large animal models show a benefit, but most murine studies fail to demonstrate protection where the ligands (IL-1α/β), the cytokine activator (IL-1–converting enzyme), or the receptor (IL-1R) have been knocked out. Recently, a number of large double-blind randomised controlled clinical studies targeting IL-1 have failed. Enthusiasm for IL-1 as a target in OA is rapidly dwindling.
Collapse
Affiliation(s)
- Tonia L Vincent
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| |
Collapse
|
136
|
Abstract
PURPOSE OF THE REVIEW Osteoarthritis is widely regarded as a spectrum of conditions that affect all joint tissues, typified by a common entity: cartilage loss. Here, we review recent progress and challenges in chondroprotection and discuss new strategies to prevent cartilage loss in osteoarthritis. RECENT FINDINGS Advances in clinical, molecular, and cellular characterization are enabling improved stratification of osteoarthritis subtypes. Integration of next-generation sequencing and "omics" approaches with clinically relevant readouts shows promise in delineating both subtypes of disease and meaningful trial end points. Novel delivery strategies are enabling joint-specific delivery. Chondroprotection requires a whole joint approach, stratification of patient groups, and use of patient-relevant end points. Drug development should continue to explore new targets, while using modern technologies and recent knowledge to re-visit unsuccessful therapeutics from the past. The overarching goal for chondroprotection is to provide the right treatment(s) for the right patient at the right time.
Collapse
Affiliation(s)
- Jolet Y Mimpen
- The Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, OX3 7LD, UK
| | - Sarah J B Snelling
- The Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, OX3 7LD, UK.
| |
Collapse
|