101
|
Autologous Biologic Treatment for Equine Musculoskeletal Injuries: Platelet-Rich Plasma and IL-1 Receptor Antagonist Protein. Vet Clin North Am Equine Pract 2011; 27:275-98. [DOI: 10.1016/j.cveq.2011.05.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
102
|
Cohen SB, Proudman S, Kivitz AJ, Burch FX, Donohue JP, Burstein D, Sun YN, Banfield C, Vincent MS, Ni L, Zack DJ. A randomized, double-blind study of AMG 108 (a fully human monoclonal antibody to IL-1R1) in patients with osteoarthritis of the knee. Arthritis Res Ther 2011; 13:R125. [PMID: 21801403 PMCID: PMC3239365 DOI: 10.1186/ar3430] [Citation(s) in RCA: 189] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 01/06/2011] [Accepted: 07/29/2011] [Indexed: 11/24/2022] Open
Abstract
Introduction AMG 108 is a fully human, immunoglobulin subclass G2 (IgG2) monoclonal antibody that binds the human interleukin-1 (IL-1) receptor type 1, inhibiting the activity of IL-1a and IL-1b. In preclinical studies, IL-1 inhibition was shown to be beneficial in models of osteoarthritis (OA). The purpose of this two-part study was to evaluate the safety and pharmacokinetics (PK; Part A) and clinical effect (Part B) of AMG 108 in a double-blind, placebo-controlled, multiple-dose study in patients with OA of the knee. Methods In Part A, patients received placebo or AMG 108 subcutaneously (SC; 75 mg or 300 mg) or intravenously (IV; 100 mg or 300 mg) once every 4 weeks for 12 weeks; in Part B, patients received placebo or 300 mg AMG 108 SC, once every 4 weeks for 12 weeks. The clinical effect of AMG 108 was measured in Part B by using the Western Ontario and McMaster Universities (WOMAC) osteoarthritis index pain score. Results In Part A, 68 patients were randomized, and 64 received investigational product. In Part B, 160 patients were randomized, and 159 received investigational product. AMG 108 was well tolerated. Most adverse events (AEs), infectious AEs, serious AEs and infections, as well as withdrawals from the study due to AEs occurred at similar rates in both active and placebo groups. One death was reported in an 80-year-old patient (Part A, 300 mg IV AMG 108; due to complications of lobar pneumonia). AMG 108 serum concentration-time profiles exhibited nonlinear PK. The AMG 108 group in Part B had statistically insignificant but numerically greater improvement in pain compared with the placebo group, as shown by the WOMAC pain scores (median change, -63.0 versus -37.0, respectively). Conclusions The safety profile of AMG 108 SC and IV was comparable with placebo in patients with OA of the knee. Patients who received AMG 108 showed statistically insignificant but numerically greater improvements in pain; however, minimal, if any, clinical benefit was observed. Trial Registration This study is registered with ClinicalTrials.gov with the identifier NCT00110942.
Collapse
Affiliation(s)
- Stanley B Cohen
- Rheumatology, Metroplex Clinical Research Center, 8144 Walnut Hill Lane, Dallas, TX 75231, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Bilge O, Doral MN, Atesok K, Atay OA, Donmez G, Turhan E, Uzumcugil A, Leblebicioglu G, Kaya D, Bilgili H, Sargon M. The effects of the synovium on chondrocyte growth: an experimental study. Knee Surg Sports Traumatol Arthrosc 2011; 19:1214-23. [PMID: 21290114 DOI: 10.1007/s00167-010-1391-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 12/31/2010] [Indexed: 10/18/2022]
Abstract
PURPOSE The objective of this study was to evaluate the effects of synovium on the proliferation of the cartilage tissue and chondrocytes using a rabbit knee model as an in vivo synovial culture medium. METHODS Twelve New Zealand rabbits were used as the animal model in this investigation. Standard size chondral and osteochondral cartilage grafts were taken from, respectively, the left and right knees of all the animals. Two groups of 6 animals were formed: in Group I (synovium group), grafts were placed into the synovial tissue and in group II (patellar tendon group) behind the patellar tendon of the corresponding knees. After 4 months, samples were collected and evaluated macroscopically by measuring their dimensions (vertical = D1, horizontal = D2, and depth = D3) and volumes, and histologically by counting the chondrocyte number using camera lucida method. RESULTS Macroscopically, the increase in average D1, D2, and D3 measurements and volume in the osteochondral specimens were significantly higher compared to the chondral specimens in both groups (P < 0.05). However, no significant difference was observed between the two groups in terms of macroscopic values. Histologically, the mean chondrocyte counts in osteochondral and chondral specimens for Group I (synovium) were 20.2 and 18.1, and for Group II (patellar tendon) were 18.7 and 15.6, respectively. The mean number of chondrocytes was found to be significantly higher in osteochondral specimens than that of chondral specimens in either group (P < 0.05). Overall average chondrocyte count was significantly higher for Group I compared to Group II (P < 0.05). CONCLUSION Transplantation of the cartilage grafts into the synovial tissue in rabbit knees significantly enhanced the chondrocyte production compared with the group where the grafts were transplanted into intra-articular patellar tendon. The results of this study indicate that native synovial tissue may have the potential to be used as an in vivo culture medium for osteochondral tissue growth.
Collapse
Affiliation(s)
- Onur Bilge
- Faculty of Medicine, Department of Orthopaedics and Traumatology, Hacettepe University, Ankara, Turkey.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Attur M, Belitskaya-Lévy I, Oh C, Krasnokutsky S, Greenberg J, Samuels J, Smiles S, Lee S, Patel J, Al-Mussawir H, McDaniel G, Kraus VB, Abramson SB. Increased interleukin-1β gene expression in peripheral blood leukocytes is associated with increased pain and predicts risk for progression of symptomatic knee osteoarthritis. ARTHRITIS AND RHEUMATISM 2011; 63:1908-17. [PMID: 21717421 PMCID: PMC3128429 DOI: 10.1002/art.30360] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To evaluate whether gene expression profiles could serve as biomarkers of symptomatic knee osteoarthritis (OA) by examining gene expression profiles in peripheral blood leukocytes (PBLs) from patients with OA compared with those from non-OA controls, and to determine whether candidate genomic biomarkers (PBL expression of inflammatory genes) predict an increased risk of disease progression in patients with symptomatic radiographic knee OA. METHODS Three independent cohorts of patients with knee OA and non-OA control subjects were studied. Two cohorts (a learning cohort and a validation cohort) were recruited at New York University Hospital for Joint Diseases (NYUHJD), and 1 cohort (a validation cohort) was recruited at Duke University Medical Center. PBL gene expression was assessed using Affymetrix microarray and was confirmed by quantitative polymerase chain reaction (qPCR). Radiographic progression at 2 years was assessed in 86 patients. RESULTS We identified 173 genes that were significantly up-regulated or down-regulated (≥1.5-fold change) in OA PBLs, at a false discovery rate of 5%. Cluster analysis revealed 2 distinct subgroups among the patients with OA: those in whom the expression of interleukin-1β (IL-1β) was increased ≥2-fold compared with controls, and those in whom the expression of IL-1β was comparable with that in controls. Overexpression of IL-1β in these OA subclasses was validated using qPCR in all 3 cohorts. Patients with the inflammatory "IL-1β signature" had higher pain scores and decreased function and were at higher risk of radiographic progression of OA. CONCLUSION PBLs from patients with symptomatic knee OA display a characteristic transcriptome profile. Moreover, increased expression of IL-1β identifies a subset of patients with OA who have increased pain and are at higher risk of radiographic progression of OA.
Collapse
Affiliation(s)
- Mukundan Attur
- Division of Rheumatology, New York University School of Medicine and NYU Hospital for Joint Diseases, New York, NY 10003
| | - Ilana Belitskaya-Lévy
- Division of Biostatistics, New York University School of Medicine, New York, NY 10016
| | - Cheongeun Oh
- Division of Biostatistics, New York University School of Medicine, New York, NY 10016
| | - Svetlana Krasnokutsky
- Division of Rheumatology, New York University School of Medicine and NYU Hospital for Joint Diseases, New York, NY 10003
| | - Jeffrey Greenberg
- Division of Rheumatology, New York University School of Medicine and NYU Hospital for Joint Diseases, New York, NY 10003
| | - Jonathan Samuels
- Division of Rheumatology, New York University School of Medicine and NYU Hospital for Joint Diseases, New York, NY 10003
| | - Stephen Smiles
- Division of Rheumatology, New York University School of Medicine and NYU Hospital for Joint Diseases, New York, NY 10003
| | - Sicy Lee
- Division of Rheumatology, New York University School of Medicine and NYU Hospital for Joint Diseases, New York, NY 10003
| | - Jyoti Patel
- Division of Rheumatology, New York University School of Medicine and NYU Hospital for Joint Diseases, New York, NY 10003
| | - Hayf Al-Mussawir
- Division of Rheumatology, New York University School of Medicine and NYU Hospital for Joint Diseases, New York, NY 10003
| | | | | | - Steven B. Abramson
- Division of Rheumatology, New York University School of Medicine and NYU Hospital for Joint Diseases, New York, NY 10003
| |
Collapse
|
105
|
Abstract
The concept of using gene transfer strategies for cartilage repair originates from the idea of transferring genes encoding therapeutic factors into the repair tissue, resulting in a temporarily and spatially defined delivery of therapeutic molecules to sites of cartilage damage. This review focuses on the potential benefits of using gene therapy approaches for the repair of articular cartilage and meniscal fibrocartilage, including articular cartilage defects resulting from acute trauma, osteochondritis dissecans, osteonecrosis, and osteoarthritis. Possible applications for meniscal repair comprise meniscal lesions, meniscal sutures, and meniscal transplantation. Recent studies in both small and large animal models have demonstrated the applicability of gene-based approaches for cartilage repair. Chondrogenic pathways were stimulated in the repair tissue and in osteoarthritic cartilage using genes for polypeptide growth factors and transcription factors. Although encouraging data have been generated, a successful translation of gene therapy for cartilage repair will require an ongoing combined effort of orthopedic surgeons and of basic scientists.
Collapse
Affiliation(s)
- Henning Madry
- Saarland University, Homburg, Germany,Henning Madry, Saarland University, Kirrbergerstrasse 1, Homburg, 66424 Germany
| | | | | |
Collapse
|
106
|
Allen KD, Adams SB, Mata BA, Shamji MF, Gouze E, Jing L, Nettles DL, Latt LD, Setton LA. Gait and behavior in an IL1β-mediated model of rat knee arthritis and effects of an IL1 antagonist. J Orthop Res 2011; 29:694-703. [PMID: 21437948 PMCID: PMC3100769 DOI: 10.1002/jor.21309] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 10/11/2010] [Indexed: 02/04/2023]
Abstract
Interleukin-1 beta (IL1β) is a proinflammatory cytokine that mediates arthritic pathologies. Our objectives were to evaluate pain and limb dysfunction resulting from IL1β over-expression in the rat knee and to investigate the ability of local IL1 receptor antagonist (IL1Ra) delivery to reverse-associated pathology. IL1β over-expression was induced in the right knees of 30 Wistar rats via intra-articular injection of rat fibroblasts retrovirally infected with human IL1β cDNA. A subset of animals received a 30 µl intra-articular injection of saline or human IL1Ra on day 1 after cell delivery (0.65 µg/µl hIL1Ra, n = 7 per group). Joint swelling, gait, and sensitivity were investigated over 1 week. On day 8, animals were sacrificed and joints were collected for histological evaluation. Joint inflammation and elevated levels of endogenous IL1β were observed in knees receiving IL1β-infected fibroblasts. Asymmetric gaits favoring the affected limb and heightened mechanical sensitivity (allodynia) reflected a unilateral pathology. Histopathology revealed cartilage loss on the femoral groove and condyle of affected joints. Intra-articular IL1Ra injection failed to restore gait and sensitivity to preoperative levels and did not reduce cartilage degeneration observed in histopathology. Joint swelling and degeneration subsequent to IL1β over-expression is associated limb hypersensitivity and gait compensation. Intra-articular IL1Ra delivery did not result in marked improvement for this model; this may be driven by rapid clearance of administered IL1Ra from the joint space. These results motivate work to further investigate the behavioral consequences of monoarticular arthritis and sustained release drug delivery strategies for the joint space.
Collapse
Affiliation(s)
- Kyle D. Allen
- Department of Orthopaedic Surgery, Duke University Medical Center, Duke University, Durham, NC, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Samuel B Adams
- Department of Orthopaedic Surgery, Duke University Medical Center, Duke University, Durham, NC, USA
| | - Brian A. Mata
- Department of Orthopaedic Surgery, Duke University Medical Center, Duke University, Durham, NC, USA
| | - Mohammed F. Shamji
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Division of Neurosurgery, The Ottawa Hospital, Ottawa, ON, Canada
| | - Elvire Gouze
- INSERM U563, Centre de Physiopathologie de Toulouse Purpan, BP3028, 31024 Toulouse Cedex 3, France
| | - Liufang Jing
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Dana L. Nettles
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - L. Daniel Latt
- Department of Orthopaedic Surgery, Duke University Medical Center, Duke University, Durham, NC, USA
| | - Lori A. Setton
- Department of Orthopaedic Surgery, Duke University Medical Center, Duke University, Durham, NC, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
107
|
Santangelo KS, Pieczarka EM, Nuovo GJ, Weisbrode SE, Bertone AL. Temporal expression and tissue distribution of interleukin-1β in two strains of guinea pigs with varying propensity for spontaneous knee osteoarthritis. Osteoarthritis Cartilage 2011; 19:439-48. [PMID: 21251992 PMCID: PMC3073778 DOI: 10.1016/j.joca.2011.01.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 12/13/2010] [Accepted: 01/03/2011] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To provide a comprehensive immunohistochemical (IHC) map of the temporal expression and tissue distribution of interleukin-1β (IL-1β) through progression of osteoarthritis (OA) in two strains of guinea pigs with varying propensity for spontaneous knee joint disease. METHODS OA-prone Hartley and OA-resistant Strain 13 guinea pigs were collected at 60, 120, 180, 240, 360, and 480 days of age (N=4 animals per strain per date). IHC was performed on whole joint preparations; the distribution of IL-1β expression on coronal sections was mapped, semi-quantitatively scored, and correlated to OA grade using Mankin criteria with guinea pig-specific modifications. OA and IHC indices were compared among times and between strains using the Kruskal-Wallis one-way analysis of variance by ranks followed by Dunn's post test. RESULTS OA indices for both strains increased from 60 to 480 days of age; a statistically higher score (P ≤ 0.01) was found in Hartley animals at 180, 240, 360, and 480 days. At 60 days of age, IL-1β expression was detected in cartilage, menisci, synovium, and subchondral bone in both strains. Persistent and statistically increased (P<0.05) IL-1β expression was found in these same tissues in Hartley animals at 120 and 180 days, while Strain 13 animals demonstrated a significant reduction in positive immunostaining. Statistical differences in IHC indices between strains beyond 240 days of age were restricted to synovium (days 240 and 480) and subchondral bone (days 360 and 480). CONCLUSIONS As expected, histologic OA proceeded in an accelerated manner in Hartley animals relative to Strain 13 animals. The OA-prone strain did not demonstrate reduced IL-1β expression during adult maturity as occurred in the OA-resistant strain, and this persistent expression may have corresponded to early incidence of OA. Future interventional studies are warranted to explore whether dysregulation of IL-1β expression may contribute to premature onset of spontaneous disease in the Hartley guinea pig.
Collapse
Affiliation(s)
- K. S. Santangelo
- Departments of Veterinary Clinical Sciences and Biosciences, The Ohio State University, Columbus, OH, USA
| | - E. M. Pieczarka
- Departments of Veterinary Clinical Sciences and Biosciences, The Ohio State University, Columbus, OH, USA
| | - G. J. Nuovo
- Department of Pathology, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - S. E. Weisbrode
- Departments of Veterinary Clinical Sciences and Biosciences, The Ohio State University, Columbus, OH, USA
| | - A. L. Bertone
- Departments of Veterinary Clinical Sciences and Biosciences, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
108
|
Al-Saffar F, Ganabadi S, Fakuraz S. Response of Channa striatus Extract Against Monosodium Iodoacetate Induced Osteoarthritis in Rats. ACTA ACUST UNITED AC 2011. [DOI: 10.3923/javaa.2011.460.469] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
109
|
Kokebie R, Aggarwal R, Lidder S, Hakimiyan AA, Rueger DC, Block JA, Chubinskaya S. The role of synovial fluid markers of catabolism and anabolism in osteoarthritis, rheumatoid arthritis and asymptomatic organ donors. Arthritis Res Ther 2011; 13:R50. [PMID: 21435227 PMCID: PMC3132039 DOI: 10.1186/ar3293] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 02/09/2011] [Accepted: 03/24/2011] [Indexed: 12/20/2022] Open
Abstract
Introduction The purpose of this study was to correlate the level of anabolic and catabolic biomarkers in synovial fluid (SF) from patients with rheumatoid arthritis (RA), patients with osteoarthritis (OA) and asymptomatic organ donors. Methods SF was collected from the knees of 45 OA, 22 RA patients and 20 asymptomatic organ donors. Eight biomarkers were selected and analyzed by using an enzyme-linked immunosorbent assay: interleukin (IL)-1, IL-6, IL-8 and IL-11; leukemia-inhibitory factor (LIF); cartilage oligomeric protein (COMP); osteocalcin; and osteogenic protein 1 (OP-1). Data are expressed as medians (interquartile ranges). The effects of sex and disease activity were assessed on the basis of the Western Ontario and McMaster Universities index score for patients with OA and on the basis of white blood cell count, erythrocyte sedimentation rate and C-reactive protein level for patients with RA. Results The mean ages (± SD) of the patients were as follows: 53 ± 9 years for patients with OA, 54 ± 11 years for patients with RA and 52 ± 7 years for asymptomatic organ donors. No effect of participants' sex was identified. In the SF of patients with RA, four of five cytokines were higher than those in the SF of patients with OA and those of asymptomatic organ donors. The most significant differences were found for IL-6 and IL-8, where IL-6 concentration in SF of patients with RA was almost threefold higher than that in patients with OA and fourfold higher than that in asymptomatic donor controls: 354.7 pg/ml (1,851.6) vs. 119.4 pg/ml (193.2) vs. 86.97 pg/ml (82.0) (P < 0.05 and P < 0.05, respectively). IL-8 concentrations were higher in SF of patients with RA than that in patients with OA as well as that in asymptomatic donor controls: 583.6 pg/ml (1,086.4) vs. 429 pg/ml (87.3) vs. 451 pg/ml (170.1) (P < 0.05 and P < 0.05, respectively). No differences were found for IL-11 in the SF of patients with RA and that of patients with OA, while a 1.4-fold difference was detected in the SF of patients with OA and that of asymptomatic donor controls: 296.2 pg/ml (257.2) vs. 211.6 pg/ml (40.8) (P < 0.05). IL-1 concentrations were the highest in the SF of RA patients (9.26 pg/ml (11.1)); in the SF of asymptomatic donors, it was significantly higher than that in patients with OA (9.083 pg/ml (1.6) vs. 7.76 pg/ml (2.6); P < 0.05). Conversely, asymptomatic donor control samples had the highest LIF concentrations: 228.5 pg/ml (131.6) vs. 128.4 pg/ml (222.7) in the SF of patients with RA vs. 107.5 pg/ml (136.9) in the SF of patients with OA (P < 0.05). OP-1 concentrations were twofold higher in the SF of patients with RA than those in patients with OA and threefold higher than those in asymptomatic donor control samples (167.1 ng/ml (194.8) vs. 81.79 ng/ml (116.0) vs. 54.49 ng/ml (29.3), respectively; P < 0.05). The differences in COMP and osteocalcin were indistinguishable between the groups, as were the differences between active and inactive OA and RA. Conclusions Activation of selected biomarkers corresponds to the mechanisms that drive each disease. IL-11, LIF and OP-1 may be viewed as a cluster of biomarkers significant for OA; while profiling of IL-1, IL-6, IL-8, LIF and OP-1 may be more significant in RA. Larger, better-defined patient cohorts are necessary to develop a biomarker algorithm for prognostic use.
Collapse
Affiliation(s)
- Rediet Kokebie
- Section of Rheumatology, Department of Internal Medicine, Rush University Medical Center, 1653 West Congress Parkway, Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
110
|
Coleman CM, Curtin C, Barry FP, O'Flatharta C, Murphy JM. Mesenchymal stem cells and osteoarthritis: remedy or accomplice? Hum Gene Ther 2011; 21:1239-50. [PMID: 20649459 DOI: 10.1089/hum.2010.138] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multipotent mesenchymal stromal or stem cells (MSCs) are likely to be agents of connective tissue homeostasis and repair. Because the hallmark of osteoarthritis (OA) is degeneration and failure to repair connective tissues it is compelling to think that these cells have a role to play in OA. Indeed, MSCs have been implicated in the pathogenesis of OA and, in turn, progression of the disease has been shown to be therapeutically modulated by MSCs. This review discusses current knowledge on the potential of both marrow- and local joint-derived MSCs in OA, the mode of action of the cells, and possible effects of the osteoarthritic niche on the function of MSCs. The use of stem cells for repair of isolated cartilage lesions and strategies for modulation of OA using local cell delivery are discussed as well as therapeutic options for the future to recruit and appropriately activate endogenous progenitors and/or locally systemically administered MSCs in the early stages of the disease. The use of gene therapy protocols, particularly as they pertain to modulation of inflammation associated with the osteoarthritic niche, offer an additional option in the treatment of this chronic disease. In summary, elucidation of the etiology of OA and development of technologies to detect early disease, allied to an increased understanding of the role MSCs in aging and OA, should lead to more targeted and efficacious treatments for this debilitating chronic disease in the future.
Collapse
Affiliation(s)
- Cynthia M Coleman
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | | | | | | | | |
Collapse
|
111
|
Chen B, Qin J, Wang H, Magdalou J, Chen L. Effects of adenovirus-mediated bFGF, IL-1Ra and IGF-1 gene transfer on human osteoarthritic chondrocytes and osteoarthritis in rabbits. Exp Mol Med 2011; 42:684-95. [PMID: 20733349 DOI: 10.3858/emm.2010.42.10.067] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The study investigated the effects of adenovirus-mediated gene transfection of basic fibroblast growth factor (bFGF), bFGF combined with interleukin-1 receptor antagonist protein (IL-Ra) and/or insulin-like growth factor-1 (IGF-1) both in human osteoarthritis (OA) chondrocytes and rabbits OA model. Human OA chondrocytes were delivered by adenovirus-mediated bFGF, IL-Ra and IGF-1 vectors, respectively. Chondrocyte proliferation, glycosaminoglycan (GAG) content, expression of type II collagen, ADAMTS-5, MMP-13, MMP-3 and TIMP-1 were determined. Rabbit OA model was induced by anterior cruciate ligament transaction (ACLT) in knees. Adenoviral vectors encoding human bFGF, IL-Ra and IGF-1 were injected intraarticularly into the knee joints after ACLT. The effects of adenovirus-mediated gene transfection on rabbit OA were evaluated. In vitro, the transfected genes were expressed in cell supernatant of human OA chondrocytes. AdbFGF group significantly promoted chondrocyte proliferation, and increased GAG and type II collagen synthesis than in the OA group. As two or three genes were transfected in different combinations, there was significant enhancement on the GAG content, type II collagen synthesis, and TIMP-1 levels, while ADAMTS-5, MMP-13, and MMP-3 levels were reduced. In vivo, the transfected genes were expressed in synovial fluid of rabbits. Intraarticular delivery of bFGF enhanced the expression of type II collagen in cartilage and decreased cartilage Mankin score compared with the OA control group (P=0.047; P<0.01, respectively). Multiple-gene transfection in different combinations showed better results than bFGF transfection alone. This study suggests that bFGF gene transfection is effective in treating experimental OA. Multiple gene transfection has better biologic effects on OA.
Collapse
Affiliation(s)
- Biao Chen
- Department of Orthopaedic Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | | | | | | | | |
Collapse
|
112
|
Jun JB, Kim JK, Kim TH, Na YI, Choi CH, Kim YH. Inhibition of the IL-1β-induced Expression of Matrix Metalloproteinases by Controlled Release of IL-1 Receptor Antagonist Using Injectable and Thermo-reversible Gels in Human Osteoarthritis Chondrocytes. JOURNAL OF RHEUMATIC DISEASES 2011. [DOI: 10.4078/jrd.2011.18.2.85] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Jae-Bum Jun
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
| | - Jang Kyoung Kim
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea
| | - Tae-Hwan Kim
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
| | - Young-In Na
- Institute of Rheumatism, Hanyang University, Seoul, Korea
| | - Choong Hyeok Choi
- Department of Surgery for Rheumatism, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
| | - Yong-Hee Kim
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea
| |
Collapse
|
113
|
Principles of tissue engineering and cell- and gene-based therapy. Rheumatology (Oxford) 2011. [DOI: 10.1016/b978-0-323-06551-1.00018-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
114
|
Evans CH, Ghivizzani SC, Robbins PD. Getting arthritis gene therapy into the clinic. Nat Rev Rheumatol 2010; 7:244-9. [PMID: 21135882 DOI: 10.1038/nrrheum.2010.193] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gene transfer technologies enable the controlled, targeted and sustained expression of gene products at precise anatomical locations, such as the joint. In this way, they offer the potential for more-effective, less-expensive treatments of joint diseases with fewer extra-articular adverse effects. A large body of preclinical data confirms the utility of intra-articular gene therapy in animal models of rheumatoid arthritis and osteoarthritis. However, relatively few clinical trials have been conducted, only one of which has completed phase II. This article summarizes the status in 2010 of the clinical development of gene therapy for arthritis, identifies certain constraints to progress and suggests possible solutions.
Collapse
Affiliation(s)
- Christopher H Evans
- Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, RN-115, Boston, MA 02215, USA.
| | | | | |
Collapse
|
115
|
Umlauf D, Frank S, Pap T, Bertrand J. Cartilage biology, pathology, and repair. Cell Mol Life Sci 2010; 67:4197-211. [PMID: 20734104 PMCID: PMC11115553 DOI: 10.1007/s00018-010-0498-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 07/27/2010] [Accepted: 08/04/2010] [Indexed: 02/07/2023]
Abstract
Osteoarthritis is one of the most common forms of musculoskeletal disease and the most prominent type of arthritis encountered in all countries. Although great efforts have been made to investigate cartilage biology and osteoarthritis pathology, the treatment has lagged behind that of other arthritides, as there is a lack of effective disease-modifying therapies. Numerous approaches for dealing with cartilage degradation have been tried, but enjoyed very little success to develop approved OA treatments with not only symptomatic improvement but also structure-modifying effect. In this review we discuss the most recent findings regarding the regulation of cartilage biology and pathology and highlight their potential therapeutic values.
Collapse
Affiliation(s)
- Daniel Umlauf
- Institute of Experimental Musculoskeletal Medicine IEMM, University Hospital Muenster, Domagkstrasse 3, 48149 Muenster, Germany
| | - Svetlana Frank
- Institute of Experimental Musculoskeletal Medicine IEMM, University Hospital Muenster, Domagkstrasse 3, 48149 Muenster, Germany
| | - Thomas Pap
- Institute of Experimental Musculoskeletal Medicine IEMM, University Hospital Muenster, Domagkstrasse 3, 48149 Muenster, Germany
| | - Jessica Bertrand
- Institute of Experimental Musculoskeletal Medicine IEMM, University Hospital Muenster, Domagkstrasse 3, 48149 Muenster, Germany
| |
Collapse
|
116
|
Abstract
Osteoarthritis (OA) is associated with cartilage destruction, subchondral bone remodeling and inflammation of the synovial membrane, although the etiology and pathogenesis underlying this debilitating disease are poorly understood. Secreted inflammatory molecules, such as proinflammatory cytokines, are among the critical mediators of the disturbed processes implicated in OA pathophysiology. Interleukin (IL)-1β and tumor necrosis factor (TNF), in particular, control the degeneration of articular cartilage matrix, which makes them prime targets for therapeutic strategies. Animal studies provide support for this approach, although only a few clinical studies have investigated the efficacy of blocking these proinflammatory cytokines in the treatment of OA. Apart from IL-1β and TNF, several other cytokines including IL-6, IL-15, IL-17, IL-18, IL-21, leukemia inhibitory factor and IL-8 (a chemokine) have also been shown to be implicated in OA and could possibly be targeted therapeutically. This Review discusses the current knowledge regarding the role of proinflammatory cytokines in the pathophysiology of OA and addresses the potential of anticytokine therapy in the treatment of this disease.
Collapse
|
117
|
Chevalier X, Kemta-Lepka F. Are biologics a treatment option in osteoarthritis? ACTA ACUST UNITED AC 2010. [DOI: 10.2217/thy.10.66] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
118
|
Wehling P, Reinecke J, Baltzer AWA, Granrath M, Schulitz KP, Schultz C, Krauspe R, Whiteside TW, Elder E, Ghivizzani SC, Robbins PD, Evans CH. Clinical responses to gene therapy in joints of two subjects with rheumatoid arthritis. Hum Gene Ther 2010; 20:97-101. [PMID: 18986219 DOI: 10.1089/hum.2008.075] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
This paper provides the first evidence of a clinical response to gene therapy in human arthritis. Two subjects with rheumatoid arthritis received ex vivo, intraarticular delivery of human interleukin-1 receptor antagonist (IL-1Ra) cDNA. To achieve this, autologous synovial fibroblasts were transduced with a retrovirus, MFG-IRAP, carrying IL-1Ra as the transgene, or remained as untransduced controls. Symptomatic metacarpophalangeal (MCP) joints were injected with control or transduced cells. Joints were clinically evaluated on the basis of pain; the circumference of MCP joint 1 was also measured. After 4 weeks, joints underwent surgical synovectomy. There were no adverse events in either subject. The first subject responded dramatically to gene transfer, with a marked and rapid reduction in pain and swelling that lasted for the entire 4 weeks of the study. Remarkably, joints receiving IL-1Ra cDNA were protected from flares that occurred during the study period. Analysis of RNA recovered after synovectomy revealed enhanced expression of IL-1Ra and reduced expression of matrix metalloproteinase-3 and IL-1beta. The second subject also responded with reduced pain and swelling. Thus, gene transfer to human, rheumatoid joints can be accomplished safely to produce clinical benefit, at least in the short term. Using this ex vivo procedure, the transgene persisted within the joint for at least 1 month. Further clinical studies are warranted.
Collapse
Affiliation(s)
- Peter Wehling
- Department of Orthopedic Surgery, University of Düsseldorf School of Medicine, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
119
|
Chevalier X. [Evolution of the pharmacological management of osteoarthritis: the biologics]. Presse Med 2010; 39:1164-71. [PMID: 20843646 DOI: 10.1016/j.lpm.2010.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 05/04/2010] [Indexed: 12/11/2022] Open
Abstract
Proinflammatory cytokines such as IL-1 β et TNF α are implicated in osteoarthritis where they contribute to the local synovitis. Blocking IL-1 β is a promising tool in experimental animal's model of osteoarthritis (OA). So far in human clinical trials using IL-1 blockers, the results are inconclusive. The blockade of TNF α seems more appropriated for inflammatory forms of hand OA with some encouraging results. Intra-articular administration of biologics is hampered by their short resident time into the joint. Gene therapy is one of the possible way to prolong the local intra-articular delivery of biologics.
Collapse
Affiliation(s)
- Xavier Chevalier
- Hôpital Henri-Mondor, service de rhumatologie, 94010 Créteil, France.
| |
Collapse
|
120
|
Fox BA, Stephens MM. Treatment of knee osteoarthritis with Orthokine-derived autologous conditioned serum. Expert Rev Clin Immunol 2010; 6:335-45. [PMID: 20441419 DOI: 10.1586/eci.10.17] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Osteoarthritis (OA) is the most prevalent arthritis in the world with increasing numbers of people expected to acquire the disease as the population ages. Therapies commonly used to manage the disease have limited efficacy and some carry significant risks. Current data suggest that the anti-inflammatory cytokine IL-1 receptor antagonist (IL-1Ra) can alter the inflammatory response and cartilage erosion present in OA. Intra-articular gene expression of IL-1Ra has shown promising results in animal models to provide symptomatic improvement and minimize osteoarthritic changes. Orthogen AG (Dusseldorf, Germany) has developed a method to produce an autologous conditioned serum (ACS) rich in IL-1Ra marketed as Orthokine. Study participants treated with ACS have improved pain and function; however, these results are preliminary and need confirmation. If ongoing trials prove that ACS can retard cartilage degeneration and reduce inflammation, the management of OA would be dramatically altered, perhaps providing a mechanism to prevent the disease or at least its progression.
Collapse
Affiliation(s)
- Beth Anne Fox
- Department of Family Medicine, East Tennessee State University, Kingsport, TN 37660, USA.
| | | |
Collapse
|
121
|
Cucchiarini M, Terwilliger EF, Kohn D, Madry H. Remodelling of human osteoarthritic cartilage by FGF-2, alone or combined with Sox9 via rAAV gene transfer. J Cell Mol Med 2010; 13:2476-2488. [PMID: 18705695 DOI: 10.1111/j.1582-4934.2008.00474.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Compensating for the loss of extracellular cartilage matrix, as well as counteracting the alterations of the chondrocyte phenotype in osteoarthritis are of key importance to develop effective therapeutic strategies against this disorder. In the present study, we analysed the benefits of applying a potent gene combination to remodel human osteoarthritic (OA) cartilage. We employed the promising recombinant adeno-associated virus (rAAV) vector to deliver the mitogenic fibroblast growth factor 2 (FGF-2) factor, alone or simultaneously with the transcription factor Sox9 as a key activator of matrix synthesis, to human normal and OA articular chondrocytes. We evaluated the effects of single (FGF-2) or combined (FGF-2/SOX9) transgene expression upon the regenerative activities of chondrocytes in three dimensional cultures in vitro and in cartilage explants in situ. Single overexpression of FGF-2 enhanced the survival and proliferation of both normal and OA chondrocytes, without stimulating the matrix synthetic processes in the increased pools of cells. The mitogenic properties of FGF-2 were maintained when SOX9 was co-overexpressed and concomitant with an increase in the production of proteoglycans and type-II collagen, suggesting that the transcription factor was capable of counterbalancing the effects of FGF-2 on matrix accumulation. Also important, expression of type-X collagen, a marker of hypertrophy strongly decreased following treatment by the candidate vectors. Most remarkably, the levels of activities achieved in co-treated human OA cartilage were similar to or higher than those observed in normal cartilage. The present findings show that combined expression of candidate factors in OA cartilage can re-establish key features of normal cartilage and prevent the pathological shift of metabolic homeostasis. These data provide further motivation to develop coupled gene transfer approaches via rAAV for the treatment of human OA.
Collapse
Affiliation(s)
- Magali Cucchiarini
- Laboratory for Experimental Orthopaedics, Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg, Germany
| | - Ernest F Terwilliger
- Division of Experimental Medicine, Harvard Institutes of Medicine and Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Dieter Kohn
- Laboratory for Experimental Orthopaedics, Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg, Germany
| | - Henning Madry
- Laboratory for Experimental Orthopaedics, Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
122
|
Goekoop RJ, Kloppenburg M, Kroon HM, Frölich M, Huizinga TWJ, Westendorp RGJ, Gussekloo J. Low innate production of interleukin-1beta and interleukin-6 is associated with the absence of osteoarthritis in old age. Osteoarthritis Cartilage 2010; 18:942-7. [PMID: 20417290 DOI: 10.1016/j.joca.2010.03.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 02/28/2010] [Accepted: 03/19/2010] [Indexed: 02/02/2023]
Abstract
OBJECTIVE We investigated whether innate differences in cytokine response were associated with the absence of osteoarthritis (OA) in old age. DESIGN In 82 participants from a cross-sectional birth cohort, radiographs of hands, hips and knees were taken at the age of 90 years. OA was defined as a Kellgren-Lawrence score of at least two. "Free from OA" was defined at patient level as absence of hip and knee OA, and presence of OA in maximally two hand joints. The innate cytokine response was determined in whole-blood samples upon stimulation with lipopolysaccharide. Logistic regression analyses were used to investigate associations between absence of OA in relation to tertiles of interleukin (IL)-1beta, IL-6, tumor necrosis factor (TNF)-alpha, IL-1 receptor antagonist (RA) and IL-10. Adjustments were made for gender and body mass index. RESULTS Sixteen percent of the participants were "free from OA". Subjects in the lowest tertile of Il-1beta production had a 11-fold increased chance to be free of OA [odds ratio (OR) 11.3, confidence intervals (CI) 95% 1.1-115.9], subjects in the lowest tertile of IL-6 production had an almost 7-fold increased chance to be free of OA (OR 6.7, 95% CI 1.1-41.2). Absence of hand OA was associated with low innate production of IL-6 and IL-1RA, absence of hip OA was associated with low innate IL-1beta production. No associations were found for TNF-alpha and IL-10. CONCLUSIONS Low innate capacity to produce the pro-inflammatory cytokines IL-1beta and IL-6 is associated with the absence of OA in old age.
Collapse
Affiliation(s)
- R J Goekoop
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
123
|
Lotz MK, Kraus VB. New developments in osteoarthritis. Posttraumatic osteoarthritis: pathogenesis and pharmacological treatment options. Arthritis Res Ther 2010; 12:211. [PMID: 20602810 PMCID: PMC2911903 DOI: 10.1186/ar3046] [Citation(s) in RCA: 210] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Joint trauma can lead to a spectrum of acute lesions, including osteochondral fractures, ligament or meniscus tears and damage to the articular cartilage. This is often associated with intraarticular bleeding and causes posttraumatic joint inflammation. Although the acute symptoms resolve and some of the lesions can be surgically repaired, joint injury triggers a chronic remodeling process in cartilage and other joint tissues that ultimately manifests as osteoarthritis in a majority of cases. The objective of the present review is to summarize information on pathogenetic mechanisms involved in the acute and chronic consequences of joint trauma and discuss potential pharmacological interventions. The focus of the review is on the early events that follow joint trauma since therapies for posttraumatic joint inflammation are not available and this represents a unique window of opportunity to limit chronic consequences.
Collapse
Affiliation(s)
- Martin K Lotz
- Department of Molecular and Experimental Medicine, The Scripps-Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | |
Collapse
|
124
|
Rutgers M, Saris DBF, Dhert WJA, Creemers LB. Cytokine profile of autologous conditioned serum for treatment of osteoarthritis, in vitro effects on cartilage metabolism and intra-articular levels after injection. Arthritis Res Ther 2010; 12:R114. [PMID: 20537160 PMCID: PMC2911907 DOI: 10.1186/ar3050] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2009] [Revised: 05/03/2010] [Accepted: 06/10/2010] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Intraarticular administration of autologous conditioned serum (ACS) recently demonstrated some clinical effectiveness in treatment of osteoarthritis (OA). The current study aims to evaluate the in vitro effects of ACS on cartilage proteoglycan (PG) metabolism, its composition and the effects on synovial fluid (SF) cytokine levels following intraarticular ACS administration. METHODS The effect of conditioned serum on PG metabolism of cultured OA cartilage explants was compared to unconditioned serum. The effect of serum conditioning on levels of interleukin-1beta (IL-1beta), IL-4, IL-6, IL-10, IL-13, interferon gamma (IFN-gamma), tumor necrosis factor alpha (TNF-alpha), osteoprotegerin (OPG), oncostatin M (OSM), interleukin-1 receptor (IL-1ra) and transforming growth factor beta (TGF-beta) were measured by multiplex ELISA. As TNF-alpha levels were found to be increased in conditioned serum, the effect of TNF-alpha inhibition by etanercept on PG metabolism was studied in cartilage explants cultured in the presence of conditioned serum. Furthermore, cytokine levels in SF were measured three days after intraarticular ACS injection in OA patients to verify their retention time in the joint space. RESULTS PG metabolism was not different in the presence of conditioned serum compared to unconditioned serum. Levels of the anti-inflammatory cytokines IL-1ra, TGF-beta, IL-10 as well as of pro-inflammatory cytokines IL-1beta, IL-6, TNF-alpha and OSM were increased. IL-4, IL-13 and IFN-gamma levels remained similar, while OPG levels decreased. TNF-alpha inhibition did not influence PG metabolism in cartilage explant culture in the presence of conditioned serum. Although OPG levels were higher and TGF-beta levels were clearly lower in ACS than in SF, intraarticular ACS injection in OA patients did not result in significant changes in these cytokine levels. CONCLUSIONS ACS for treatment of osteoarthritis contains increased levels of anti-inflammatory as well as pro-inflammatory cytokines, in particular TNF-alpha, but conditioned serum does not seem to have a net direct effect on cartilage metabolism, even upon inhibition of TNF-alpha. The fast intraarticular clearance of cytokines in the injected ACS may explain the limited effects found previously in vivo.
Collapse
Affiliation(s)
- Marijn Rutgers
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Daniël BF Saris
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Wouter JA Dhert
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Faculty of Veterinary Sciences, Utrecht University, Yalelaan 1, De Uithof, 3584 CL Utrecht, The Netherlands
| | - Laura B Creemers
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
125
|
Ulrich-Vinther M. Gene therapy methods in bone and joint disorders. ACTA ORTHOPAEDICA. SUPPLEMENTUM 2010. [DOI: 10.1080/17453690610046512] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
126
|
Attur M, Wang HY, Kraus VB, Bukowski JF, Aziz N, Krasnokutsky S, Samuels J, Greenberg J, McDaniel G, Abramson SB, Kornman KS. Radiographic severity of knee osteoarthritis is conditional on interleukin 1 receptor antagonist gene variations. Ann Rheum Dis 2010; 69:856-61. [PMID: 19934104 PMCID: PMC2925146 DOI: 10.1136/ard.2009.113043] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2009] [Indexed: 11/03/2022]
Abstract
BACKGROUND A lack of biomarkers that identify patients at risk for severe osteoarthritis (OA) complicates development of disease-modifying OA drugs. OBJECTIVE To determine whether inflammatory genetic markers could stratify patients with knee OA into high and low risk for destructive disease. METHODS Genotype associations with knee OA severity were assessed in two Caucasian populations. Fifteen single nucleotide polymorphisms (SNPs) in six inflammatory genes were evaluated for association with radiographic severity and with synovial fluid mediators in a subset of the patients. RESULTS Interleukin 1 receptor antagonist (IL1RN) SNPs (rs419598, rs315952 and rs9005) predicted Kellgren-Lawrence scores independently in each population. One IL1RN haplotype was associated with lower odds of radiographic severity (OR=0.15; 95% CI 0.065 to 0.349; p<0.0001), greater joint space width and lower synovial fluid cytokine levels. Carriage of the IL1RN haplotype influenced the age relationship with severity. CONCLUSION IL1RN polymorphisms reproducibly contribute to disease severity in knee OA and may be useful biomarkers for patient selection in disease-modifying OA drug trials.
Collapse
Affiliation(s)
- Mukundan Attur
- New York University Hospital for Joint Diseases, New York, USA
| | | | | | - Jack F Bukowski
- Interleukin Genetics, Waltham, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Nazneen Aziz
- Interleukin Genetics, Waltham, Massachusetts, USA
| | | | | | | | - Gary McDaniel
- Duke University Medical Center, Durham, North Carolina, USA
| | | | | |
Collapse
|
127
|
Moxley G, Meulenbelt I, Chapman K, van Diujn CM, Slagboom PE, Neale MC, Smith AJP, Carr AJ, Loughlin J. Interleukin-1 region meta-analysis with osteoarthritis phenotypes. Osteoarthritis Cartilage 2010; 18:200-7. [PMID: 19733643 DOI: 10.1016/j.joca.2009.08.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 07/24/2009] [Accepted: 08/24/2009] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Several research groups have examined osteoarthritis (OA) association with Interleukin-1 (IL-1) region markers and haplotypes. The results have been suggestive for hand OA, negative for knee OA, and conflicting for hip OA. DESIGN Our aim was to address conflicts employing meta-analytical methods on data from 1238 European-descent cases with various OA phenotypes and 1269 European-descent controls from four study centers. We imputed some missing genotype data and reconstructed IL-1 region extended haplotypes. A previously reported 7-marker IL1A-IL1B-IL1RN extended risk haplotype was tested for association with each specific index phenotype. RESULTS For hip OA, data from three centers showed heterogeneity of extended-risk-haplotype effect, two panels showing trend toward risk and another showing protection, with overall odds ratio (OR) 1.24 (95% Confidence interval (CI) 0.45-3.41, P 0.67). The heterogeneity fell partly along control ascertainment lines, chiefly between controls ascertained as spouses of arthroplasty patients and controls identified through population radiographic survey. For knee OA, the results showed no heterogeneity and no significant extended-risk-haplotype effect. For hand OA, the results showed little heterogeneity and a modest trend toward positive association (summary OR 1.34, 95% CI 0.83-2.17 P 0.23). Using a Bayesian partition modeling approach, the 7-marker extended haplotypes showed no significant effect on any OA phenotype examined. A 3-single-nucleotide polymorphism (SNP) IL1B-IL1RN haplotype rs1143627-rs16944-rs419598 showed a trend toward hand OA association (posterior probability of association 0.72) with the most prominent feature being protection from a specific haplotype representing a partial mirror image of the extended risk haplotype (OR estimated at 0.46). CONCLUSIONS The meta-analysis data do not confirm but only suggest that some hand and hip OA risk could be associated with the IL-1 region, particularly centered in IL1B and possibly also IL1RN.
Collapse
Affiliation(s)
- G Moxley
- Virginia Commonwealth University, Richmond, VA 23298-0263, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Osteoarthritis: can anti-cytokine therapy play a role in treatment? Clin Rheumatol 2010; 29:451-5. [PMID: 20108016 DOI: 10.1007/s10067-009-1352-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Revised: 10/13/2009] [Accepted: 12/17/2009] [Indexed: 10/19/2022]
Abstract
Osteoarthritis (OA) is the most common joint disorder worldwide, and it has an enormous socioeconomic impact both in the United States and throughout the world. The degree of articular inflammation is usually associated with the disease's progression, indicating that this process could contribute to articular damage. IL-1 beta and anti-TNF alpha are the two major cytokines players in the physiopathology of OA. Hence, we aimed to review the current literature on the effects of IL-1 and TNF-alpha neutralization as a new OA therapy. In vitro and experimental models showed a reduction in cartilage destruction with IL-1 inhibition therapy by IL-1 receptor antagonists (IL-1Ra). Despite this favorable evidence in animal models, studies on the inhibition of IL-1R in humans are still scarce. Although there is clear evidence that TNF-alpha plays a role in the pathophysiology of OA, only a few experimental trials have investigated the efficacy of blocking this pro-inflammatory cytokine in the treatment of OA. So far, the few studies available in humans using anti-TNF-alpha and IL-1 receptor antagonist are not remarkable, suggesting that further investigation and new therapeutic approaches are needed.
Collapse
|
129
|
Cucchiarini M, Heiligenstein S, Kohn D, Madry H. [Molecular tools to remodel osteoarthritic articular cartilage : growth, transcription, and signaling factors]. DER ORTHOPADE 2009; 38:1063-70. [PMID: 19876615 DOI: 10.1007/s00132-009-1495-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Osteoarthritis (OA) is a chronic disorder of the diarthrodial joints, mostly characterized by gradual deterioration of the articular cartilage. This disease still has no effective treatment. An emerging strategy for treating OA is based on molecular concepts using growth factors, transcription factors, and signaling molecules in light of their effects on the restoration of cartilage integrity. Recent studies have demonstrated that overexpression of such candidate molecules using direct gene transfer or ex vivo protocols is capable of stimulating cell proliferation and matrix synthesis in normal human and OA cartilage explants in vitro as well as in animal models in vivo. As a result, the structure of the articular cartilage can be improved. More insights into the pathophysiology of human OA and further studies in animal models are needed, however, to facilitate clinical translation of these molecular approaches. In conclusion, recent experimental findings permit cautious optimism, holding promise for treating human OA in the future.
Collapse
Affiliation(s)
- M Cucchiarini
- Labor für Experimentelle Orthopädie, Klinik für Orthopädie und Orthopädische Chirurgie, Universitätsklinikum des Saarlandes, Kirrbergerstrasse 37, 66421, Homburg/Saar, Deutschland.
| | | | | | | |
Collapse
|
130
|
|
131
|
Sanderson RO, Beata C, Flipo RM, Genevois JP, Macias C, Tacke S, Vezzoni A, Innes JF. Systematic review of the management of canine osteoarthritis. Vet Rec 2009; 164:418-24. [PMID: 19346540 DOI: 10.1136/vr.164.14.418] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
This review assesses the evidence for the efficacy of therapies used in the management of osteoarthritis in dogs on the basis of papers published in peer-reviewed journals in English between 1985 and July 2007. Sixty-eight papers were identified and evaluated. They considered four alternative therapies, one use of functional food, two intra-articular agents, six nutraceutical agents, 21 pharmacological agents, two physical therapies, three surgical techniques and two combinations of weight control. There was a high level of comfort (strong evidence) for the efficacy of carprofen, firocoxib and meloxicam, and a moderate level of comfort for the efficacy of etodolac in modifying the signs of osteoarthritis. There was a moderate level of comfort for the efficacy of glycosaminoglycan polysulphate, licofelone, elk velvet antler and a functional food containing green-lipped mussel for the modification of the structures involved in the disease. There was weak or no evidence in support of the use of doxycycline, electrostimulated acupuncture, extracorporeal shockwave therapy, gold wire acupuncture, hyaluronan, pentosan polysulphate, P54FP (extract of turmeric), tiaprofenic acid or tibial plateau levelling osteotomy.
Collapse
Affiliation(s)
- R O Sanderson
- Small Animal Teaching Hospital and Musculoskeletal Research Group, Faculty of Veterinary Science, University of Liverpool, Leahurst, Neston, Cheshire CH64 7TE
| | | | | | | | | | | | | | | |
Collapse
|
132
|
Scott I, Midha A, Rashid U, Ball S, Walding A, Kerry P, Delaney S, Cruwys S. Correlation of gene and mediator expression with clinical endpoints in an acute interleukin-1beta-driven model of joint pathology. Osteoarthritis Cartilage 2009; 17:790-7. [PMID: 19010066 DOI: 10.1016/j.joca.2008.09.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Accepted: 09/30/2008] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Interleukin-1beta (IL-1beta) plays a key role in the pathogenesis of chronic joint diseases, including osteoarthritis (OA), and drives a cascade of inflammatory and destructive responses within the synovial joint. Animal models of arthritis support the role of IL-1beta in joint pathology, however, the molecular changes downstream of IL-1beta are poorly understood in vivo. This study aimed to evaluate the intra-articular (i.a.) injection of IL-1beta in the rat joint as an acute model of joint disease and associate gene and mediator expression with clinical endpoints and pathological changes. METHODS The effects of i.a. administration of a pathologically relevant dose of IL-1beta on joint swelling, mechanical hyperalgesia, histopathology, gene expression and biochemical changes were measured from 2 to 24h. RESULTS IL-1beta-induced joint swelling and mechanical hyperalgesia. Gene expression analysis of joint tissue and biochemical analysis of joint lavage fluid identified pro-inflammatory and destructive mediators induced by IL-1beta. Histopathology of joint tissues showed evidence of synovitis and connective tissue inflammation, but not cartilage destruction. However, biochemical analysis of glucosaminoglycan levels in joint lavage fluids indicated cartilage breakdown and might be a sensitive marker of cartilage pathology. CONCLUSIONS Intra-articular injection of IL-1beta is a reproducible acute model of joint pathology that is potentially useful to evaluate IL-1 pathway inhibitors. The correlation of molecular events with clinical and pathological changes in this model has enhanced the understanding of the role of IL-1beta in joint disease. Methods developed for gene expression analysis using multi-gene microfluidics cards and for biochemical analysis of joint lavage fluid might have utility for characterisation of other arthritis models and corresponding human disease.
Collapse
Affiliation(s)
- I Scott
- Department of Molecular Biology, AstraZeneca Charnwood, Loughborough, UK.
| | | | | | | | | | | | | | | |
Collapse
|
133
|
Chevalier X, Goupille P, Beaulieu AD, Burch FX, Bensen WG, Conrozier T, Loeuille D, Kivitz AJ, Silver D, Appleton BE. Intraarticular injection of anakinra in osteoarthritis of the knee: a multicenter, randomized, double-blind, placebo-controlled study. ACTA ACUST UNITED AC 2009; 61:344-52. [PMID: 19248129 DOI: 10.1002/art.24096] [Citation(s) in RCA: 397] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To evaluate the clinical response, safety, and tolerability of a single intraarticular injection of anakinra in patients with symptomatic osteoarthritis (OA) of the knee. METHODS Patients with OA of the knee were enrolled in a multicenter, double-blind, placebo-controlled study and randomized 2:1:2 to receive a single intraarticular injection of placebo, anakinra 50 mg, or anakinra 150 mg in their symptomatic knee. Patients were evaluated for 12 weeks postinjection. The primary end point was the change in the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) score from baseline to week 4. Safety assessments included the evaluation of adverse events (AEs), laboratory tests, and vital signs. Pharmacokinetic parameters were assessed in a subset of patients. RESULTS Of 170 patients who enrolled, 160 (94%) completed the study. The mean improvements from baseline to week 4 in the WOMAC score were not statistically different between the placebo group and the patients who received 50 mg of anakinra (P = 0.67) or 150 mg of anakinra (P = 0.77). Anakinra was well tolerated. No withdrawals due to AEs or serious AEs, and no serious infections or deaths were reported. No clinically significant trends were noted in laboratory values or vital signs. Pharmacokinetic parameters demonstrated that the mean terminal half-life of anakinra in serum after intraarticular injection was approximately 4 hours. CONCLUSION Anakinra was well tolerated as a single 50-mg or 150-mg intraarticular injection in patients with OA of the knee. However, anakinra was not associated with improvements in OA symptoms compared with placebo.
Collapse
Affiliation(s)
- X Chevalier
- Department of Rheumatology, Hospital Henri Mondor, University Paris XII, Créteil, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Rutgers M, Saris DBF, Auw Yang KG, Dhert WJA, Creemers LB. Joint injury and osteoarthritis: soluble mediators in the course and treatment of cartilage pathology. Immunotherapy 2009; 1:435-45. [DOI: 10.2217/imt.09.14] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Osteoarthritis is a disabling disease of the aging generation, which results in loss of quality of life and increased healthcare costs. Cytokines appear to play an important role in the cartilaginous degeneration characterizing the pathological process. Increasing experience is being gained with cytokine-modulating therapies aimed at interfering with effects of chondrodegradative cytokines in the synovial fluid. Although in vitro and in vivo effectiveness of several of these therapies has been demonstrated, clinical effectiveness remains disputable, which may be related to the low levels of inflammatory cytokines found in osteoarthritic joints. By contrast, directly after joint trauma, which has been shown to predipose to early osteoarthritis, synovial fluid cytokine levels are strongly increased. Cytokine-modulating therapies, however, have hardly been considered for this indication. Increased knowledge of intra-articular soluble mediators correlating with cartilage pathology will lead to further development of cytokine-modulating products and, eventually, to effective inhibition of cartilage degeneration, in both the osteoarthritic as well as injured joints.
Collapse
Affiliation(s)
- Marijn Rutgers
- University Medical Center Utrecht, Department of Orthopaedics, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Daniël BF Saris
- University Medical Center Utrecht, Department of Orthopaedics, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Kiem Gie Auw Yang
- University Medical Center Utrecht, Department of Orthopaedics, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Wouter JA Dhert
- University Medical Center Utrecht, Department of Orthopaedics, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - Laura B Creemers
- University Medical Center Utrecht, Department of Orthopaedics, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
135
|
Baltzer AWA, Moser C, Jansen SA, Krauspe R. Autologous conditioned serum (Orthokine) is an effective treatment for knee osteoarthritis. Osteoarthritis Cartilage 2009; 17:152-60. [PMID: 18674932 DOI: 10.1016/j.joca.2008.06.014] [Citation(s) in RCA: 211] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Accepted: 06/18/2008] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is prevalent and difficult to treat. Autologous conditioned serum (ACS), marketed under the trade name Orthokine, is a novel, injectable antiarthritic derived from the patient's own blood. The present study is the first time ACS has undergone a controlled clinical trial. METHOD We investigated 376 patients with knee OA in a prospective, randomized, patient- and observer-blinded, placebo-controlled trial using an intention-to-treat analysis (ITT). The clinical effects of ACS were compared to hyaluronan (HA) and saline (placebo) as assessed by patient-administered outcome instruments (Western Ontario and McMaster Universities osteoarthritis index, global patient assessment, visual analog scale, Short-Form 8) after 7, 13 and 26 weeks. After 104 weeks an observer-blinded follow-up was carried out. Frequency and severity of adverse events were used as safety parameters. RESULTS In all treatment groups, intra-articular injections produced a reduction in symptoms as well as an improvement in quality of life. However, the effects of ACS were significantly superior to those of HA and saline for all outcome measures and time points, and improvements were clinically relevant; there were no differences between the effects of HA and saline. The frequency of adverse events was comparable in the ACS and saline groups, but higher in the HA group. CONCLUSION The data demonstrate that ACS injection considerably improves clinical signs and symptoms of OA. It remains to be determined whether ACS is disease-modifying, chondroprotective, or chondroregenerative.
Collapse
Affiliation(s)
- A W A Baltzer
- Centre for Molecular Orthopaedics, Düsseldorf, Germany
| | | | | | | |
Collapse
|
136
|
Evans CH, Ghivizzani SC, Robbins PD. Gene therapy of the rheumatic diseases: 1998 to 2008. Arthritis Res Ther 2009; 11:209. [PMID: 19232068 PMCID: PMC2688220 DOI: 10.1186/ar2563] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
During the decade since the launch of Arthritis Research, the application of gene therapy to the rheumatic diseases has experienced the same vicissitudes as the field of gene therapy as a whole. There have been conceptual and technological advances and an increase in the number of clinical trials. However, funding has been unreliable and a small number of high-profile deaths in human trials, including one in an arthritis gene therapy trial, have provided ammunition to skeptics. Nevertheless, steady progress has been made in a number of applications, including rheumatoid arthritis and osteoarthritis, Sjögren syndrome, and lupus. Clinical trials in rheumatoid arthritis have progressed to phase II and have provided the first glimpses of possible efficacy. Two phase I protocols for osteoarthritis are under way. Proof of principle has been demonstrated in animal models of Sjögren syndrome and lupus. For certain indications, the major technological barriers to the development of genetic therapies seem to have been largely overcome. The translational research necessary to turn these advances into effective genetic medicines requires sustained funding and continuity of effort.
Collapse
Affiliation(s)
- Christopher H Evans
- Center for Advanced Orthopaedic Studies, Harvard Medical School, BIDMC-RN115, 330 Brookline Avenue, Boston, MA 02215, USA.
| | | | | |
Collapse
|
137
|
Jouzeau JY, Moulin D, Koufany M, Sebillaud S, Bianchi A, Netter P. [Pathophysiological relevance of peroxisome proliferators activated receptors (PPAR) to joint diseases - the pro and con of agonists]. ACTA ACUST UNITED AC 2008; 202:289-312. [PMID: 19094928 DOI: 10.1051/jbio:2008034] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Peroxisome proliferators activated receptors (PPAR) are ligand-inducible nuclear transacting factors comprising three subtypes, PPARalpha, PPARbeta/delta and PPARgamma, which play a key role in lipids and glucose homeostasis. All PPAR subtypes have been identified in joint or inflammatory cells and their activation resulted in a transcriptional repression of pro-inflammatory cytokines (IL-1, TNFalpha), early inflammatory genes (NOS(2), COX-2, mPGES-1) or matrix metalloproteases (MMP-1, MMP-13), at least for the gamma subtype. PPAR full agonists were also shown to stimulate IL-1 receptor antagonist (IL-1Ra) production by cytokine-stimulated articular cells in a subtype-dependent manner. These anti-inflammatory and anti-catabolic properties were confirmed in animal models of joint diseases where PPAR agonists reduced synovial inflammation while preventing cartilage destruction or inflammatory bone loss, although many effects required much higher doses than needed to restore insulin sensitivity or to lower circulating lipid levels. However, these promising effects of PPAR full agonists were hampered by their ability to reduce the growth factor-dependent synthesis of extracellular matrix components or to induce chondrocyte apoptosis, by the possible contribution of immunosuppressive properties to their anti-arthritic effects, by the increased adipocyte differentiation secondary to prolonged stimulation of PPARgamma, and by a variable contribution of PPAR subtypes depending on the system. Clinical data are scarce in rheumatoid arthritis (RA) patients whereas thousands of patients worldwilde, treated with PPAR agonists for type 2 diabetes or dyslipidemia, are paradoxically prone to suffer from osteoarthritis (OA). Whereas high dosage of full agonists may expose RA patients to cardiovascular adverse effects, the proof of concept that PPAR agonists have therapeutical relevance to OA may benefit from an epidemiological follow-up of joint lesions in diabetic or hyperlipidemic patients treated for long periods of time with glitazones or fibrates. Additionally, cellular and animal studies are required to assess whether partial agonists of PPAR (SPPARMs) may preserve therapeutical properties with potentially less safety concern.
Collapse
|
138
|
Wehling P, Reinecke J, Baltzer AA, Granrath M, Schulitz KP, Schultz C, Krauspe R, Whiteside T, Elder E, Ghivizzani SC, Robbins PD, Evans CH. Clinical responses to gene therapy in joints of two subjects with rheumatoid arthritis. Hum Gene Ther 2008. [DOI: 10.1089/hgt.2008.075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
139
|
Ghivizzani SC, Gouze E, Gouze JN, Kay JD, Bush ML, Watson RS, Levings PP, Nickerson DM, Colahan PT, Robbins PD, Evans CH. Perspectives on the use of gene therapy for chronic joint diseases. Curr Gene Ther 2008; 8:273-86. [PMID: 18691023 DOI: 10.2174/156652308785160638] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Advances in molecular and cellular biology have identified a wide variety of proteins including targeted cytokine inhibitors, immunomodulatory proteins, cytotoxic mediators, angiogenesis inhibitors, and intracellular signalling molecules that could be of great benefit in the treatment of chronic joint diseases, such as osteo- and rheumatoid arthritis. Unfortunately, protein-based drugs are difficult to administer effectively. They have a high rate of turnover, requiring frequent readministration, and exposure in non-diseased tissue can lead to serious side effects. Gene transfer technologies offer methods to enhance the efficacy of protein-based therapies, enabling the body to produce these molecules locally at elevated levels for extended periods. The proof of concept of gene therapies for arthritis has been exhaustively demonstrated in multiple laboratories and in numerous animal models. This review attempts to condense these studies and to discuss the relative benefits and limitations of the methods proposed and to discuss the challenges toward translating these technologies into clinical realities.
Collapse
Affiliation(s)
- Steven C Ghivizzani
- Gene Therapy Laboratory, Department of Orthopaedics & Rehabilitation, University of Florida College of Medicine, Gainesville FL 32610, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Kadri A, Ea HK, Bazille C, Hannouche D, Lioté F, Cohen-Solal ME. Osteoprotegerin inhibits cartilage degradation through an effect on trabecular bone in murine experimental osteoarthritis. ACTA ACUST UNITED AC 2008; 58:2379-86. [PMID: 18668550 DOI: 10.1002/art.23638] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
OBJECTIVE To characterize bone microarchitectural changes and to test the hypothesis that disrupting local cytokine equilibrium could modify cartilage degradation in a murine model of experimental osteoarthritis (OA). METHODS Ten-week-old male C57BL/6 mice underwent medial meniscectomy of their right knees and a sham operation of their left knees. The mice received intraperitoneal injections of osteoprotegerin (OPG) (10 mg/kg), interleukin-1 receptor antagonist (IL-1Ra) (100 mg/kg), or phosphate buffered saline for 6 weeks. The microarchitecture of the trabecular bone, the OA score, and expression of ADAMTS-4 and ADAMTS-5 were assessed. Proteoglycan release was measured in cartilage explant cultures in the presence of IL-1Ra and OPG. RESULTS In the meniscectomized knees, bone volume/tissue volume (BV/TV) was lower, whereas trabecular separation, the OA score, and aggrecanase expression were higher than in the sham-operated knees. After treatment with OPG, BV/TV was significantly increased and trabecular separation was reduced in the knees that underwent meniscectomy. The OA score and the number of ADAMTS-positive cells were significantly decreased by treatment with OPG but were not affected by IL-1Ra. Moreover, OPG did not directly reduce the release of proteoglycans from cartilage explant cultures. CONCLUSION In an experimental model of OA, meniscectomy induced bone loss and cartilage degradation at 6 weeks. Systemic administration of OPG prevented bone and cartilage degradation in vivo but had no effect on cartilage in vitro. These data collectively indicate that bone could be a contributor in the early stages of OA pathogenesis. They further suggest that disruption of RANKL/OPG balance might result in the degradation of cartilage subjected to mechanical loading. Specific targeting of the bone cytokine network might help to prevent OA.
Collapse
Affiliation(s)
- A Kadri
- INSERM U606, Université Paris 7, Paris, France
| | | | | | | | | | | |
Collapse
|
141
|
Abstract
As the article in the current issue by Shinoda and colleagues shows, during the last two decades, there has been a dramatic increase in the understanding of basic biology behind chronic temporomandibular joint (TMJ) pain, inflammation and destruction. The involvement and contribution of cytokines to TMJ pain and inflammation must now be considered as established, evident and fundamental. Based on the present knowledge, it is now possible to design and investigate novel therapeutic strategies. These new and very encouraging approaches include manipulation of cytokine function, immune reactivity and the behaviour of inflammatory cells while maintaining the integrity of the affected tissue.
Collapse
|
142
|
Davies CM, Guilak F, Weinberg JB, Fermor B. Reactive nitrogen and oxygen species in interleukin-1-mediated DNA damage associated with osteoarthritis. Osteoarthritis Cartilage 2008; 16:624-30. [PMID: 17945515 PMCID: PMC2430155 DOI: 10.1016/j.joca.2007.09.012] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Accepted: 09/01/2007] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is associated with increased levels of reactive nitrogen and oxygen species and pro-inflammatory cytokines, such as interleukin-1 (IL-1). Nitric oxide (NO) can mediate a number of the catabolic effects of IL-1 in articular cartilage. The aims of this study were to determine if OA cartilage shows evidence of DNA damage, and if IL-1 could induce DNA damage in non-OA cartilage by increasing NO or superoxide. METHODS Articular chondrocytes were isolated from porcine femoral condyles and embedded in 1.2% alginate. The effects of 24h incubation with IL-1, the nitric oxide synthase 2 (NOS2)-selective inhibitor, the free radical scavenger superoxide dismutase (SOD), the NO donor NOC18, or the combined NO and peroxynitrite donor SIN-1 on DNA damage were tested, using the "comet" assay. NO production was measured using the Griess assay. The type of oxidative damage present was assessed using a modified comet assay. RESULTS OA cartilage had significantly more DNA damage than non-OA cartilage (P<0.001). IL-1 caused an increase in DNA damage (P<0.01), which was associated with increased NO production (P<0.01). Both oxidative DNA strand breaks and base modifications of purines and pyrimidines were observed. IL-1-induced DNA damage was inhibited by an NOS2 inhibitor or by SOD (P<0.01). Furthermore, NOC18 or SIN-1 caused DNA damage (P<0.001). CONCLUSION Our work shows chondrocytes in osteoarthritic cartilage exhibit DNA damage, and that IL-1 induces DNA damage and reactive oxygen and nitrogen species in non-OA chondrocytes in alginate.
Collapse
Affiliation(s)
- Catrin M. Davies
- Department of Surgery, Duke University Medical Center, Durham, NC 27710
| | - Farshid Guilak
- Department of Surgery, Duke University Medical Center, Durham, NC 27710
| | - J. Brice Weinberg
- Department of Medicine, VA and Duke Medical Centers, Durham, NC 27705
| | - Beverley Fermor
- Department of Surgery, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
143
|
Abstract
Once articular cartilage is injured, it has a very limited capacity for self repair. Although current surgical therapeutic procedures for cartilage repair are clinically useful, they cannot restore a normal articular surface. Current research offers a growing number of bioactive reagents, including proteins and nucleic acids, that may be used to augment various aspects of the repair process. As these agents are difficult to administer effectively, gene-transfer approaches are being developed to provide their sustained synthesis at sites of repair. To augment regeneration of articular cartilage, therapeutic genes can be delivered to the synovium or directly to the cartilage lesion. Gene delivery to the cells of the synovial lining is generally considered more suitable for chondroprotective approaches, based on the expression of anti-inflammatory mediators. Gene transfer targeted at cartilage defects can be achieved by either direct vector administration to cells located at or surrounding the defects, or by transplantation of genetically modified chondrogenic cells into the defect. Several studies have shown that exogenous cDNAs encoding growth factors can be delivered locally to sites of cartilage damage, where they are expressed at therapeutically relevant levels. Furthermore, data is beginning to emerge indicating that efficient delivery and expression of these genes is capable of influencing a repair response toward the synthesis of a more hyaline cartilage repair tissue in vivo. This review presents the current status of gene therapy for cartilage healing and highlights some of the remaining challenges.
Collapse
Affiliation(s)
- Andre F. Steinert
- Orthopaedic Center for Musculoskeletal Research König-Ludwig-Haus, Julius-Maximilians-University, Würzburg, Germany
| | - Ulrich Nöth
- Orthopaedic Center for Musculoskeletal Research König-Ludwig-Haus, Julius-Maximilians-University, Würzburg, Germany
| | - Rocky S. Tuan
- Cartilage Biology and Orthopaedics Branch National Institute of Arthritis, and Musculoskeletal and Skin Diseases National Institutes of Health, Department of Health and Human Services Bethesda, MD, U.S.A
| |
Collapse
|
144
|
Goupille P, Mulleman D, Chevalier X. Is interleukin-1 a good target for therapeutic intervention in intervertebral disc degeneration: lessons from the osteoarthritic experience. Arthritis Res Ther 2008; 9:110. [PMID: 18086327 PMCID: PMC2246235 DOI: 10.1186/ar2324] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
IL-1 plays a key role in disc degeneration and could be a valid target for inhibiting this process. IL-1 receptor antagonist (IL-1ra) might be a good candidate to inhibit IL-1 activity. However, many questions need to be addressed before contemplating therapy in humans. IL-1 blockade is also a great challenge in osteoarthritis and results from animal models suggest that IL-1ra may have beneficial effects. The clinical benefit of a local injection of IL-1ra in knee osteoarthritis may be limited by the antagonist's short half-life. Further studies with longer-lasting antagonists are needed to explore this new therapeutic approach.
Collapse
|
145
|
Shamji MF, Betre H, Kraus VB, Chen J, Chilkoti A, Pichika R, Masuda K, Setton LA. Development and characterization of a fusion protein between thermally responsive elastin-like polypeptide and interleukin-1 receptor antagonist: sustained release of a local antiinflammatory therapeutic. ACTA ACUST UNITED AC 2007; 56:3650-61. [PMID: 17968946 DOI: 10.1002/art.22952] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Interleukin-1 receptor antagonist (IL-1Ra) has been evaluated for the intraarticular treatment of osteoarthritis. Such administration of proteins may have limited utility because of their rapid clearance and short half-life in the joint. The fusion of a drug to elastin-like polypeptides (ELPs) promotes the formation of aggregating particles that form a "drug depot" at physiologic temperatures, a phenomenon intended to prolong the presence of the drug. The purpose of this study was to develop an injectable drug depot composed of IL-1Ra and ELP domains and to evaluate the properties and bioactivity of the recombinant ELP-IL-1Ra fusion protein. METHODS Fusion proteins between IL-1Ra and 2 distinct sequences and molecular weights of ELP were overexpressed in Escherichia coli. Environmental sensitivity was demonstrated by turbidity and dynamic light scattering as a function of temperature. IL-1Ra domain activity was evaluated by surface plasmon resonance, and in vitro antagonism of IL-1-mediated lymphocyte and thymocyte proliferation, as well as IL-1-induced tumor necrosis factor alpha (TNFalpha) expression and matrix metalloproteinase 3 (MMP-3) and ADAMTS-4 messenger RNA expression in human intervertebral disc fibrochondrocytes. IL-1Ra immunoreactivity was assessed before and after proteolytic degradation of the ELP partner. RESULTS Both fusion proteins underwent supramolecular aggregation at subphysiologic temperatures and slowly resolubilized at 37 degrees C. Interaction with IL-1 receptor was slower in association but equivalent in dissociation as compared with the commercial antagonist. Anti-IL-1 activity was demonstrated by inhibition of lymphocyte and thymocyte proliferation and by decreased TNFalpha expression and ADAMTS-4 and MMP-3 transcription by fibrochondrocytes. ELP domain proteolysis liberated a peptide of comparable size and immunoreactivity as the commercial IL-1Ra. This peptide was more bioactive against lymphocyte proliferation, nearly equivalent to the commercial antagonist. CONCLUSION The ELP-IL-1Ra fusion protein proved to retain the characteristic ELP inverse phase-transitioning behavior as well as the bioactivity of the IL-1Ra domain. This technology represents a novel drug carrier designed to prolong the presence of bioactive peptides following intraarticular delivery.
Collapse
|
146
|
Moxley G, Han J, Stern AG, Riley BP. Potential influence of IL1B haplotype and IL1A-IL1B-IL1RN extended haplotype on hand osteoarthritis risk. Osteoarthritis Cartilage 2007; 15:1106-12. [PMID: 17532232 DOI: 10.1016/j.joca.2007.03.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Accepted: 03/29/2007] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To assess osteoarthritis (OA) association with the human interleukin-1 (IL-1) region. DESIGN Sixty-four European-descent cases with radiographic hand OA and 48 European-descent controls were genotyped at nine single nucleotide polymorphism (SNP), one variable-number-of-tandem-repeat (VNTR), and one microsatellite marker extending across loci for IL-1alpha (IL1A), IL-1beta (IL1B), and IL-1 receptor antagonist (IL1RN). The genotype data were used to reconstruct individual locus haplotypes, and then locus haplotypes were used as superalleles for extended haplotype reconstruction. RESULTS Nine different extended IL1A-IL1B-IL1RN haplotypes occurred at a frequency 0.05 or greater in either cases or controls. Only two IL1A-IL1B-IL1RN extended haplotypes were consistent with previously described extended risk haplotypes and totaled n=9 in cases and n=3 in controls [odds ratio (OR) 2.1, Haldane's chi(2) 1.67, one-sided P 0.1]. Our prior report showed hand OA association with homozygous IL1B rs1143633 minor allele genotype. All except one extended risk haplotype copy also had the IL1B rs1143633 minor allele. The rs1143633 genotype association was explained by one common six-SNP IL1B haplotype bearing rs1143633 minor allele and also risk alleles at rs1143634, rs1143627, and rs16944, component markers of the previously described extended risk haplotypes. The IL1B haplotype bearing all three risk alleles was found in 16 haplotype-homozygous hand OA cases and in four haplotype-homozygous controls and conferred OR 3.4 among homozygotes (nominal P value 0.006). CONCLUSION Our evidence broadly supports the genetic association of OA phenotypes with an IL-1 region extended risk haplotype and specifically IL1B genotype. The extended risk haplotype previously associated with hip OA appears to be less frequent and has weaker genetic effect in hand OA. Hand OA risk is conferred by homozygous state for the IL1B haplotype characteristic of the extended risk haplotype.
Collapse
Affiliation(s)
- G Moxley
- Virginia Commonwealth University, Richmond, VA, USA.
| | | | | | | |
Collapse
|
147
|
Trippel S, Cucchiarini M, Madry H, Shi S, Wang C. Gene therapy for articular cartilage repair. Proc Inst Mech Eng H 2007; 221:451-9. [PMID: 17822147 DOI: 10.1243/09544119jeim237] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Articular cartilage serves as the gliding surface of joints. It is susceptible to damage from trauma and from degenerative diseases. Restoration of damaged articular cartilage may be achievable through the use of cell-regulatory molecules that augment the reparative activities of the cells, inhibit the cells' degradative activities, or both. A variety of such molecules have been identified. These include insulin-like growth factor I, fibroblast growth factor 2, bone morphogenetic proteins 2, 4, and 7, and interleukin-1 receptor antagonist. It is now possible to transfer the genes encoding such molecules into articular cartilage and synovial lining cells. Although preliminary, data from in-vitro and in-vivo studies suggest that gene therapy can deliver such potentially therapeutic agents to protect existing cartilage and to build new cartilage.
Collapse
Affiliation(s)
- S Trippel
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 541 Clinical Drive, Suite 600, Indianapolis, IP 46202-5111, USA.
| | | | | | | | | |
Collapse
|
148
|
Abstract
Osteoarthritis (OA) is the most common form of arthritis and one of the leading causes of disability worldwide. The incidence of OA increases with age, and as longevity increases, it will cause a significant socioeconomic burden. No disease-modifying therapy is available for OA. Recent research has highlighted the role of inflammation in the progression of OA. Interleukin-1 appears to have a significant role in disease progression. Since the interleukin-1 receptor antagonist (IL-1Ra) anakinra has been used successfully in the treatment of the inflammation and bone destruction of rheumatoid arthritis, some have suggested that it may be able to retard the disease progression of OA. This article reviews the data on the use of anakinra in OA treatment.
Collapse
|
149
|
Frisbie DD, Kawcak CE, Werpy NM, Park RD, McIlwraith CW. Clinical, biochemical, and histologic effects of intra-articular administration of autologous conditioned serum in horses with experimentally induced osteoarthritis. Am J Vet Res 2007; 68:290-6. [PMID: 17331019 DOI: 10.2460/ajvr.68.3.290] [Citation(s) in RCA: 170] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To assess the clinical, biochemical, and histologic effects of intra-articular administration of autologous conditioned serum (ACS) in the treatment of experimentally induced osteoarthritis in horses. ANIMALS 16 horses. PROCEDURES Osteoarthritis was induced arthroscopically in 1 middle carpal joint of all horses. In 8 placebo- and 8 ACS-treated horses, 6 mL of PBS solution or 6 mL of ACS was injected into the osteoarthritis-affected joint on days 14, 21, 28, and 35, respectively; PBS solution was administered in the other sham-operated joints. Evaluations included clinical assessment of lameness and synovial fluid analysis (performed biweekly); gross pathologic and histologic examinations of cartilage and synovial membrane samples were performed at necropsy. RESULTS No adverse treatment-related events were detected. Horses that were treated with ACS had significant clinical improvement in lameness, unlike the placebo-treated horses. Among the osteoarthritis-affected joints, ACS treatment significantly decreased synovial membrane hyperplasia, compared with placebo-treated joints; although not significant, the ACS-treated joints also appeared to have less gross cartilage fibrillation and synovial membrane hemorrhage. The synovial fluid concentration of interleukin-1 receptor antagonist (assessed by use of mouse anti-interleukin-1 receptor antagonist antibody) was increased following treatment with ACS. CONCLUSIONS AND CLINICAL RELEVANCE Results of this controlled study indicated that there was significant clinical and histologic improvement in osteoarthritis-affected joints of horses following treatment with ACS, compared with placebo treatment. On the basis of these findings, further controlled clinical trials to assess this treatment are warranted, and investigation of the mechanisms of action of ACS should be pursued concurrently.
Collapse
Affiliation(s)
- David D Frisbie
- Equine Orthopaedic Research Center, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | | | | | | | | |
Collapse
|
150
|
Maccoux LJ, Salway F, Day PJR, Clements DN. Expression profiling of select cytokines in canine osteoarthritis tissues. Vet Immunol Immunopathol 2007; 118:59-67. [PMID: 17524496 DOI: 10.1016/j.vetimm.2007.04.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Revised: 03/23/2007] [Accepted: 04/12/2007] [Indexed: 12/13/2022]
Abstract
The objective of this study was to investigate the level of expression of five cytokines in four different articular tissues from the joints of dogs with and without osteoarthritis (OA). Articular tissues were harvested from the stifle (fat, cranial cruciate ligament, synovial membrane) or hip (articular cartilage) from eight dogs with OA secondary to cranial cruciate ligament disease (stifle) or hip dysplasia (hip), undergoing routine surgical treatment for the condition, and from five dogs euthanatized without orthopaedic disease. The mRNA transcript numbers for interleukin-1beta (IL-1beta), interleukin-6 (IL-6), interleukin-8 (IL-8), interleukin-10 (IL-10) and interleukin-17 (IL-17) were determined by quantitative real-time reverse transcription polymerase chain reaction (RT-qPCR). Increased expression of IL-1beta, IL-6 and IL-10 in OA synovial membrane, increased expression of IL-1beta and IL-6 in ruptured (OA) ligament, and reduced expression of IL-8 in OA synovial membrane were identified. Cytokine expression was detected in multiple tissues within the articular joint, but differential expression in OA was detected primarily in the synovial membrane and cranial cruciate ligament.
Collapse
Affiliation(s)
- Lindsey J Maccoux
- Centre for Integrated Genomic Medical Research, University of Manchester, The Stopford Building, Oxford Road, Manchester, UK
| | | | | | | |
Collapse
|