1
|
The effect of NSAIDs on postfracture bone healing: a meta-analysis of randomized controlled trials. OTA Int 2021; 4:e092. [PMID: 34746650 PMCID: PMC8568409 DOI: 10.1097/oi9.0000000000000092] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/18/2020] [Accepted: 08/12/2020] [Indexed: 01/08/2023]
Abstract
To determine whether nonsteroidal anti-inflammatory drugs (NSAIDs) have an adverse effect on bone healing by evaluating all available human randomized controlled trials (RCTs) on this subject. Data Sources A comprehensive search of electronic databases (PubMed, MEDLINE, and Cross-References) until October 2018 comparing the occurrence of nonunion in patients who received NSAIDs to the control group through RCTs. Study Selection Inclusion criteria were English-only studies, and the type of studies was restricted to RCTs. Data Extraction Two authors independently extracted data from the selected studies, and the data collected were compared to verify agreement. Data Synthesis Nonunion was the main outcome evaluated in each study. Regression analysis was used to estimate the relative risk comparing the duration and the type of NSAIDs by calculating the odds ratio (OR) for dichotomous variables. Studies were weighed by the inverse of the variance of the outcome, and a fixed-effects model was used for all analyses. Conclusions Six RCTs (609 patients) were included. The risk of nonunion was higher in the patients who were given NSAIDs after the fracture with an OR of 3.47. However, once the studies were categorized into the duration of treatment with NSAIDs, those who received NSAIDs for a short period (<2 weeks) did not show any significant risk of nonunion compared to those who received NSAIDs for a long period (>4 weeks). Indomethacin was associated with a significant higher nonunion rate and OR ranging from 1.66 to 9.03 compared with other NSAIDs that did not show a significant nonunion risk.
Collapse
|
2
|
Effect of NSAID Use on Bone Healing in Pediatric Fractures: A Preliminary, Prospective, Randomized, Blinded Study. J Pediatr Orthop 2020; 40:e683-e689. [PMID: 32555047 DOI: 10.1097/bpo.0000000000001603] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND This study aimed to investigate if nonsteroidal anti-inflammatory drugs (NSAIDs) used in the acute phase of bone healing in children with fractures result in delayed union or nonunion as compared with patients who do not take NSAIDs for pain control during this same time period. METHODS In this prospective, randomized, parallel, single-blinded study, skeletally immature patients with long bone fractures were randomized to 1 of 2 groups for their postfracture pain management. The NSAID group was prescribed weight-based ibuprofen, whereas the control group was not allowed any NSAID medication and instead prescribed weight-based acetaminophen. Both groups were allowed to use oxycodone for breakthrough pain. The primary outcome was fracture healing assessed at 2, 6, and 10 weeks. RESULTS One-hundred-two patients were enrolled between February 6, 2014 and September 23, 2016. Ninety-five patients (with 97 fractures) completed a 6-month follow-up (46 patients with 47 fractures in the control group and 49 patients 50 fractures in the NSAID group). None achieved healing at 1 to 2 weeks. By 6 weeks, 37 of 45 patients (82%) of control group and 46 out of 50 patients (92%) of ibuprofen group had healed fractures (P=0.22). At 10 to 12 week follow-up, 46 (98%) of the control group fractures were healed and 50 (100%) of the ibuprofen group fractures were healed. All were healed by 6 months. Healing was documented at a mean of 40 days in the control group and 31 days in the ibuprofen group (P=0.76). The mean number of days breakthrough oxycodone was used was 2.4 days in the control group and 1.9 days in the NSAID group (P=0.48). CONCLUSION Ibuprofen is an effective medication for fracture pain in children and its use does not impair clinical or radiographic long bone fracture healing in skeletally immature patients. LEVEL OF EVIDENCE Level I-therapeutic.
Collapse
|
3
|
Ali MU, Usman M, Patel K. Effects of NSAID use on bone healing: A meta-analysis of retrospective case–control and cohort studies within clinical settings. TRAUMA-ENGLAND 2019. [DOI: 10.1177/1460408619886211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Introduction This meta-analysis aims to determine whether non-steroidal anti-inflammatory drug (NSAID) use is significantly associated with adverse bone healing outcomes within clinical settings, including trauma and elective spine settings. It will also explore bone healing outcomes with the type, route, dosage and duration of NSAID exposure and aims to demonstrate the effects of various other confounding factors on bone healing outcomes. Methods Electronic databases including MedLine, Embase and Cochrane were searched from January 1975 to December 2017. A distinct analysis of observational long bone and spine studies in adult populations was performed, assessing the effects of NSAID exposure on bone healing. Studies from paediatric population and randomised trials also underwent separate assessment in the synthesis. Meta-analysis was conducted in compliance with QUORUM and PRISMA guidelines. Results Quantitative assessment of observational studies suggested a significant risk of bone healing complications with NSAID exposure across each clinical group: long bone (p = 0.0004) and spine (p = 0.02). Analysis of paediatric studies revealed a non-significant association of poor outcomes with NSAID use (p = 0.36), while assessment of randomised trials demonstrated a statistically significant risk of complications following NSAID administration (p = 0.04). Meta-regression further suggested smoking to be a substantial confounder associated with adverse bone healing outcomes (p < 0.00001). Conclusion Inclusion of only fair and moderate-quality retrospective cohort and case–control studies in the current synthesis limits the clinical application of its findings and therefore warrants the need for further research. Thus, attempts to conduct high-quality prospective cohort and randomised trials to study the effects of NSAID use on bone healing would be very helpful and will provide a basis for more extensive research in future.
Collapse
Affiliation(s)
| | - Mehvish Usman
- Trauma and Orthopaedics, Manchester Royal Infirmary, Manchester, UK
| | - Kuntal Patel
- Trauma and Orthopaedics, Royal Lancaster Infirmary, Lancaster, UK
| |
Collapse
|
4
|
Claus CF, Lytle E, Tong D, Sigler D, Lago D, Bahoura M, Dosanjh A, Lawless M, Slavnic D, Kelkar P, Houseman C, Bono P, Richards B, Soo TM. The effect of ketorolac on posterior thoracolumbar spinal fusions: a prospective double-blinded randomised placebo-controlled trial protocol. BMJ Open 2019; 9:e025855. [PMID: 30670528 PMCID: PMC6347897 DOI: 10.1136/bmjopen-2018-025855] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Ketorolac has been shown to provide quality postoperative pain control and decrease opioid requirement with minimal side effects following spinal surgery. However, the literature addressing its use in spinal fusions is highly variable in both its effectiveness and complications, such as pseudarthrosis. Recent literature postulates that ketorolac may not affect fusion rates and large randomised controlled trials are needed to demonstrate ketorolac as a safe and effective adjuvant treatment to opioids for postoperative pain control. METHODS AND ANALYSIS This is a multihospital, prospective, double-blinded, randomised placebo-controlled trial. Data concerning fusion rates, postoperative opioid use, pain scores, length of stay will be recorded with the aim of demonstrating that the use of ketorolac does not decrease thoracolumbar spinal fusion rates while identifying possible adverse events related to short-term minimal effective dose compared with placebo. Additionally, this investigation aims to demonstrate a decrease in postoperative opioid use demonstrated by a decrease in morphine equivalence while showing equivalent postoperative pain control and decrease the average length of stay. ETHICS AND DISSEMINATION Ethical approval was obtained at all participating hospitals by the institutional review board. The results of this study will be submitted for publication in peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT03278691.
Collapse
Affiliation(s)
- Chad F Claus
- Division of Neurosurgery, Department of Surgery, Ascension Providence Hospital, Michigan State University, College of Human Medicine, Southfield, Michigan, USA
| | - Evan Lytle
- Division of Neurosurgery, Department of Surgery, Ascension Providence Hospital, Michigan State University, College of Human Medicine, Southfield, Michigan, USA
| | - Doris Tong
- Division of Neurosurgery, Department of Surgery, Ascension Providence Hospital, Michigan State University, College of Human Medicine, Southfield, Michigan, USA
| | - Diana Sigler
- Department of Pharmacy, Ascension Providence Hospital, Southfield, Michigan, USA
| | - Dominick Lago
- Division of Neuro-anesthesia, Department of Anesthesia, Ascension Providence Hospital, Michigan State University, College of Human Medicine, Southfield, Michigan, USA
| | - Matthew Bahoura
- Division of Neurosurgery, Department of Surgery, Ascension Providence Hospital, Michigan State University, College of Human Medicine, Southfield, Michigan, USA
| | - Amarpal Dosanjh
- Division of Neurosurgery, Department of Surgery, Ascension Providence Hospital, Michigan State University, College of Human Medicine, Southfield, Michigan, USA
| | - Michael Lawless
- Division of Neurosurgery, Department of Surgery, Ascension Providence Hospital, Michigan State University, College of Human Medicine, Southfield, Michigan, USA
| | - Dejan Slavnic
- Division of Neurosurgery, Department of Surgery, Ascension Providence Hospital, Michigan State University, College of Human Medicine, Southfield, Michigan, USA
| | - Prashant Kelkar
- Division of Neurosurgery, Department of Surgery, Ascension Providence Hospital, Michigan State University, College of Human Medicine, Southfield, Michigan, USA
| | - Clifford Houseman
- Division of Neurosurgery, Department of Surgery, Ascension Providence Hospital, Michigan State University, College of Human Medicine, Southfield, Michigan, USA
| | - Peter Bono
- Division of Neurosurgery, Department of Surgery, Ascension Providence Hospital, Michigan State University, College of Human Medicine, Southfield, Michigan, USA
| | - Boyd Richards
- Division of Neurosurgery, Department of Surgery, Ascension Providence Hospital, Michigan State University, College of Human Medicine, Southfield, Michigan, USA
| | - Teck M Soo
- Division of Neurosurgery, Department of Surgery, Ascension Providence Hospital, Michigan State University, College of Human Medicine, Southfield, Michigan, USA
| |
Collapse
|
5
|
Bahney CS, Zondervan RL, Allison P, Theologis A, Ashley JW, Ahn J, Miclau T, Marcucio RS, Hankenson KD. Cellular biology of fracture healing. J Orthop Res 2019; 37:35-50. [PMID: 30370699 PMCID: PMC6542569 DOI: 10.1002/jor.24170] [Citation(s) in RCA: 286] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/27/2018] [Indexed: 02/04/2023]
Abstract
The biology of bone healing is a rapidly developing science. Advances in transgenic and gene-targeted mice have enabled tissue and cell-specific investigations of skeletal regeneration. As an example, only recently has it been recognized that chondrocytes convert to osteoblasts during healing bone, and only several years prior, seminal publications reported definitively that the primary tissues contributing bone forming cells during regeneration were the periosteum and endosteum. While genetically modified animals offer incredible insights into the temporal and spatial importance of various gene products, the complexity and rapidity of healing-coupled with the heterogeneity of animal models-renders studies of regenerative biology challenging. Herein, cells that play a key role in bone healing will be reviewed and extracellular mediators regulating their behavior discussed. We will focus on recent studies that explore novel roles of inflammation in bone healing, and the origins and fates of various cells in the fracture environment. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Chelsea S. Bahney
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Robert L. Zondervan
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| | - Patrick Allison
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan
| | - Alekos Theologis
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Jason W. Ashley
- Department of Biology, Eastern Washington University, Cheney, Washington
| | - Jaimo Ahn
- Department of Biology, Eastern Washington University, Cheney, Washington
| | - Theodore Miclau
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Ralph S. Marcucio
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
6
|
Wei F, Xiao Y. Modulation of the Osteoimmune Environment in the Development of Biomaterials for Osteogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1077:69-86. [DOI: 10.1007/978-981-13-0947-2_5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
7
|
Riggin CN, Qu F, Kim DH, Huegel J, Steinberg DR, Kuntz AF, Soslowsky LJ, Mauck RL, Bernstein J. Electrospun PLGA Nanofiber Scaffolds Release Ibuprofen Faster and Degrade Slower After In Vivo Implantation. Ann Biomed Eng 2017; 45:2348-2359. [PMID: 28653294 DOI: 10.1007/s10439-017-1876-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 06/15/2017] [Indexed: 01/04/2023]
Abstract
While delayed delivery of non-steroidal anti-inflammatory drugs (NSAIDs) has been associated with improved tendon healing, early delivery has been associated with impaired healing. Therefore, NSAID use is appropriate only if the dose, timing, and mode of delivery relieves pain but does not impede tissue repair. Because delivery parameters can be controlled using drug-eluting nanofibrous scaffolds, our objective was to develop a scaffold for local controlled release of ibuprofen (IBP), and characterize the release profile and degradation both in vitro and in vivo. We found that when incubated in vitro in saline, scaffolds containing IBP had a linear release profile. However, when implanted subcutaneously in vivo or when incubated in vitro in serum, scaffolds showed a rapid burst release. These data demonstrate that scaffold properties are dependent on the environment in which they are placed and the importance of using serum, rather than saline, for initial in vitro evaluation of biofactor release from biodegradable scaffolds.
Collapse
Affiliation(s)
- Corinne N Riggin
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA.,Department of Bioengineering, University of Pennsylvania, Suite 240 Skirkanich Hall, 210 South 33rd Street, Philadelphia, PA, 19104, USA
| | - Feini Qu
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA.,School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA, 19104, USA
| | - Dong Hwa Kim
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA
| | - Julianne Huegel
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA
| | - David R Steinberg
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA
| | - Andrew F Kuntz
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA
| | - Louis J Soslowsky
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA.,Department of Bioengineering, University of Pennsylvania, Suite 240 Skirkanich Hall, 210 South 33rd Street, Philadelphia, PA, 19104, USA
| | - Robert L Mauck
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA.,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA.,Department of Bioengineering, University of Pennsylvania, Suite 240 Skirkanich Hall, 210 South 33rd Street, Philadelphia, PA, 19104, USA
| | - Joseph Bernstein
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 University & Woodland Avenue, Philadelphia, PA, 19104, USA. .,McKay Orthopaedic Research Lab, University of Pennsylvania, 424 Stemmler Hall, 36th Street & Hamilton Walk, Philadelphia, PA, 19104, USA.
| |
Collapse
|
8
|
Chen Z, Ni S, Han S, Crawford R, Lu S, Wei F, Chang J, Wu C, Xiao Y. Nanoporous microstructures mediate osteogenesis by modulating the osteo-immune response of macrophages. NANOSCALE 2017; 9:706-718. [PMID: 27959374 DOI: 10.1039/c6nr06421c] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The osteoimmune environment plays indispensable roles in bone regeneration because the early immune environment that exists during the regenerative process promotes the recruitment and differentiation of osteoblastic lineage cells. The response of immune cells growing on nanotopographic surfaces and the microenvironment they generate should be considered when evaluating nanotopography-mediated osteogenesis, which are topics that are generally neglected in the field. In this study, we investigated the modulatory effects of nanoporous anodic alumina with different sized pores on macrophage responses and their subsequent effects on the osteogenic differentiation of bone marrow stromal cells (BMSCs). The nanopore structure and the pore size were found to be important adhesive cues for macrophages, which affected their spreading and cell shape, subsequently regulated the expression and activation of autophagy pathway components (LC3A/B, Beclin-1, Atg3, Atg7, and P62) and modulated the inflammatory response, osteoclastic activities, and release of osteogenic factors. Subsequently, the osteogenic pathways (Wnt and BMP) of BMSCs were found to be regulated by different nanopore-induced inflammatory environments, which affected the osteogenic differentiation outcomes. This study is the first to emphasize the effects of immune cells on nanotopography-mediated osteogenesis, which could lead to a new strategy for the development of advanced nanobiomaterials for tissue engineering, nanomedicine and immunotherapeutic applications.
Collapse
Affiliation(s)
- Zetao Chen
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, Guangdong, People's Republic of China and Institute of Health and Biomedical Innovation & the Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane 4059, Australia.
| | - Siyu Ni
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University Shanghai, 201620, People's Republic of China
| | - Shengwei Han
- Institute of Health and Biomedical Innovation & the Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane 4059, Australia.
| | - Ross Crawford
- Institute of Health and Biomedical Innovation & the Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane 4059, Australia.
| | - Shifeier Lu
- Institute of Health and Biomedical Innovation & the Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane 4059, Australia.
| | - Fei Wei
- Institute of Health and Biomedical Innovation & the Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane 4059, Australia.
| | - Jiang Chang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, People's Republic of China.
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, People's Republic of China.
| | - Yin Xiao
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, Guangdong, People's Republic of China and Institute of Health and Biomedical Innovation & the Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane 4059, Australia.
| |
Collapse
|
9
|
Goodman SB, Ma T, Genovese M, Lane Smith R. Cox-2 Selective Inhibitors and Bone. Int J Immunopathol Pharmacol 2016; 16:201-5. [PMID: 14611721 DOI: 10.1177/039463200301600303] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are widely prescribed medications for relief of pain and inflammation. Recent animal studies using models of fracture healing and bone ingrowth suggest that NSAIDs (both non-selective NSAIDs and selective COX-2 inhibitors) adversely affect these bone-related processes. The dose and time-relationships of these medications and their resulting effects on bone have not yet been fully elucidated. Furthermore, whether COX-2 inhibitors and non-selective NSAIDs lead to clinically relevant adverse effects on bone healing in humans is unknown.
Collapse
Affiliation(s)
- S B Goodman
- Dept Orthopaedic Surgery and Div Immunol Rheumatol, Stanford University Medical Center, Stanford, CA 94305, USA.
| | | | | | | |
Collapse
|
10
|
Chan JK, Glass GE, Ersek A, Freidin A, Williams GA, Gowers K, Espirito Santo AI, Jeffery R, Otto WR, Poulsom R, Feldmann M, Rankin SM, Horwood NJ, Nanchahal J. Low-dose TNF augments fracture healing in normal and osteoporotic bone by up-regulating the innate immune response. EMBO Mol Med 2016; 7:547-61. [PMID: 25770819 PMCID: PMC4492816 DOI: 10.15252/emmm.201404487] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The mechanism by which trauma initiates healing remains unclear. Precise understanding of these events may define interventions for accelerating healing that could be translated to the clinical arena. We previously reported that addition of low-dose recombinant human TNF (rhTNF) at the fracture site augmented fracture repair in a murine tibial fracture model. Here, we show that local rhTNF treatment is only effective when administered within 24 h of injury, when neutrophils are the major inflammatory cell infiltrate. Systemic administration of anti-TNF impaired fracture healing. Addition of rhTNF enhanced neutrophil recruitment and promoted recruitment of monocytes through CCL2 production. Conversely, depletion of neutrophils or inhibition of the chemokine receptor CCR2 resulted in significantly impaired fracture healing. Fragility, or osteoporotic, fractures represent a major medical problem as they are associated with permanent disability and premature death. Using a murine model of fragility fractures, we found that local rhTNF treatment improved fracture healing during the early phase of repair. If translated clinically, this promotion of fracture healing would reduce the morbidity and mortality associated with delayed patient mobilization.
Collapse
Affiliation(s)
- James K Chan
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Graeme E Glass
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Adel Ersek
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Andrew Freidin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Garry A Williams
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Kate Gowers
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Rosemary Jeffery
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - William R Otto
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - Richard Poulsom
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - Marc Feldmann
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Sara M Rankin
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Nicole J Horwood
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | |
Collapse
|
11
|
Fong K, Truong V, Foote CJ, Petrisor B, Williams D, Ristevski B, Sprague S, Bhandari M. Predictors of nonunion and reoperation in patients with fractures of the tibia: an observational study. BMC Musculoskelet Disord 2013; 14:103. [PMID: 23517574 PMCID: PMC3614478 DOI: 10.1186/1471-2474-14-103] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 03/15/2013] [Indexed: 11/10/2022] Open
Abstract
Background Tibial shaft fractures are the most common long bone fracture and are prone to complications such as nonunion requiring reoperations to promote fracture healing. We aimed to determine the fracture characteristics associated with tibial fracture nonunion, and their predictive value on the need for reoperation. We further aimed to evaluate the predictive value of a previously-developed prognostic index of three fracture characteristics on nonunion and reoperation rate. Methods We conducted an observational study and developed a risk factor list from previous literature and key informants in the field of orthopaedic surgery, as well as via a sample-to-redundancy strategy. We evaluated 22 potential risk factors for the development of tibial fracture nonunion in 200 tibial fractures. We also evaluated the predictive value of a previously-identified prognostic risk index on secondary intervention and/or reoperation rate. Two individuals independently extracted the data from 200 patient electronic medical records. An independent reviewer assessed the initial x-ray, the post-operative x-ray, and all available sequential x-rays. Regression and chi-square analysis was used to evaluate potential associations. Results In our cohort of patients, 37 (18.5%) had a nonunion and 27 (13.5%) underwent a reoperation. Patients with a nonunion were 97 times (95% CI 25.8-366.5) more likely to have a reoperation. Multivariable logistic regression revealed that fractures with less than 25% cortical continuity were predictive of nonunion (odds ratio = 4.72; p = 0.02). Such fractures also accounted for all of the reoperations identified in our sample. Furthermore, our data provided preliminary validation of a previous risk index predictive of reoperation that includes the presence of a fracture gap post-fixation, open fracture, and transverse fracture type as variables, with an aggregate of fracture gap and an open fracture yielding patients with the highest risk of developing a nonunion. Conclusions We identified a significant association between degree of cortical continuity and the development of a nonunion and risk for reoperation in tibial shaft fractures. In addition, our study supports the predictive value of a previous prognostic index, which inform discussion of prognosis following operative management of tibial fractures.
Collapse
Affiliation(s)
- Katie Fong
- Division of Orthopaedic Surgery, McMaster University, Hamilton, Canada
| | | | | | | | | | | | | | | |
Collapse
|
12
|
The effect of nonsteroidal anti-inflammatory drugs on tissue healing. Knee Surg Sports Traumatol Arthrosc 2013; 21:540-9. [PMID: 22744434 DOI: 10.1007/s00167-012-2095-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 06/04/2012] [Indexed: 01/29/2023]
Abstract
PURPOSE Non-selective (NSAIDs) and selective (COX-2) nonsteroidal anti-inflammatory drugs are commonly used for their analgesic and anti-inflammatory effects. Their role after orthopaedic surgery has been infrequently described and remains controversial because of unclear effects on soft tissue and bone healing. This study critically reviews the available literature to describe the effects of NSAIDs and COX-2 inhibitors on soft tissue and bone healing. METHODS A Medline search was performed using NSAIDs or COX-2 inhibitors and tissue healing. The combined search yielded 637 articles. Following exclusion, 44 articles were deemed relevant with 9 articles on soft tissue healing and 35 articles on bone healing. The available evidence is based primarily on animal data (39 studies), with considerable variation in study methods. RESULTS In regard to soft tissue healing, there is insufficient evidence of a detrimental effect when using either NSAIDs or COX-2 inhibitors at standard doses for ≤2 weeks. For soft tissue to bone healing, a limited number of studies demonstrate impairment in healing. However, with respect to bone healing, indomethacin appears to have a clear detrimental effect, with less substantial evidence for other NSAIDs. CONCLUSIONS Short-term, low-dose use of NSAIDs and COX-2 inhibitors does not appear to have a detrimental effect following soft tissue injury, but is inhibitory in cases involving bony healing. However, additional well-controlled human studies are necessary to draw more definitive conclusions regarding their role. Clinically, the prudent use of anti-inflammatory medications following sports medicine injuries and surgeries appears to be a reasonable option in clinical practice unless bone healing is required. LEVEL OF EVIDENCE III.
Collapse
|
13
|
Schemitsch EH, Bhandari M, Guyatt G, Sanders DW, Swiontkowski M, Tornetta P, Walter SD, Zdero R, Goslings JC, Teague D, Jeray K, McKee MD. Prognostic factors for predicting outcomes after intramedullary nailing of the tibia. J Bone Joint Surg Am 2012; 94:1786-93. [PMID: 23032589 PMCID: PMC3448300 DOI: 10.2106/jbjs.j.01418] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Prediction of negative postoperative outcomes after long-bone fracture treatment may help to optimize patient care. We recently completed the Study to Prospectively Evaluate Reamed Intramedullary Nails in Patients with Tibial Fractures (SPRINT), a large, multicenter trial of reamed and unreamed intramedullary nailing of tibial shaft fractures in 1226 patients. Using the SPRINT data, we conducted an investigation of baseline and surgical factors to determine any associations with an increased risk of adverse events within one year of intramedullary nailing. METHODS Using multivariable logistic regression analysis, we investigated fifteen baseline and surgical factors for any associations with an increased risk of negative outcomes. RESULTS There was an increased risk of negative events in patients with a high-energy mechanism of injury (odds ratio [OR] = 1.57; 95% confidence interval [CI], 1.05 to 2.35), a stainless steel compared with a titanium nail (OR = 1.52; 95% CI, 1.10 to 2.13), a fracture gap (OR = 2.40; 95% CI, 1.47 to 3.94), and full weight-bearing status after surgery (OR = 1.63; 95% CI, 1.00 to 2.64). There was no increased risk with the use of nonsteroidal anti-inflammatory agents, late or early time to surgery, or smoking status. Open fractures had a higher risk of events among patients treated with reamed nailing (OR = 3.26; 95% CI, 2.01 to 5.28) but not in patients treated with unreamed nailing (OR = 1.50; 95% CI, 0.92 to 2.47). Patients with open fractures who had wound management either without any additional procedures or with delayed primary closure had a decreased risk of events compared with patients who required subsequent, more complex reconstruction (OR = 0.18 [95% CI, 0.09 to 0.35] and 0.29 [95% CI, 0.14 to 0.62], respectively). CONCLUSIONS We identified several baseline fracture and surgical characteristics that may increase the risk of adverse events in patients with tibial shaft fractures. Surgeons should consider the predictors identified in our analysis to inform patients treated for tibial shaft fractures. LEVEL OF EVIDENCE Prognostic Level II. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Emil H Schemitsch
- Division of Orthopaedics, Department of Surgery, University of Toronto, St. Michael's Hospital, Suite 800, 55 Queen Street East, Toronto, ON M5C 1R6, Canada.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Lu C, Xing Z, Wang X, Mao J, Marcucio RS, Miclau T. Anti-inflammatory treatment increases angiogenesis during early fracture healing. Arch Orthop Trauma Surg 2012; 132:1205-13. [PMID: 22622792 DOI: 10.1007/s00402-012-1525-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Indexed: 02/09/2023]
Abstract
OBJECTIVES Both inflammation and angiogenesis are crucial for normal fracture healing. The goal of this work was to determine how anti-inflammatory treatment affects angiogenesis during early stages of fracture repair. METHODS Tibia fractures were created in adult mice and animals were treated with indomethacin (2 mg/kg/day), a non-steroidal anti-inflammatory drug, or PBS once a day beginning from 1 day before fracture and continuing to 6 days after fracture. Animals were killed at 7, 14, and 28 days after injury for histomorphometric analysis of fracture healing. A second group of animals were killed at 3 and 7 days after injury to measure tissue levels of VEGF and interleukin-1 beta (IL-1β). A third group of animals were killed at 3 and 7 days after injury for stereology analysis of macrophage and neutrophil infiltration and tissue vascularization. RESULTS Indomethacin significantly decreased bone and cartilage formation at 7 days after fracture compared to controls. Indomethacin decreased the tissue levels of IL-1β at 3 days after fracture but did not affect the recruitment of macrophages or neutrophils to injured limbs. Indomethacin-treated fractures had similar length density and surface density of vasculature as the controls at 3 days after injury. At 7 days after fracture, vasculature in indomethacin-treated fractures exhibited higher length density and surface density than that in controls. By 28 days after injury, indomethacin-treated fractures still exhibited defects in fracture repair. CONCLUSIONS Anti-inflammatory treatments using indomethacin impair bone and cartilage formation and increase tissue vascularization in the callus during early fracture healing.
Collapse
Affiliation(s)
- Chuanyong Lu
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, San Francisco General Hospital, University of California at San Francisco, San Francisco, CA 94110, USA
| | | | | | | | | | | |
Collapse
|
15
|
Kurmis AP, Kurmis TP, O'Brien JX, Dalén T. The effect of nonsteroidal anti-inflammatory drug administration on acute phase fracture-healing: a review. J Bone Joint Surg Am 2012; 94:815-23. [PMID: 22552671 DOI: 10.2106/jbjs.j.01743] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND The analgesic efficacy of nonsteroidal anti-inflammatory drugs (NSAIDs) is well established, and these agents often form an integral part of posttraumatic pain management. However, potentially deleterious effects of resulting prostaglandin suppression on fracture-healing have been suggested. METHODS A systematic literature review involving searches of electronic databases and online sources was performed to identify articles exploring the influence of NSAIDs on fracture-healing. RESULTS A structured search approach identified 316 papers as potentially relevant to the topic, and these were manually reviewed. The majority described small-scale studies that were retrospective or observational in nature, with limited control of potentially confounding variables, or presented little key information that was not also present in other studies. CONCLUSIONS Although increasing evidence from animal studies suggests that cyclooxygenase-2 (COX-2) inhibition suppresses early fracture-healing, in vivo studies involving human subjects have not provided convincing evidence to substantiate this concern. We found no robust evidence to attest to a significant and appreciable patient detriment resulting from the short-term use of NSAIDs following a fracture. The balance of evidence in the available literature appears to suggest that a short-duration NSAID regimen is a safe and effective supplement to other modes of post-fracture pain control, without a significantly increased risk of sequelae related to disrupted healing.
Collapse
Affiliation(s)
- Andrew P Kurmis
- Department of Orthopaedics, Repatriation General Hospital, Daws Road, Daw Park 5041, South Australia, Australia.
| | | | | | | |
Collapse
|
16
|
Pountos I, Georgouli T, Calori GM, Giannoudis PV. Do nonsteroidal anti-inflammatory drugs affect bone healing? A critical analysis. ScientificWorldJournal 2012; 2012:606404. [PMID: 22272177 PMCID: PMC3259713 DOI: 10.1100/2012/606404] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Accepted: 10/18/2011] [Indexed: 12/21/2022] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) play an essential part in our approach to control pain in the posttraumatic setting. Over the last decades, several studies suggested that NSAIDs interfere with bone healing while others contradict these findings. Although their analgesic potency is well proven, clinicians remain puzzled over the potential safety issues. We have systematically reviewed the available literature, analyzing and presenting the available in vitro animal and clinical studies on this field. Our comprehensive review reveals the great diversity of the presented data in all groups of studies. Animal and in vitro studies present so conflicting data that even studies with identical parameters have opposing results. Basic science research defining the exact mechanism with which NSAIDs could interfere with bone cells and also the conduction of well-randomized prospective clinical trials are warranted. In the absence of robust clinical or scientific evidence, clinicians should treat NSAIDs as a risk factor for bone healing impairment, and their administration should be avoided in high-risk patients.
Collapse
Affiliation(s)
- Ippokratis Pountos
- Academic Department of Trauma & Orthopaedics, School of Medicine, University of Leeds, Leeds LS1 3EX, UK
| | | | | | | |
Collapse
|
17
|
High-dose ketorolac affects adult spinal fusion: a meta-analysis of the effect of perioperative nonsteroidal anti-inflammatory drugs on spinal fusion. Spine (Phila Pa 1976) 2011; 36:E461-8. [PMID: 20489674 DOI: 10.1097/brs.0b013e3181dfd163] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Meta-analysis of literature. OBJECTIVE To evaluate the effect of perioperative nonsteroidal anti-inflammatory drugs (NSAIDs) on the success rate of adult spinal fusion. SUMMARY OF BACKGROUND DATA NSAIDs are commonly used to treat postsurgical orthopedic pain. Studies on animal models have shown a significant inhibiting effect of NSAIDs on osteogenesis process, on which spinal fusion also depends. Recently, great interest has been shown in the effect of NSAIDs on the success rate of adult spinal fusion. Clinical trials have tested the effect of perioperative NSAIDs in spinal fusion procedures. A cumulative result of these studies would give more credit to the final conclusions. METHODS A systematic search of electronic databases and references from eligible articles was conducted. Comparative studies reporting on the results of primary spinal fusion including treatment group of NSAIDs perioperatively were regarded eligible. A pooled estimate of effect size was produced using both random and fixed effect model. RESULTS Five retrospective comparative studies (n = 1403 participants) were included in the present study. The mean age of these patients was more than 40 years and none of them had NSAIDs for longer than 14 days following spinal fusion surgery. High-dose ketorolac showed a statistically significant adverse effect on spinal fusion (P = 0.001, RR = 2.87, 95% CI = 1.53 = - 5.38) with no statistical heterogeneity (I = 3%, P = 0.38), whereas normal-dose NSAIDs (ketorolac, diclofenac sodium, celecoxib, or rofecoxib) did not appear to produce inferior results than the no-NSAIDs group (P = 0.30, RR = 1.39, 95% CI = 0.74 - 2.61) with no statistical heterogeneity (I² = 0%, P = 0.50). CONCLUSION Although randomized controlled trials would be optimal for meta-analyses, the data of this review revealed that short-time (<14 days) exposure to normal-dose NSAIDs (ketorolac, diclofenac sodium, celecoxib, or rofecoxib) were safe after spinal fusion, whereas short-time (<14 days) exposure to high-dose ketorolac increased the risk of nonunion, which meant that the effect of perioperative NSAIDs on spinal fusion might be dose-dependent. Further studies would be needed to find out whether long-time exposure to normal-dose NSAIDs could also increase the risk of nonunion and which type of NSAIDs would like to have a worse effect on spinal fusion.
Collapse
|
18
|
Affiliation(s)
- Sigbjørn Dimmen
- Orthopaedic Department, Ullevaal Hospital, Oslo University Hospital, Kirkeveien 166, 0407 Oslo, Norway.
| |
Collapse
|
19
|
Cottrell J, O’Connor JP. Effect of Non-Steroidal Anti-Inflammatory Drugs on Bone Healing. Pharmaceuticals (Basel) 2010; 3:1668-1693. [PMID: 27713323 PMCID: PMC4034003 DOI: 10.3390/ph3051668] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 05/05/2010] [Accepted: 05/24/2010] [Indexed: 01/11/2023] Open
Abstract
Nonspecific and COX-2 selective nonsteroidal anti-inflammatory drugs (NSAIDs) function by inhibiting the cyclooxygenase isoenzymes and effectively reduce pain and inflammation attributed to acute or chronic musculoskeletal pathologies. However, use of NSAIDs as an analgesic is thought to negatively contribute to bone healing. This review strived to provide a thorough unbiased analysis of the current research conducted on animals and humans regarding NSAIDs and their effect on bone healing. Specifically, this review discusses the role of animal models, dosing regiments, and outcome parameters when examining discrepancies about NSAIDS and their effects on bone regeneration. The role of COX-2 in bone regeneration needs to be better defined in order to further elucidate the impact of NSAIDs on bone healing.
Collapse
Affiliation(s)
| | - J. Patrick O’Connor
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-973-972-5011; Fax: +1-973-972-5594
| |
Collapse
|
20
|
Xing Z, Lu C, Hu D, Yu YY, Wang X, Colnot C, Nakamura M, Wu Y, Miclau T, Marcucio RS. Multiple roles for CCR2 during fracture healing. Dis Model Mech 2010; 3:451-8. [PMID: 20354109 DOI: 10.1242/dmm.003186] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bone injury induces an inflammatory response that involves neutrophils, macrophages and other inflammatory cells. The recruitment of inflammatory cells to sites of injury occurs in response to specific signaling pathways. The CC chemokine receptor type 2 (CCR2) is crucial for recruiting macrophages, as well as regulating osteoclast function. In this study, we examined fracture healing in Ccr2-/- mice. We first demonstrated that the expression of Ccr2 transcripts and the filtration of macrophages into fracture calluses were most robust during the early phases of fracture healing. We then determined that the number of macrophages at the fracture site was significantly lower in Ccr2-/- mice compared with wild-type controls at 3 days after injury. As a result, impaired vascularization, decreased formation of callus, and delayed maturation of cartilage were observed at 7 days after injury in mutant mice. At day 14, Ccr2-/- mice had less bone in their calluses. At day 21, Ccr2-/- mice had larger calluses and more bone compared with wild-type mice, suggesting a delayed remodeling. In addition, we examined the effect of Ccr2 mutation on osteoclasts. We found that a lack of Ccr2 did not affect the number of osteoclasts within fracture calluses at 21 days after injury. However, Ccr2-/- osteoclasts exhibited a decreased ability to resorb bone compared with wild-type cells, which could contribute to the delayed remodeling of fracture calluses observed in Ccr2-/- mice. Collectively, these results indicate that a deficiency of Ccr2 reduces the infiltration of macrophages and impairs the function of osteoclasts, leading to delayed fracture healing.
Collapse
Affiliation(s)
- Zhiqing Xing
- Orthopaedic Trauma Institute, Laboratory for Skeletal Regeneration and Department of Orthopaedic Surgery, San Francisco General Hospital, University of California at San Francisco, San Francisco, CA 94110, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wixted JJ, Fanning PJ, Gaur T, O'Connell SL, Silva J, Mason-Savas A, Ayers DC, Stein GS, Lian JB. Enhanced fracture repair by leukotriene antagonism is characterized by increased chondrocyte proliferation and early bone formation: a novel role of the cysteinyl LT-1 receptor. J Cell Physiol 2009; 221:31-9. [PMID: 19544365 DOI: 10.1002/jcp.21809] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Inflammatory mediators and drugs which affect inflammation can influence the healing of injured tissues. Leukotrienes are potent inflammatory mediators, and similar to prostaglandins, are metabolites of arachidonic acid which can have positive or negative effects on bone and cartilage tissues. Here we tested the hypothesis that blocking the negative regulation of leukotrienes, would lead to enhanced endochondral bone formation during fracture repair. A closed femoral fracture was created in mice. Animals were divided into three groups for treatment with either montelukast sodium, a cysteinyl leukotriene type 1 receptor antagonist (trade name Singulair), zileuton, a 5-lipoxygenase enzyme inhibitor (trade name Zyflo), or carrier alone. The fractures were analyzed using radiographs, quantitative gene expression, histology and histomorphometry, and immunohistochemistry. Both the montelukast sodium group and the zileuton group exhibited enhanced fracture repair when compared with controls. Both treatment groups exhibited increased callous size and earlier bone formation when compared to controls as early as day 7. Gene expression analysis of treatment groups showed increased markers of chondrocyte proliferation and differentiation, and increased early bone formation markers when compared with controls. Treatment with montelukast sodium directly targeted the cysteinyl leukotriene type 1 receptor, leading to increased chondrocyte proliferation at early time points. These novel findings suggests a potential mechanism by which the cysteinyl leukotriene type 1 receptor acts as a negative regulator of chondrocyte proliferation, with important and previously unrecognized implications for both fracture repair, and in a broader context, systemic chondrocyte growth and differentiation.
Collapse
Affiliation(s)
- John J Wixted
- Department of Orthopedic Surgery and Physical Rehabilitation, University of Massachusetts Medical School, Worcester, Massachusetts, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Parecoxib and indomethacin delay early fracture healing: a study in rats. Clin Orthop Relat Res 2009; 467:1992-9. [PMID: 19319614 PMCID: PMC2706352 DOI: 10.1007/s11999-009-0783-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Accepted: 02/27/2009] [Indexed: 01/31/2023]
Abstract
Nonsteroidal antiinflammatory drugs (NSAIDs) are used to reduce inflammatory response and pain. These drugs have been reported to impair bone metabolism. Parecoxib, a specific COX-2 inhibitor, exerts an inhibitory effect on the mineralization of fracture callus after a tibial fracture in rats. Decreased bone mineral density (BMD) at a fracture site may indicate impairment of early healing, casting doubt on the safety of using COX-2 inhibitors during the early treatment of diaphyseal fractures. Forty-two female Wistar rats were randomly allocated to three groups. They were given parecoxib, indomethacin, or saline intraperitoneally for 7 days after being subjected to a closed tibial fracture stabilized with an intramedullary nail. Two and 3 weeks after surgery, the bone density at the fracture site was measured using dual energy xray absorptiometry (DEXA). Three weeks after the operation the rats were euthanized and the healing fractures were mechanically tested in three-point cantilever bending. Parecoxib decreased BMD at the fracture site for 3 weeks after fracture, indomethacin for 2 weeks. Both parecoxib and indomethacin reduced the ultimate bending moment and the bending stiffness of the healing fractures after 3 weeks. These results suggest COX inhibitors should be avoided in the early phase after fractures.
Collapse
|
23
|
Dimmen S, Nordsletten L, Engebretsen L, Steen H, Madsen JE. The effect of parecoxib and indometacin on tendon-to-bone healing in a bone tunnel. ACTA ACUST UNITED AC 2009; 91:259-63. [DOI: 10.1302/0301-620x.91b2.21471] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Conventional non-steroidal anti-inflammatory drugs (NSAIDs) and newer specific cyclo-oxygenase-2 (cox-2) inhibitors are commonly used in musculoskeletal trauma and orthopaedic surgery to reduce the inflammatory response and pain. These drugs have been reported to impair bone metabolism. In reconstruction of the anterior cruciate ligament the hamstring tendons are mainly used as the graft of choice, and a prerequisite for good results is healing of the tendons in the bone tunnel. Many of these patients are routinely given NSAIDs or cox-2 inhibitors, although no studies have elucidated the effects of these drugs on tendon healing in the bone tunnel. In our study 60 female Wistar rats were randomly allocated into three groups of 20. One received parecoxib, one indometacin and one acted as a control. In all the rats the tendo-Achillis was released proximally from the calf muscles. It was then pulled through a drill hole in the distal tibia and sutured anteriorly. The rats were given parecoxib, indometacin or saline intraperitoneally twice daily for seven days. After 14 days the tendon/bone-tunnel interface was subjected to mechanical testing. Significantly lower maximum pull-out strength (p < 0.001), energy absorption (p < 0.001) and stiffness (p = 0.035) were found in rats given parecoxib and indometacin compared with the control group, most pronounced with parecoxib.
Collapse
Affiliation(s)
- S. Dimmen
- Orthopaedic Centre, Ullevaal University Hospital and Faculty of Medicine, University of Oslo, N-0407 Oslo, Norway
| | - L. Nordsletten
- Orthopaedic Centre, Ullevaal University Hospital and Faculty of Medicine, University of Oslo, N-0407 Oslo, Norway
| | - L. Engebretsen
- Orthopaedic Centre, Ullevaal University Hospital and Faculty of Medicine, University of Oslo, N-0407 Oslo, Norway
| | - H. Steen
- Institute for Surgical Research, Rikshospitalet-Radiumhospitalet Medical Centre and Faculty of Medicine, University of Oslo, N-0027, Oslo, Norway
| | - J. E. Madsen
- Orthopaedic Centre, Ullevaal University Hospital and Faculty of Medicine, University of Oslo, N-0407 Oslo, Norway
| |
Collapse
|
24
|
Dimmen S, Nordsletten L, Engebretsen L, Steen H, Madsen JE. Negative effect of parecoxib on bone mineral during fracture healing in rats. Acta Orthop 2008; 79:438-44. [PMID: 18626809 DOI: 10.1080/17453670710015373] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Non-steroidal anti-inflammatory drugs (NSAIDs) are conventional cyclooxygen-ase (cox) inhibitors commonly used in musculoskeletal trauma to reduce the inflammatory response and pain, but they also seem to affect bone metabolism. Parecoxib is a cox inhibitor that selectively inhibits cox-2. Through their selective mechanism of action, these newer drugs are supposed to reduce the gastrointestinal side effects of conventional cox inhibitors. The effects on bone metabolism and healing have, however, not been fully elucidated. Thus, there are reasons for concern regarding the potential negative effects of these drugs on bone metabolism and bone repair. We investigated the effects of short-term administration of parecoxib on bone mineral formation and bone healing in rats. ANIMALS AND METHODS 26 female Wistar rats were given parecoxib intraperitoneally for 7 days after a closed tibial fracture that was stabilized with an intra-medullary nail, and 26 animals were given saline. At 2, 3, and 6 weeks after surgery bone mineral density (BMD) at the fracture site was measured using dualenergy X-ray absorptiometry (DEXA). 6 weeks after the fracture, 14 rats from the parecoxib group and 16 rats from the placebo group were killed for mechanical testing, and the rest of the animals were killed for tissue analysis. The healing fractures and the intact contralateral tibias were mechanically tested by three-point cantilever bending. RESULTS The BMD at the fracture site was calculated as the average of the results after 2,3, and 6 weeks. Mean BMD was lower in the parecoxib group, 0.23 (SD 0.06) g/ cm2, than in the control group, 0.27 (SD 0.05) g/cm2 (p = 0.01). There were no statistically significant differences in mechanical properties of the healing fractures after 6 weeks. However, the study may have lacked sufficient statistical power to determine whether a negative effect on healing had occurred. INTERPRETATION No mechanical differences were detected between the control and treatment groups after 6 weeks, but they may have been present earlier in the fracture healing process. Our findings do, however, indicate that parecoxib given postoperatively for a week has a negative effect on mineralization during the early phase of fracture healing.
Collapse
Affiliation(s)
- Sigbøjrn Dimmen
- Institute of Surgical Research and Biomechanics Laboratory, Orthopedics Department, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Norway.
| | | | | | | | | |
Collapse
|
25
|
Vuolteenaho K, Moilanen T, Moilanen E. Non-steroidal anti-inflammatory drugs, cyclooxygenase-2 and the bone healing process. Basic Clin Pharmacol Toxicol 2007; 102:10-4. [PMID: 17973900 DOI: 10.1111/j.1742-7843.2007.00149.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Traditional non-steroidal anti-inflammatory drugs (NSAID) and selective cyclooxygenase-2 (COX-2) inhibitors are widely used in the treatment of pain, including bone fracture pain and orthopaedic post-operative pain. The gastrointestinal and cardiovascular adverse effects of NSAIDs are acknowledged, but their effects on bone are less widely known. Prostaglandins play an important role in the regulation of osteoblast and osteoclast functions, and inhibition of prostaglandin production retards bone formation. Therefore, NSAIDs could be expected to have significant consequences in divergent clinical situations where bone formation or remodelling is a contributing factor. The present survey reviews current experimental and clinical evidence related to two of those conditions (i.e. on ectopic bone formation and on bone fracture healing). NSAIDs are used clinically to prevent ectopic bone formation (also known as heterotopic ossification) (e.g. after total hip arthroplasty or trauma). The efficacy of NSAIDs in the avoidance of heterotopic ossification has been documented in controlled clinical trials, but the inherent risks (e.g. on healing processes and on loosening of prostheses) need further studies. At the same time, NSAIDs are widely used in the treatment of fracture pain, and their inhibitory effects on the ongoing bone healing process have raised concerns. Results of fracture healing studies in animals treated with NSAIDs or in mice lacking COX-2 gene show that inhibition or deficiency of COX-2 impairs the bone healing process. The limited clinical data also support the assumption that inhibition of COX-2 by non-selective or COX-2-selective NSAIDs delays fracture healing. However, the clinical significance of the effect in various patient groups needs to be carefully assessed and further investigations are needed to characterize the patients at the highest risk for NSAID-induced delayed fracture healing and its complications. In the meantime, use of NSAIDs in fracture patients should be cautious, keeping in mind the benefits of pain relief and inhibition of ectopic bone formation on one hand, and the risks of non-union and retarded union on the other hand.
Collapse
Affiliation(s)
- Katriina Vuolteenaho
- The Immunopharmacology Research Group, Medical School, University of Tampere and Research Unit, Tampere University Hospital, Tampere, Finland
| | | | | |
Collapse
|
26
|
Karachalios T, Boursinos L, Poultsides L, Khaldi L, Malizos KN. The effects of the short-term administration of low therapeutic doses of anti-COX-2 agents on the healing of fractures. ACTA ACUST UNITED AC 2007; 89:1253-60. [PMID: 17905969 DOI: 10.1302/0301-620x.89b9.19050] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We have evaluated the effect of the short-term administration of low therapeutic doses of modern COX-2 inhibitors on the healing of fractures. A total of 40 adult male New Zealand rabbits were divided into five groups. A mid-diaphyseal osteotomy of the right ulna was performed and either normal saline, prednisolone, indometacin, meloxicam or rofecoxib was administered for five days. Radiological, biomechanical and histomorphometric evaluation was performed at six weeks. In the group in which the highly selective anti-COX-2 agent, rofecoxib, was used the incidence of radiologically-incomplete union was similar to that in the control group. All the biomechanical parameters were statistically significantly lower in both the prednisolone and indometacin (p = 0.01) and in the meloxicam (p = 0.04) groups compared with the control group. Only the fracture load values were found to be statistically significantly lower (p = 0.05) in the rofecoxib group. Histomorphometric parameters were adversely affected in all groups with the specimens of the rofecoxib group showing the least negative effect. Our findings indicated that the short-term administration of low therapeutic doses of a highly selective COX-2 inhibitor had a minor negative effect on bone healing.
Collapse
Affiliation(s)
- T Karachalios
- Orthopaedic Department, School of Medicine, Faculty of Health Sciences, University of Thessaly, Larisa, Greece.
| | | | | | | | | |
Collapse
|
27
|
Gerstenfeld LC, Al-Ghawas M, Alkhiary YM, Cullinane DM, Krall EA, Fitch JL, Webb EG, Thiede MA, Einhorn TA. Selective and nonselective cyclooxygenase-2 inhibitors and experimental fracture-healing. Reversibility of effects after short-term treatment. J Bone Joint Surg Am 2007; 89:114-25. [PMID: 17200318 DOI: 10.2106/jbjs.f.00495] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Cyclooxygenase-2-specific anti-inflammatory drugs (coxibs) and nonspecific nonsteroidal anti-inflammatory drugs have been shown to inhibit experimental fracture-healing. The present study tested the hypothesis that these effects are reversible after short-term treatment. METHODS With use of a standard model of fracture-healing, identical ED50 dosages of either a nonsteroidal anti-inflammatory drug (ketorolac), a coxib (valdecoxib), or vehicle (control) were orally administered to rats for either seven or twenty-one days and fracture-healing was assessed with biomechanical, histological, and biochemical analyses. RESULTS When healing was assessed at twenty-one days, the seven-day treatment produced only a trend for a higher rate of nonunion in valdecoxib and ketorolac-treated animals as compared with controls. No differences were observed at thirty-five days. The twenty-one-day treatment produced significantly more nonunions in valdecoxib-treated animals as compared with either ketorolac-treated or control animals (p < 0.05), but these differences disappeared by thirty-five days. The dose-specific inhibition of these drugs on prostaglandin E2 levels and the reversibility of the effects after drug withdrawal were assessed in fracture calluses and showed that ketorolac treatment led to twofold to threefold lower levels of prostaglandin E2 than did valdecoxib. Withdrawal of either drug after six days led to a twofold rebound in these levels by fourteen days. Histological analysis showed delayed remodeling of calcified cartilage and reduced bone formation in association with valdecoxib treatment. CONCLUSIONS Cyclooxygenase-2-specific drugs inhibit fracture-healing more than nonspecific nonsteroidal anti-inflammatory drugs, and the magnitude of the effect is related to the duration of treatment. However, after the discontinuation of treatment, prostaglandin E2 levels are gradually restored and the regain of strength returns to levels similar to control.
Collapse
Affiliation(s)
- L C Gerstenfeld
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Boston University Medical Center, 715 Albany Street, R-205, Boston, MA 02118, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Velásquez-Forero F, García P, Triffitt JT, Llach F. Prostaglandin E1 increases in vivo and in vitro calcitriol biosynthesis in rabbits. Prostaglandins Leukot Essent Fatty Acids 2006; 75:107-15. [PMID: 16876395 DOI: 10.1016/j.plefa.2006.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2005] [Revised: 03/27/2006] [Accepted: 03/29/2006] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Prostaglandins have an anabolic effect on bone. Possible mediation of this effect is via calcitriol. This study determines in vivo and in vitro effects of PGE(1) on calcitriol synthesis. METHODOLOGY In vivo: rabbits received intravenous vehicle or prostaglandin E(1) (50 microg/day) for 20 days before measurements of serum total and ionic calcium, magnesium and phosphorus levels, total and bone-specific alkaline phosphatases, 25(OH)D(3), calcitriol, parathyroid hormone and calcitonin. In vitro: rabbit proximal renal tubules were incubated with 25(OH)D(3) (8 microM) together with PGE(1) (2.82 x 10(-6) M) and the prostaglandin receptor inhibitor AH6809 (10(-4) M) in selected samples. After 5 or 30 min incubation, calcitriol production was measured by radioimmunoassay and data analysed statistically. RESULTS In vivo, in groups receiving PGE(1), levels of total Ca, Mg and calcitriol increased significantly and 25 dihydroxyvitamin D(3), parathyroid hormone and calcitonin remained unchanged. In vitro, PGE(1) increased calcitriol biosynthesis and the prostaglandin inhibitor AH6809 reduced calcitriol levels significantly after prolonged incubation. CONCLUSIONS In vivo and in vitro results demonstrate that PGE(1) stimulates calcitriol synthesis. This study represent a major advancement in knowledge of bone metabolism.
Collapse
Affiliation(s)
- F Velásquez-Forero
- Laboratorio de Metabolismo Mineral Oseo, Hospital Infantil de México Federico Gómez, Y programa de doctorado en Ciencias Biológicas, Universidad Autónoma Metropolitana, México City, México.
| | | | | | | |
Collapse
|
29
|
Bergenstock M, Min W, Simon AM, Sabatino C, O'Connor JP. A comparison between the effects of acetaminophen and celecoxib on bone fracture healing in rats. J Orthop Trauma 2005; 19:717-23. [PMID: 16314720 DOI: 10.1097/01.bot.0000184144.98071.5d] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVES This study compared the acute treatment effects of systemic analgesics with (celecoxib) and without anti-inflammatory activity (acetaminophen) on bone fracture healing. STUDY DESIGN Longitudinal time study of fracture healing in rats. METHODS Closed, mid diaphyseal femur fractures were produced in female Sprague-Dawley rats. The rats were treated for 10 days after fracture with 60 or 300 mg/kg of acetaminophen, 3 or 6 mg/kg of celecoxib, or vehicle by once-daily oral dosing. Fracture healing was measured after 8 weeks by radiographic examination, mechanical testing, and histology. RESULTS Radiographic scoring indicated that acute celecoxib treatment significantly impaired fracture healing; acetaminophen treatment had no negative effect. Mechanical testing supported the radiographic observations. No negative effects of celecoxib or acetaminophen treatment on the structural properties (peak torque and torsional rigidity) of the healing femurs were detected. In contrast, celecoxib treatment, but not acetaminophen treatment, significantly reduced the material properties (maximum shear stress and shear modulus) of the healing femurs (P < 0.001). Post-mechanical testing examination of the healing femurs found that 73% of the vehicle-treated or acetaminophen-treated femurs had healed as unions (30/41), 27% failed as incomplete unions (11/41), and none failed as nonunions (0%). In contrast, only 21% of the fractured femurs from the celecoxib treated rats had healed as unions (7/34), 53% failed as incomplete unions (18/34), and 26% failed as nonunions (9/34). The proportion of nonunions among the celecoxib-treated rats was significantly higher compared with the control and acetaminophen-treated rats (P < 0.001). Histologic examination indicated that similar to previous studies, celecoxib treatment, but not acetaminophen treatment, altered normal fracture callus morphology in which cartilage rather than new bone abuts the fracture site. CONCLUSIONS No negative effect from acute acetaminophen treatment on fracture healing was detected. In contrast, acute treatment with celecoxib, a selective cyclooxygenase-2 inhibitor with anti-inflammatory activity, significantly impaired fracture healing.
Collapse
Affiliation(s)
- Marika Bergenstock
- Department of Orthopaedics, UMDNJ-New Jersey Medical School, Newark, NJ 07103, USA
| | | | | | | | | |
Collapse
|
30
|
Goodman SB, Ma T, Mitsunaga L, Miyanishi K, Genovese MC, Smith RL. Temporal effects of a COX-2-selective NSAID on bone ingrowth. J Biomed Mater Res A 2005; 72:279-87. [PMID: 15666361 DOI: 10.1002/jbm.a.30231] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The effects of a short course of a COX-2 inhibitor on bone healing when the drug is discontinued are unknown. We examined the effects of rofecoxib on bone ingrowth over a 6-week period using a well-defined animal model. The Bone Harvest Chamber was implanted bilaterally in mature rabbits. After osseointegration of the chamber, the following treatments were given for 6 weeks each, followed by a harvest in each case: control-no drug; oral rofecoxib (12.5 mg/day) for the first 2 of 6 weeks; washout period-no drug; oral rofecoxib for the last 2 of 6 weeks; washout period-no drug; rofecoxib given continuously for all 6 weeks. Harvested specimens were snap-frozen, cut into serial 6-microm sections, and stained with hematoxylin and eosin and alkaline phosphatase (osteoblast marker), and processed using immunohistochemistry to identify the vitronectin receptor (osteoclast-like cells). Rofecoxib given continuously for 6 weeks yielded statistically less bone ingrowth compared to the control treatment. Rofecoxib given during the initial or final 2 weeks of a 6-week treatment did not appear to interfere with bone ingrowth. This suggests that the effects of COX-2 inhibitors on bone are less profound when the drug is administered for a short period of time.
Collapse
Affiliation(s)
- Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University Medical Center, Stanford, California 94305-5341, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
Pediatric patients benefit from patient-controlled analgesia (PCA), which eliminates the need for painful intramuscular injections of opioids and improves the child's sense of control. Age is often used inappropriately as a criterion for PCA use in children. Children must be carefully screened for their cognitive and physical ability to manage their pain using PCA. Family-controlled analgesia and nurse-controlled analgesia may be considered in select cases as alternatives to PCA in children with cognitive or physical disabilities. PCA dosage regimens must be individualized on the basis of age. Monitoring parameters must be age appropriate. Potential adverse effects of PCA therapy, including respiratory depression, nausea, vomiting, and pruritus, can be prevented or controlled. Clinicians must become aware of age-related and developmental differences in the pharmacokinetic, pharmacodynamic, and monitoring parameters for the pediatric patient. The safety and efficacy of PCA in pediatric patients has been established, and its role has increased beyond postoperative pain management.
Collapse
Affiliation(s)
- Victoria Tutag Lehr
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy & Health Sciences, Wayne State University, Detroit, MI, USA
| | | |
Collapse
|
32
|
Gerstenfeld LC, Thiede M, Seibert K, Mielke C, Phippard D, Svagr B, Cullinane D, Einhorn TA. Differential inhibition of fracture healing by non-selective and cyclooxygenase-2 selective non-steroidal anti-inflammatory drugs. J Orthop Res 2003; 21:670-5. [PMID: 12798067 DOI: 10.1016/s0736-0266(03)00003-2] [Citation(s) in RCA: 262] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) specifically inhibit cyclooxygenase (COX) activity and are widely used as anti-arthritics, post-surgical analgesics, and for the relief of acute musculoskeletal pain. Recent studies suggest that non-specific NSAIDs, which inhibit both COX-1 and COX-2 isoforms, delay bone healing. The objectives of this study were 2-fold; first, to measure the relative changes in the normal expression of COX-1 and COX-2 mRNAs over a 42 day period of fracture healing and second, to compare the effects of a commonly used non-specific NSAID, ketorolac, with a COX-2 specific NSAID, Parecoxib (a pro-drug of valdecoxib), on this process. Simple, closed, transverse fractures were generated in femora of male Sprague-Dawley rats weighing approximately 450 g each. Total RNA was prepared from the calluses obtained prior to fracture and at 1, 3, 5, 7, 10, 14, 21, 35 and 42 days post-fracture and levels of COX-1 and COX-2 mRNA were measured using real time PCR. While the relative levels of COX-1 mRNA remained constant over a 21-day period, COX-2 mRNA levels showed peak expression during the first 14 days of healing and returned to basal levels by day 21. Mechanical properties of the calluses were then assessed at 21 and 35 days post-fracture in untreated animals and animals treated with either ketorolac or high or low dose parecoxib. At both 21 and 35 days after fracture, calluses in the group treated with the ketorolac showed a significant reduction in mechanical strength and stiffness when compared with controls (p<0.05). At the 21-day time point, calluses of the parecoxib treated animals showed a lower mean mechanical strength than controls, but the inhibition was not statistically significant. Based on physical analysis of the bones, 3 of 12 (25%) of the ketorolac-treated and 1 of 12 (8%) of the high dose parecoxib-treated animals showed failure to unite their fractures by 21 days, while all fractures in both groups showed union by 35 days. Histological analysis at 21 days showed that the calluses in the ketorolac-treated group contained substantial amounts of residual cartilage while neither the control nor the parecoxib-treated animals showed comparable amounts of cartilage at this stage. These results demonstrate that ketorolac and parecoxib delay fracture healing in this model, but in this study daily administration of ketorolac, a non-selective COX inhibitor had a greater affect on this process. They further demonstrate that a COX-2 selective NSAID, such as parecoxib (valdecoxib), has only a small effect on delaying fracture healing even at doses that are known to fully inhibit prostaglandin production.
Collapse
Affiliation(s)
- Louis C Gerstenfeld
- Department of Orthopaedic Surgery, Orthopaedic Research Laboratory, Boston University Medical Center, Boston, MA 02118, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Bhandari M, Tornetta P, Sprague S, Najibi S, Petrisor B, Griffith L, Guyatt GH. Predictors of reoperation following operative management of fractures of the tibial shaft. J Orthop Trauma 2003; 17:353-61. [PMID: 12759640 DOI: 10.1097/00005131-200305000-00006] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Accurate prediction of likelihood of reoperation in patients with tibial shaft fractures would facilitate optimal management. Previous studies were limited by small sample sizes and noncomprehensive examination of possible risk factors. OBJECTIVE We conducted an observational study to determine which prognostic factors were associated with an increased risk of reoperation following operative treatment in a heterogeneous population of patients with tibial shaft fractures. DESIGN Retrospective observational study. SETTING Level 1 trauma center. METHODS We identified 200 patients with tibial shaft fractures from two university-affiliated centers. Two reviewers independently abstracted data regarding 20 possible prognostic variables, reviewed preoperative and postoperative radiographs, and documented reoperations (defined as any surgical procedure </=1 year after the initial surgery that was aimed specifically at achieving bony union of the fracture, including bone grafts, implant exchanges, or débridement for infections). We chose a Cox proportion hazards model to conduct a survival analysis for time to reoperation and constructed a multivariable model to estimate the relative risk of reoperation and associated 95%confidence interval (CI) for each predictor variable. MAIN OUTCOME MEASURES Time to reoperation following the initial surgery. RESULTS Complete follow-up information was available for 192 of 200 (96%) patients. Three variables predicted reoperation: the presence of an open fracture wound (relative risk 4.32, 95% CI 1.76 to 11.26), lack of cortical continuity between the fracture ends following fixation (relative risk 8.33, 95% CI 3.03 to 25.0), and the presence of a transverse fracture (relative risk 20.0, 95% CI 4.34 to 142.86). CONCLUSIONS We identified a set of three simple prognostic variables (open fracture, transverse fracture, and postoperative fracture gap) that can assist surgeons in predicting reoperation following operative treatment of tibial shaft fractures.
Collapse
Affiliation(s)
- Mohit Bhandari
- Department of Clinical Epidemiology, McMaster University, Hamilton, Ontario, Canada.
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
BACKGROUND Nonsteroidal anti-inflammatory drugs have been implicated in the development of delayed unions and nonunion after fractures in animal models. Previous investigations have identified two important factors as determinants of delayed fracture healing: early drug administration and a dose-dependent effect. OBJECTIVE The purpose of this investigation was to study the effect of tenoxicam, a nonsteroidal anti-inflammatory drug, on the fracture healing process in rat tibiae. METHODS Fifty-eight Wistar rats were randomly divided in four groups (I, II, III, and IV). Group I (control group, n=12) was given 0.1ml saline solution per day intramuscularly. Groups II (n=12), III (n=12), and IV (n=12) were administered 10mg per kg per day of tenoxicam intramuscularly. Administration of substances was begun on a week before to 48h after the fracturing procedure and continued during the entire experiment. Callus formation was studied histologically and histomorphologically, using light microscopy. In addition, a histologic grading based on the morphologic stage of fracture healing was carried out at 4 weeks, according to the criteria proposed by Allen et al. RESULTS There was a significant difference in treatment effect between Group I (saline solution) and Groups II, III, and IV (tenoxicam) (P=0.07). Histologically and histomorphologically, there were qualitative and quantitative delay in callus formation at all tenoxicam groups. This was more pronounced the earlier the nonsteroidal anti-inflammatory drug was started, although no significant difference could be detected between Groups II, III, and IV (P>(alpha=10%)). Four weeks after fracture, Group I (n=3) showed complete osseous union, Groups II (n=3) and III (n=3), complete cartilaginous union, and Group IV (n=3), incomplete osseous union, according to Allen et al. By using this rating scale, the difference between control and drug-treated groups was statistically significant (P<0.1). CONCLUSION Under studied conditions, this investigation shows that administration of tenoxicam intramuscularly delays fracture healing process in rat tibiae. These results suggest the hypothesis that early drug administration may delay bone healing after experimental fractures in animals, although it could not be detected statistically significant.
Collapse
Affiliation(s)
- Vincenzo Giordano
- Departamento de Ortopedia e Traumatologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | | | | | | | | |
Collapse
|
35
|
Yugoshi LI, Sala MA, Brentegani LG, Lamano Carvalho TL. Histometric study of socket healing after tooth extraction in rats treated with diclofenac. Braz Dent J 2003; 13:92-6. [PMID: 12238809 DOI: 10.1590/s0103-64402002000200003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The purpose of the present study was to investigate if diclofenac administration interferes with the time course of alveolar wound healing in rats. Forty-two Wistar rats were used, 21 rats received 10 mg/kg/day of diclofenac one day before and 4 days after extraction of the right maxillary incisors and 21 rats received saline. The animals were sacrificed 7, 14 and 21 days after tooth extraction. Progressive new bone formation and a decrease in the volume fraction of blood clot and connective tissue from 1 to 3 weeks after tooth extraction was quantified using the histometric point-counting method. Diclofenac treatment caused a significant delay in new bone formation in association with an impairment of blood clot remission/organization.
Collapse
Affiliation(s)
- Luciana Ibara Yugoshi
- Department of Morphology, Stomatology and Physiology, Faculty of Dentistry of Ribeirão Preto, University of São Paulo, Brazil
| | | | | | | |
Collapse
|
36
|
Goodman S, Ma T, Trindade M, Ikenoue T, Matsuura I, Wong N, Fox N, Genovese M, Regula D, Smith RL. COX-2 selective NSAID decreases bone ingrowth in vivo. J Orthop Res 2002; 20:1164-9. [PMID: 12472224 DOI: 10.1016/s0736-0266(02)00079-7] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Whether non-steroidal anti-inflammatory drug (NSAID)-induced suppression of bone ingrowth is due to cyclooxygenase-1 (COX-1) inhibition, cyclooxygenase-2 (COX-2) inhibition, or through a yet unidentified pathway is unknown. In this study, the effects of a non-specific COX-1 and COX-2 inhibitor, versus a specific COX-2 inhibitor on bone ingrowth and tissue differentiation are examined in vivo. Harvest chambers were implanted unilaterally in the tibiae of eight mature, New Zealand white rabbits. After a 6-week period for osseointegration of the chamber, the following oral treatments were given for 4 weeks each, followed by a harvest in each case: drinking water with no NSAID (control 1), Naproxen sodium--a COX-1 and COX-2 non-specific inhibitor at a dose of 110 mg/kg/day in the drinking water, drinking water with no NSAID (control 2), and Rofecoxib-a COX-2 inhibitor at a dose of 12.5 mg/day inserted directly into the rabbit's mouth. Harvested specimens were snap frozen, cut into serial 6 microm sections and stained with hematoxylin and eosin for general morphological characterization, and alkaline phosphatase (osteoblast marker). Sections were also processed for immunoperoxidase staining using monoclonal antibodies to identify cells expressing the vitronectin receptor (osteoclast-like cells). With drinking water alone, the percentage of bone ingrowth averaged 24.8 +/- 2.9% and 29.9 +/- 4.5% respectively. Naproxen sodium in the drinking water and oral Rofecoxib decreased bone ingrowth significantly (15.9 +/- 3.3%. p = 0.031 and 18.5 +/- 2+/-4%, p = 0.035 compared to drinking water respectively). Both Naproxen sodium (p = 0.026) and Rofecoxib (p = 0.02) decreased the number of CD51 positive osteoclast-like cells per section compared with drinking water alone. Rofecoxib decreased the area of osteoblasts per section area (p = 0.014) compared to controls, although the value for Naproxen sodium did not reach statistical significance. The results of the present study suggest that bone formation is suppressed by oral administration of an NSAID which contains a COX-2 inhibitor. COX-2 inhibitors currently taken for arthritis and other conditions may potentially delay fracture healing and bone ingrowth.
Collapse
Affiliation(s)
- Stuart Goodman
- Division of Orthopaedic Surgery, Orthopaedic Research Laboratory, Stanford University Medical Center, 300 Pasteur Drive, R144, Stanford, CA 94305, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Martin GJ, Boden SD, Titus L. Recombinant human bone morphogenetic protein-2 overcomes the inhibitory effect of ketorolac, a nonsteroidal anti-inflammatory drug (NSAID), on posterolateral lumbar intertransverse process spine fusion. Spine (Phila Pa 1976) 1999; 24:2188-93; discussion 2193-4. [PMID: 10562982 DOI: 10.1097/00007632-199911010-00003] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN An animal model of posterolateral intertransverse process spine fusion healing. OBJECTIVE To evaluate the effect of systemic ketorolac, alone and in combination with locally applied recombinant human bone morphogenetic protein-2, on spine fusion healing. SUMMARY OF BACKGROUND DATA The effect of nonsteroidal anti-inflammatory drugs on bone graft healing in animals remains controversial. However, most studies point to the inhibition of fracture repair, especially during the early healing period. METHODS Forty-nine adult New Zealand white rabbits underwent single-level lumbar fusion with autologous iliac bone graft. Two mini-osmotic pumps were implanted subcutaneously and filled with saline as a control or ketorolac. Rabbits were divided into three groups: 1) control (saline in pump); 2) nonsteroidal anti-inflammatory drug (ketorolac in pump); 3) nonsteroidal anti-inflammatory drug (ketorolac in pump) and bone morphogenetic protein (bone graft soaked in a 3.0 mg solution of recombinant human bone morphogenetic protein-2. All rabbits were killed after 6 weeks. RESULTS In the control group, 75% (12 in 16) of the surviving rabbits were judged to have solidly fused lumbar spines as compared with only 35% (6 in 17) of the animals that received ketorolacachieved fusion (P = 0.037). Of the animals that received ketorolac and recombinant bone morphogenetic protein-2, 100% (9 in 9) fused. CONCLUSIONS The results of this study confirm the detrimental effect of a commonly used nonsteroidal anti-inflammatory drug on spinal fusion during the immediate postoperation period in a established rabbit model of posterolateral lumbar spine fusion. The addition of recombinant bone morphogenetic protein-2 to the autograft bone was able to compensate for the inhibitory effect of ketorolac on bone formation. On the basis of these data, caution is urged in the routine use of nonsteroidal anti-inflammatory drugs for postoperation analgesia in patients undergoing spine arthrodesis.
Collapse
Affiliation(s)
- G J Martin
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | |
Collapse
|
38
|
Abstract
Many mammalian and avian species have been used as models to study fracture healing. The individual animals, based on their size and other characteristics, are used to show the effects of different interventions on healing bone. The models may have fractures created by closed or open means or osteotomies. Fracture healing can be evaluated through histologic, mechanical, chemical, or biological studies. Internal fixation, external skeletal fixation, or no fixation may be used in the experiments. Results of fracture healing studies among animal species may conflict even when the same model is used in each species. These differences may be explained by anatomic, biochemical, and gene expression differences. Evaluation of the animal model is an important consideration when applying the results of any particular study to humans. Consensus regarding fracture healing develops from agreement between results of animal models and human clinical studies.
Collapse
MESH Headings
- Animals
- Biomechanical Phenomena
- Bone and Bones/metabolism
- Bone and Bones/pathology
- Bone and Bones/physiopathology
- Bone and Bones/surgery
- Disease Models, Animal
- Fracture Fixation
- Fracture Fixation, Internal
- Fracture Healing/genetics
- Fracture Healing/physiology
- Fractures, Bone/metabolism
- Fractures, Bone/pathology
- Fractures, Bone/physiopathology
- Fractures, Bone/surgery
- Fractures, Closed/physiopathology
- Fractures, Closed/surgery
- Fractures, Open/physiopathology
- Fractures, Open/surgery
- Gene Expression
- Humans
- Osteotomy
Collapse
Affiliation(s)
- D M Nunamaker
- New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square 19348, USA
| |
Collapse
|
39
|
Orgill DP, Ehret FW, Regan JF, Glowacki J, Mulliken JB. Polyethylene glycol/microfibrillar collagen composite as a new resorbable hemostatic bone wax. JOURNAL OF BIOMEDICAL MATERIALS RESEARCH 1998; 39:358-63. [PMID: 9468043 DOI: 10.1002/(sici)1097-4636(19980305)39:3<358::aid-jbm3>3.0.co;2-i] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Although bone wax is effective at achieving hemostasis, it is nonresorbable, causes a foreign body reaction, and inhibits osteogenesis. We report development of a polyethylene glycol/microfibrillar collagen composite (PEG/MFC) that has inherent hemostatic qualities, is biodegradable, and is compatible with bone repair. PEG/MFC composite (n = 42) was placed in 5 mm cranial defects in New Zealand white rabbits. Hemostasis and healing were compared to unfilled defects (n = 32) and defects filled with standard bone wax (n = 10). Both PEG/MFC and bone wax handled well and stopped bleeding. The polyethylene glycol component was resorbed by 8 h, and the microfibrillar collagen was resorbed over 2 months, eliciting only a minor inflammatory response during the first month. Defects filled with the PEG/MFC composite showed similar amounts of bony regeneration as did unfilled control defects. At 4 weeks, healing bone accounted for 43 +/- 13% in those treated with PEG/MFC and 47 +/- 19% defect area in untreated holes. In contrast, less than 1% of the area was bone in defects filled with bone wax (p < 0.05). PEG/MFC composite provided excellent bony hemostasis and did not inhibit bone growth.
Collapse
Affiliation(s)
- D P Orgill
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
40
|
Keeting PE, Li CH, Whipkey DL, Thweatt R, Xu J, Murty M, Blaha JD, Graeber GM. 1,25-dihydroxyvitamin D3 pretreatment limits prostaglandin biosynthesis by cytokine-stimulated adult human osteoblast-like cells. J Cell Biochem 1998. [DOI: 10.1002/(sici)1097-4644(19980201)68:2<237::aid-jcb10>3.0.co;2-c] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
41
|
New generation of poly(ortho esters): synthesis, characterization, kinetics, sterilization and biocompatibility. J Control Release 1997. [DOI: 10.1016/s0168-3659(97)00050-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
42
|
Leunig M, Yuan F, Gerweck LE, Berk DA, Jain RK. Quantitative analysis of angiogenesis and growth of bone: effect of indomethacin exposure in a combined in vitro-in vivo approach. RESEARCH IN EXPERIMENTAL MEDICINE. ZEITSCHRIFT FUR DIE GESAMTE EXPERIMENTELLE MEDIZIN EINSCHLIESSLICH EXPERIMENTELLER CHIRURGIE 1995; 195:275-88. [PMID: 8578003 DOI: 10.1007/bf02576798] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nonsteroidal anti-inflammatory agents have been used experimentally and clinically to suppress a variety of physiological events, including angiogenesis and formation of bone. The exact mechanisms by which indomethacin alters skeletal tissue generation are unknown, due in part to methodological limitations. By the use of an organ culture assay and an animal model using intravital microscopy in mice bearing dorsal skinfold chambers, the effect of indomethacin on growth and angiogenesis of neonatal femora was characterized over 16 days. In both assays, femora significantly elongated with time (P < 0.05). The in vitro growth rate was more rapid than in vivo and dependent on the serum concentration, culture medium and age of mice. Although enhancing the serum content promoted cellular proliferation in organ culture, it dose-dependently suppressed femoral elongation, leading at 20% fetal calf serum to growth rates identical to those observed in vivo. Indomethacin supplementation (2 and 10 mg l-1) significantly accelerated longitudinal femoral growth in organ culture (P < 0.05), whereas in vivo indomethacin (2 mg kg-1) did not modulate either angiogenesis or elongation of bone. Our in vitro data propose a central role of serum in the regulation of bone formation. Although indomethacin altered femoral growth in vitro, our findings do not suggest that indomethacin suppresses angiogenesis or growth of bone in vivo. The complexity of physiological events in vivo may be obscuring a detectable effect.
Collapse
Affiliation(s)
- M Leunig
- Steele Laboratory, Department of Radiation Oncology, Massachusetts General Hospital, Boston 02114, USA
| | | | | | | | | |
Collapse
|
43
|
Solheim E, Pinholt EM, Andersen R, Bang G, Sudmann E. Local delivery of indomethacin by a polyorthoester inhibits reossification of experimental bone defects. JOURNAL OF BIOMEDICAL MATERIALS RESEARCH 1995; 29:1141-6. [PMID: 8567712 DOI: 10.1002/jbm.820290914] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Inhibition of orthotopic reossification after surgical removal of bone is sometimes indicated and may be accomplished by implantation of interpositional materials or by systemic administration of indomethacin. However, implantation of nonresorbable foreign material may induce a chronic inflammation and predispose to infections; and systemic administration of indomethacin may induce systemic adverse effects. We studied the effect of local delivery of indomethacin by a bioerodible polyorthoester on the reossification of segmental defects of the radius in rats. We divided 45 Wistar rats into three groups, A-C. A 3.5 mm-long middiaphyseal osteoperiosteal resection of the right radius was made in each rat. The defect was filled with 15 mg of polyorthoester with 5% indomethacin in group A and 15 mg of polyorthoester without drug in group B. No material was implanted in the defects in the group C rats. The rats were killed 50 days postoperatively. The mean area of the residual defects were greater in the defects with the polyorthoester with 5% indomethacin compared with defects with polyorthoester without drug or without implant as judged by computer-assisted area measurements on radiographs. By light microscopy, no inflammation was seen and only traces of the polyorthoester could be detected in the defects filled with the polyorthoester with or without indomethacin. The results of this study suggest that the polyorthoester may be used as a bioerodible system for local delivery of indomethacin to inhibit reossification of skeletal defects without tissue reaction, unabsorbed carrier, or systemic effects.
Collapse
Affiliation(s)
- E Solheim
- Institute for Surgical Research, University of Oslo, Norway
| | | | | | | | | |
Collapse
|
44
|
Sudmann B, Anfinsen OG, Bang G, Koppang R, Stølen SO, Koppang HS, Sudmann E. Assessment in rats of a new bioerodible bone-wax-like polymer. ACTA ORTHOPAEDICA SCANDINAVICA 1993; 64:336-9. [PMID: 8322594 DOI: 10.3109/17453679308993639] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Holes drilled in rats' skull, iliac crest, and tibia were filled with beeswax or with a new, wax-like, bioerodible polyorthoester (Alzamer). Empty drill-holes served as controls. In addition, beeswax and polyorthoester were deposited between the left and the right oblique abdominal muscles, respectively. In muscle, both the beeswax and polyorthoester elicited a transient foreign body reaction. The beeswax was not resorbed in bone or muscle, whereas the polyorthoester was. Bone healing was inhibited in the iliac crest and the tibiae filled with beeswax, whereas holes filled with polyorthoester healed readily.
Collapse
Affiliation(s)
- B Sudmann
- Surgical Research Laboratory, School of Dentistry, University of Bergen, Norway
| | | | | | | | | | | | | |
Collapse
|