Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 202
Filter
1.
Sci Transl Med ; 16(749): eabp8334, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38809966

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disease driven by gain-of-function variants in activin receptor-like kinase 2 (ALK2), the most common variant being ALK2R206H. In FOP, ALK2 variants display increased and dysregulated signaling through the bone morphogenetic protein (BMP) pathway resulting in progressive and permanent replacement of skeletal muscle and connective tissues with heterotopic bone, ultimately leading to severe debilitation and premature death. Here, we describe the discovery of BLU-782 (IPN60130), a small-molecule ALK2R206H inhibitor developed for the treatment of FOP. A small-molecule library was screened in a biochemical ALK2 binding assay to identify potent ALK2 binding compounds. Iterative rounds of structure-guided drug design were used to optimize compounds for ALK2R206H binding, ALK2 selectivity, and other desirable pharmacokinetic properties. BLU-782 preferentially bound to ALK2R206H with high affinity, inhibiting signaling from ALK2R206H and other rare FOP variants in cells in vitro without affecting signaling of closely related homologs ALK1, ALK3, and ALK6. In vivo efficacy of BLU-782 was demonstrated using a conditional knock-in ALK2R206H mouse model, where prophylactic oral dosing reduced edema and prevented cartilage and heterotopic ossification (HO) in both muscle and bone injury models. BLU-782 treatment preserved the normal muscle-healing response in ALK2R206H mice. Delayed dosing revealed a short 2-day window after injury when BLU-782 treatment prevented HO in ALK2R206H mice, but dosing delays of 4 days or longer abrogated HO prevention. Together, these data suggest that BLU-782 may be a candidate for prevention of HO in FOP.


Subject(s)
Disease Models, Animal , Myositis Ossificans , Ossification, Heterotopic , Animals , Myositis Ossificans/drug therapy , Myositis Ossificans/metabolism , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/prevention & control , Mice , Humans , Activin Receptors, Type II/metabolism , Activin Receptors, Type I/metabolism , Activin Receptors, Type I/antagonists & inhibitors , Signal Transduction/drug effects
2.
Cell Prolif ; 57(1): e13521, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37340819

ABSTRACT

Trauma-induced heterotopic ossification (HO) is a complex disorder after musculoskeletal injury and characterized by aberrant extraskeletal bone formation. Recent studies shed light on critical role of dysregulated osteogenic differentiation in aberrant bone formation. Krupel-like factor 2 (KLF2) and peroxisome proliferator-activated receptor gamma (PPARγ) are master adapter proteins that link cellular responses to osteogenesis; however, their roles and relationships in HO remain elusive. Using a murine burn/tenotomy model in vivo, we identified elevated KLF2 and reduced PPARγ levels in tendon stem/progenitor cells (TSPCs) during trauma-induced HO formation. Both KLF2 inhibition and PPARγ promotion reduced mature HO, whereas the effects of PPARγ promotion were abolished by KLF2 overexpression. Additionally, mitochondrial dysfunction and reactive oxygen species (ROS) production also increased after burn/tenotomy, and improvements in mitochondrial function (ROS scavenger) could alleviate HO formation, but were abolished by KLF2 activation and PPARγ suppression by affecting redox balance. Furthermore, in vitro, we found increased KLF2 and decreased PPARγ levels in osteogenically induced TSPCs. Both KLF2 inhibition and PPARγ promotion relieved osteogenesis by improving mitochondrial function and maintaining redox balance, and effects of PPARγ promotion were abolished by KLF2 overexpression. Our findings suggest that KLF2/PPARγ axis exerts regulatory effects on trauma-induced HO through modulation of mitochondrial dysfunction and ROS production in TSPCs by affecting redox balance. Targeting KLF2/PPARγ axis and mitochondrial dysfunction can represent attractive approaches to therapeutic intervention in trauma-induced HO.


Subject(s)
Burns , Mitochondrial Diseases , Ossification, Heterotopic , Mice , Animals , Osteogenesis , PPAR gamma , Reactive Oxygen Species , Ossification, Heterotopic/drug therapy , Burns/complications
3.
BMJ Open ; 13(12): e075502, 2023 12 18.
Article in English | MEDLINE | ID: mdl-38110382

ABSTRACT

INTRODUCTION: Exaggerated inflammatory response is one of the main mechanisms underlying heterotopic ossification (HO). It has been suggested that the antifibrinolytic drug tranexamic acid (TXA) can exert a significant anti-inflammatory effect during orthopaedic surgery. However, no prospective studies have yet investigated the effects of TXA on HO recurrence in patients following open elbow arthrolysis (OEA). METHODS AND ANALYSIS: Here, we present a protocol for a single-centre, randomised, double-blind, placebo-controlled trial to investigate the effectiveness of TXA on HO recurrence after OEA in a single hospital. A minimum sample size of 138 eligible and consenting participants randomised into treatment and control groups in a 1:1 manner will be included. Patients will receive 2 g of intravenous TXA (experimental group) or placebo (normal saline, control group) administered before skin incision. The primary outcome is HO recurrence rate within 12 months after surgery. The secondary outcomes are the serum immune-inflammatory cytokines including erythrocyte sedimentation rate, C reactive protein, interleukin (IL)-6, IL-1ß, IL-13 at the first and third day postoperatively, and elbow range of motion and functional score at 1.5, 6, 9 and 12 months after surgery. After completion of the trial, the results will be reported in accordance with the extensions of the Consolidated Standards of Reporting Trials Statement for trials. The results of this study should determine whether TXA can reduce the rates of HO occurrence after OEA. ETHICS AND DISSEMINATION: Ethical approval has been granted by the Medical Ethics Committee of the Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (reference number 2022-123-(1)). The results of this study will be disseminated through presentations at academic conferences and publication in peer-reviewed journals. TRIAL REGISTRATION NUMBER: ChiCTR2300068106.


Subject(s)
Antifibrinolytic Agents , Joint Diseases , Orthopedic Procedures , Ossification, Heterotopic , Tranexamic Acid , Humans , Tranexamic Acid/therapeutic use , Elbow/surgery , China , Antifibrinolytic Agents/therapeutic use , Double-Blind Method , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/etiology , Ossification, Heterotopic/surgery , Treatment Outcome , Randomized Controlled Trials as Topic
4.
BMC Med Res Methodol ; 23(1): 269, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37957586

ABSTRACT

BACKGROUND: The design of clinical trials in rare diseases is often complicated by a lack of real-world translational knowledge. Fibrodysplasia ossificans progressiva (FOP) is an ultra-rare genetic disorder characterized by skeletal malformations and progressive heterotopic ossification (HO). Palovarotene is a selective retinoic acid receptor gamma agonist. Here, we describe the methodology of three studies in the palovarotene clinical development program in FOP and discuss insights that could inform future research, including endpoint suitability and the impact of trial design. METHODS: PVO-1A-001 (NCT02322255) was a prospective, protocol-specified, longitudinal FOP natural history study (NHS). PVO-1A-201 (NCT02190747) was a randomized, double-blind, placebo-controlled phase II trial; PVO-1A-202 (NCT02279095) was its open-label extension. Trial designs, including treatment regimens and imaging assessments, were refined between PVO-1A-201 and PVO-|1A-202, and within PVO-1A-202, based on emerging data as the studies progressed. Palovarotene doses were administered using a flare-up treatment regimen (higher dose for 2/4 weeks, followed by lower dose for 4/≥8 weeks; from flare-up onset), with or without accompanying chronic (daily) treatment. Flare-up and disease progression outcomes were assessed, including incidence and volume of new HO during flare-ups and/or annually, as well as other clinical, patient-reported, and exploratory outcomes. Safety was monitored throughout all studies. RESULTS: Overall, 114 and 58 individuals with FOP were enrolled in the NHS and phase II trials, respectively. Results of the NHS and PVO-1A-201 were published in 2022; complete results of PVO-1A-202 will be publicly available in due course. Together the studies yielded important information on endpoint suitability, including that low-dose whole-body computed tomography was the optimum imaging modality for assessing HO progression annually and that long study durations are needed to detect substantial changes in functional and patient-reported outcomes. CONCLUSIONS: A flexible clinical development program is necessary for underexplored rare diseases to overcome the many challenges faced. Here, the NHS provided a longitudinal evaluation of FOP progression and interventional trials were based on emerging data. The studies described informed the design and endpoints implemented in the phase III MOVE trial (NCT03312634) and provide a foundation for future clinical trial development. TRIAL REGISTRATION: NCT02322255 (registered 23/12/2014); NCT02190747 (registered 15/07/2014); NCT02279095 (registered 30/10/2014).


Subject(s)
Myositis Ossificans , Ossification, Heterotopic , Humans , Myositis Ossificans/drug therapy , Ossification, Heterotopic/drug therapy , Prospective Studies , Rare Diseases , Randomized Controlled Trials as Topic , Clinical Trials, Phase II as Topic
5.
Semin Arthritis Rheum ; 63: 152306, 2023 12.
Article in English | MEDLINE | ID: mdl-37976811

ABSTRACT

INTRODUCTION: Ectopic calcifications (ECs) and heterotopic ossifications (HOs) form in non-mineralized tissues, most often in subcutaneous and muscular areas. Local and systemic complications can cause severe disability. Systemic administration of sodium thiosulfate (STS) gives promising results but is difficult to use in clinical practice. OBJECTIVE: Evaluation of the efficacy and safety of topical STS in ECs and HOs. METHODS: Retrospective analysis of the CATSS-O registry that included patients receiving topical STS 25 % prepared by the pharmacy of Limoges hospital during 2014-2020. The efficacy of STS was assessed by imaging (radiography or CT) after at least 6 months' treatment. RESULTS: Among 126 patients who received STS 25 %, 35 had complete clinical and radiographic data for analysis (28 with ECs and 7 with HOs; 18 children [mean age 8.9 years, range 1.5-16], 17 adults [mean age 52.4 years, range 24-90]). Calcifications or ossifications were due to dermatomyositis (8 children, 6 adults), systemic scleroderma (6 adults) or pseudo-hypoparathyroidism 1A (7 children). They were single (37.1 %) or multiple (62.9 %). Treated regions were in the lower limbs (31.4 %), upper limbs (37.1 %) or both (28.6 %) and the axial region (2.9 %). Topical STS was clinically effective in 9/28 (32.1 %) patients with ECs and 2/7 (28.6 %) children with HOs. Three patients experienced complete disappearance of their calcifications. Response for ECs was better in children than adults (54.5% vs 17.6 %, p = 0.035). Topical STS was well tolerated. CONCLUSION: Local STS seems effective for ossifications, particularly pediatric calcifications or ossifications. Randomized and experimental studies are needed to confirm this observation and to identify the underlying mechanisms.


Subject(s)
Calcinosis , Ossification, Heterotopic , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Humans , Infant , Middle Aged , Young Adult , Calcinosis/diagnostic imaging , Calcinosis/drug therapy , Calcinosis/etiology , Ossification, Heterotopic/diagnostic imaging , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/complications , Osteogenesis , Retrospective Studies
6.
ACS Nano ; 17(23): 24187-24199, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37983164

ABSTRACT

Ankylosing spondylitis (AS) is a chronic systemic inflammatory disease that leads to serious spinal deformity and ankylosis. Persistent inflammation and progressive ankylosis lead to loss of spinal flexibility in patients with AS. Tetrahedral framework nucleic acids (tFNAs) have emerged as a one kind of nanomaterial composed of four specially designed complementary DNA single strands with outstanding biological properties. Results from in vivo experiments demonstrated that tFNAs treatment could inhibit inflammatory responses and heterotopic ossification to halt disease progression. In vitro, tFNAs were proved to influence the biological behavior of AS primary chondrocytes and inhibit the secretion of pro-inflammatory cytokines through interleukin-17 pathway. The osteogenic process of chondrocytes was as well inhibited at the transcriptional level to regulate the expression of related proteins. Therefore, we believe tFNAs had a strong therapeutic effect and could serve as a nonsurgical remedy in the future to help patients suffering from AS.


Subject(s)
Nucleic Acids , Ossification, Heterotopic , Spondylitis, Ankylosing , Humans , Spondylitis, Ankylosing/drug therapy , Spondylitis, Ankylosing/surgery , Interleukin-17 , Nucleic Acids/pharmacology , Ossification, Heterotopic/drug therapy , Inflammation/drug therapy
7.
J Cell Mol Med ; 27(22): 3491-3502, 2023 11.
Article in English | MEDLINE | ID: mdl-37605888

ABSTRACT

Heterotopic ossification (HO) is a pathological process that often occurs in soft tissues following severe trauma. There is no effective therapy for HO. The BMP signalling pathway plays an essential role in the pathogenesis of HO. Our previous study showed that AMPK negatively regulates the BMP signalling pathway and osteogenic differentiation. The present study aims to study the effect of two AMPK activators berberine and aspirin on osteogenic differentiation and HO induced by traumatic injury. The effects of two AMPK activators, berberine and aspirin, on BMP signalling and osteogenic differentiation were measured by western blot, ALP and Alizarin red S staining in C3H10T1/2 cells. A mouse model with Achilles tenotomy was employed to assess the effects of berberine and aspirin on HO using µCT and histological analysis. First, our study showed that berberine and aspirin inhibited phosphorylation of Smad1/5 induced by BMP6 and the inhibition was attributed to the down-regulation of ALK2 expression. Second, the combination of berberine and aspirin yielded more potent effects on BMP signalling. Third, we further found that there was an additive effect of berberine and aspirin combination on osteogenic differentiation. Finally, we found that berberine and aspirin blocked trauma-induced ectopic bone formation in mice, which may be through suppression of phosphorylation of Smad1/5 in injured tissues. Collectively, these findings indicate that berberine and aspirin inhibit osteogenic differentiation in C3H10T1/2 cells and traumatic HO in mice, possibly through the down-regulation of the BMP signalling pathway. Our study sheds a light on prevention and treatment of traumatic HO using AMPK pharmacological activators berberine and aspirin.


Subject(s)
Berberine , Ossification, Heterotopic , Mice , Animals , Berberine/pharmacology , Osteogenesis , AMP-Activated Protein Kinases , Aspirin/pharmacology , Cell Differentiation , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/etiology , Ossification, Heterotopic/prevention & control
8.
FASEB J ; 37(7): e23057, 2023 07.
Article in English | MEDLINE | ID: mdl-37367700

ABSTRACT

Heterotopic ossification occurs as a pathological ossification condition characterized by ectopic bone formation within soft tissues following trauma. Vascularization has long been established to fuel skeletal ossification during tissue development and regeneration. However, the feasibility of vascularization as a target of heterotopic ossification prevention remained to be further clarified. Here, we aimed to identify whether verteporfin as a widely used FDA-approved anti-vascularization drug could effectively inhibit trauma-induced heterotopic ossification formation. In the current study, we found that verteporfin not only dose dependently inhibited the angiogenic activity of human umbilical vein endothelial cells (HUVECs) but also the osteogenic differentiation of tendon stem cells (TDSCs). Moreover, YAP/ß-catenin signaling axis was downregulated by the verteporfin. Application of lithium chloride, an agonist of ß-catenin, recovered TDSCs osteogenesis and HUVECs angiogenesis that was inhibited by verteporfin. In vivo, verteporfin attenuated heterotopic ossification formation by decelerating osteogenesis and the vessels densely associated with osteoprogenitors formation, which could also be readily reversed by lithium chloride, as revealed by histological analysis and Micro-CT scan in a murine burn/tenotomy model. Collectively, this study confirmed the therapeutic effect of verteporfin on angiogenesis and osteogenesis in trauma-induced heterotopic ossification. Our study sheds light on the anti-vascularization strategy with verteporfin as a candidate treatment for heterotopic ossification prevention.


Subject(s)
Achilles Tendon , Ossification, Heterotopic , Mice , Humans , Animals , Osteogenesis , Achilles Tendon/pathology , Verteporfin/pharmacology , beta Catenin , Endothelial Cells/pathology , Lithium Chloride/pharmacology , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/etiology , Ossification, Heterotopic/prevention & control
9.
Inflammation ; 46(4): 1414-1429, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37115368

ABSTRACT

Heterotopic ossification (HO) is a pathological condition that occurs in soft tissues following severe trauma. The exact pathogenesis of HO remains unclear. Studies have shown that inflammation predisposes patients to the development of HO and triggers ectopic bone formation. Macrophages are crucial mediators of inflammation and are involved in HO development. The present study investigated the inhibitory effect and underlying mechanism of metformin on macrophage infiltration and traumatic HO in mice. Our results found that abundant levels of macrophages were recruited to the injury site during early HO progression and that early administration of metformin prevented traumatic HO in mice. Furthermore, we found that metformin attenuated macrophage infiltration and the NF-κB signaling pathway in injured tissue. The monocyte-to-macrophage transition in vitro was suppressed by metformin and this event was mediated by AMPK. Finally, we showed that inflammatory mediator's regulation by macrophages targeted preosteoblasts, leading to elevated BMP signaling, and osteogenic differentiation and driving HO formation, and this effect was blocked after the activation of AMPK in macrophages. Collectively, our study suggests that metformin prevents traumatic HO by inhibiting of NF-κB signaling in macrophages and subsequently attenuating BMP signaling and osteogenic differentiation in preosteoblasts. Therefore, metformin may serve as a therapeutic drug for traumatic HO by targeting NF-κB signaling in macrophages.


Subject(s)
Metformin , Ossification, Heterotopic , Mice , Animals , NF-kappa B/metabolism , Metformin/pharmacology , Metformin/therapeutic use , AMP-Activated Protein Kinases/metabolism , Osteogenesis , Macrophages/metabolism , Signal Transduction , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/etiology , Ossification, Heterotopic/prevention & control
10.
Adv Sci (Weinh) ; 10(15): e2207224, 2023 05.
Article in English | MEDLINE | ID: mdl-36970815

ABSTRACT

Heterotopic ossification (HO) is a double-edged sword. Pathological HO presents as an undesired clinical complication, whereas controlled heterotopic bone formation by synthetic osteoinductive materials shows promising therapeutic potentials for bone regeneration. However, the mechanism of material-induced heterotopic bone formation remains largely unknown. Early acquired HO being usually accompanied by severe tissue hypoxia prompts the hypothesis that hypoxia caused by the implantation coordinates serial cellular events and ultimately induces heterotopic bone formation in osteoinductive materials. The data presented herein shows a link between hypoxia, macrophage polarization to M2, osteoclastogenesis, and material-induced bone formation. Hypoxia inducible factor-1α (HIF-1α), a crucial mediator of cellular responses to hypoxia, is highly expressed in an osteoinductive calcium phosphate ceramic (CaP) during the early phase of implantation, while pharmacological inhibition of HIF-1α significantly inhibits M2 macrophage, subsequent osteoclast, and material-induced bone formation. Similarly, in vitro, hypoxia enhances M2 macrophage and osteoclast formation. Osteoclast-conditioned medium enhances osteogenic differentiation of mesenchymal stem cells, such enhancement disappears with the presence of HIF-1α inhibitor. Furthermore, metabolomics analysis reveals that hypoxia enhances osteoclastogenesis via the axis of M2/lipid-loaded macrophages. The current findings shed new light on the mechanism of HO and favor the design of more potent osteoinductive materials for bone regeneration.


Subject(s)
Bone Substitutes , Ossification, Heterotopic , Humans , Osteogenesis , Bone Substitutes/therapeutic use , Macrophages , Hypoxia , Ossification, Heterotopic/drug therapy , Lipids/therapeutic use
11.
J Bone Miner Res ; 38(3): 381-394, 2023 03.
Article in English | MEDLINE | ID: mdl-36583535

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is an ultra-rare, severely disabling genetic disorder of progressive heterotopic ossification (HO). The single-arm, open-label, phase 3 MOVE trial (NCT03312634) assessed efficacy and safety of palovarotene, a selective retinoic acid receptor gamma agonist, in patients with FOP. Findings were compared with FOP natural history study (NHS; NCT02322255) participants untreated beyond standard of care. Patients aged ≥4 years received palovarotene once daily (chronic: 5 mg; flare-up: 20 mg for 4 weeks, then 10 mg for ≥8 weeks; weight-adjusted if skeletally immature). The primary endpoint was annualized change in new HO volume versus NHS participants (by low-dose whole-body computed tomography [WBCT]), analyzed using a Bayesian compound Poisson model (BcPM) with square-root transformation. Twelve-month interim analyses met futility criteria; dosing was paused. An independent Data Monitoring Committee recommended trial continuation. Post hoc 18-month interim analyses utilized BcPM with square-root transformation and HO data collapsed to equalize MOVE and NHS visit schedules, BcPM without transformation, and weighted linear mixed-effects (wLME) models, alongside prespecified analysis. Safety was assessed throughout. Eighteen-month interim analyses included 97 MOVE and 101 NHS individuals with post-baseline WBCT. BcPM analyses without transformation showed 99.4% probability of any reduction in new HO with palovarotene versus NHS participants (with transformation: 65.4%). Mean annualized new HO volume was 60% lower in MOVE versus the NHS. wLME results were similar (54% reduction fitted; nominal p = 0.039). All palovarotene-treated patients reported ≥1 adverse event (AE); 97.0% reported ≥1 retinoid-associated AE; 29.3% reported ≥1 serious AE, including premature physeal closure (PPC)/epiphyseal disorder in 21/57 (36.8%) patients aged <14 years. Post hoc computational analyses using WBCT showed decreased vertebral bone mineral density, content, and strength, and increased vertebral fracture risk in palovarotene-treated patients. Thus, post hoc analyses showed evidence for efficacy of palovarotene in reducing new HO in FOP, but high risk of PPC in skeletally immature patients. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Myositis Ossificans , Ossification, Heterotopic , Humans , Myositis Ossificans/drug therapy , Bayes Theorem , Ossification, Heterotopic/drug therapy , Pyrazoles/therapeutic use
12.
J Shoulder Elbow Surg ; 31(10): 2157-2163, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35872167

ABSTRACT

BACKGROUND: The aim of this study was to assess the efficacy of 3 weeks of indomethacin, a nonselective nonsteroidal anti-inflammatory drug, in comparison to 1 week of meloxicam as prophylaxis for heterotopic ossifications (HOs) after distal biceps tendon repair. METHODS: A single-center retrospective study was performed on 78 patients undergoing distal biceps tendon repair between 2008 and 2019. From 2008 to 2016, patients received meloxicam 15 mg daily for the period of 1 week as usual care. From 2016 onward, the standard protocol was changed to indomethacin 25 mg 3 times daily for 3 weeks. All patients underwent a single-incision repair with a cortical button technique. The postoperative rehabilitation protocol was similar for all patients. The postoperative radiographs at 8-week follow-up were assessed blindly by 7 independent assessors. If HOs were present, it was classified according to the Ilahi-Gabel classification for size and according to the Gärtner-Heyer classification for density. Statistical analysis was performed to analyze the difference in HO between the patients who were treated with indomethacin and with meloxicam. RESULTS: Seventy-eight patients, with a mean age of 48.8 years (range 30-72) were included. The mean follow-up after surgery was 12 months (range 2-45). Indomethacin (21 days, 25 mg 3 times per day) was prescribed to 26 (33%) patients. The 52 other patients (67%) were prescribed meloxicam 15 mg daily for 7 days. HOs were seen in 19 patients 8 weeks postoperatively. Five of 26 patients treated with indomethacin developed HO, and 14 of 52 patients treated with meloxicam developed HO (P = .5). Two patients had symptomatic HO with minor restrictions in movement; neither patient was treated with indomethacin. Significantly more HOs were seen in patients with a longer time from injury to surgery (P = .01) The intraclass correlation score for reliability between assessors for HO scoring on postoperative radiographs was good to excellent for both classifications. CONCLUSION: In this study, HOs were seen in 24% of postoperative radiographs. Three weeks of indomethacin was not superior to meloxicam for 1 week for the prevention of HO after single-incision distal biceps tendon repair.


Subject(s)
Ossification, Heterotopic , Tendon Injuries , Adult , Aged , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Humans , Indomethacin/therapeutic use , Meloxicam/therapeutic use , Middle Aged , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/etiology , Ossification, Heterotopic/prevention & control , Reproducibility of Results , Retrospective Studies , Rupture/surgery , Tendon Injuries/surgery , Tendons
13.
J Bone Miner Res ; 37(10): 1891-1902, 2022 10.
Article in English | MEDLINE | ID: mdl-35854638

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is an ultra-rare genetic disorder characterized by progressive heterotopic ossification (HO), often heralded by flare-ups, leading to reduced movement and life expectancy. This placebo-controlled, double-blind trial (NCT02190747) evaluated palovarotene, an orally bioavailable selective retinoic acid receptor gamma agonist, for prevention of HO in patients with FOP. Patients experiencing a flare-up were enrolled in two cohorts: (1) patients ≥15 years were randomized 3:1 to palovarotene 10/5 mg (weeks 1-2/3-6) or placebo; (2) patients ≥6 years were randomized 3:3:2 to palovarotene 10/5 mg, palovarotene 5/2.5 mg (weeks 1-2/3-6), or placebo. Cohort data were pooled. The primary endpoint was the proportion of responders (no/minimal new HO at flare-up body region by plain radiograph) at week 6. Change from baseline in HO volume and new HO incidence were assessed by computed tomography (CT) at week 12. Tissue edema was assessed by magnetic resonance imaging (MRI) or ultrasound. Forty patients (aged 7-53 years) were enrolled (placebo: n = 10; palovarotene 5/2.5 mg: n = 9; palovarotene 10/5 mg: n = 21). Disease history was similar between groups. In the per-protocol population, the proportion of responders at week 6 by plain radiograph was 100% with palovarotene 10/5 mg; 88.9% with palovarotene 5/2.5 mg; 88.9% with placebo (Cochran-Armitage trend test: p = 0.17). At week 12, the proportions were 95.0% with palovarotene 10/5 mg; 88.9% with palovarotene 5/2.5 mg; 77.8% with placebo (Cochran-Armitage trend test: p = 0.15). Week 12 least-squares mean (LSmean) new HO volume, assessed by CT, was 3.8 × 103  mm3 with palovarotene 10/5 mg; 1.3 × 103  mm3 with palovarotene 5/2.5 mg; 18.0 × 103  mm3 with placebo (pairwise tests versus placebo: p ≤ 0.12). Palovarotene was well-tolerated. No patients discontinued treatment or required dose reduction; one patient had dose interruption due to elevated lipase. Although these findings were not statistically significant, they support further evaluation of palovarotene for prevention of HO in FOP in larger studies. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Myositis Ossificans , Ossification, Heterotopic , Stilbenes , Humans , Myositis Ossificans/drug therapy , Ossification, Heterotopic/diagnostic imaging , Ossification, Heterotopic/drug therapy , Pyrazoles/therapeutic use , Stilbenes/therapeutic use
14.
Eur J Pharmacol ; 928: 175109, 2022 Aug 05.
Article in English | MEDLINE | ID: mdl-35738451

ABSTRACT

Heterotopic ossification (HO) is the formation of bony tissues in the extraskeletal system. To date, no effective therapy has been developed for the treatment of HO, although increasing evidences have shown that inhibition of TGF-ß signaling has potential as a new therapeutic approach for attenuating HO progression. Results from previous clinical trials have demonstrated that patients with malignant tumors exhibit excellent tolerability to Galunisertib, a TGF-ß receptor I kinase inhibitor. However, its therapeutic potential in preventing HO and inhibitory effect on osteogenesis remain unclear. In this study, we demonstrated that intragastrical administration of Galunisertib, at a concentration as low as 10 mg/kg, was not only fairly effective in preventing HO development in a dose-dependent manner, but also generated a non-toxic response in a novel Achilles tendon puncture-induced traumatic HO model in mice. Moreover, Galunisertib treatment in the early phases of HO development, including the inflammatory and chondrogenic period, resulted in better therapeutic effects instead of eliminating already formed bony tissues. Mechanistically, Galunisertib suppressed the osteogenic differentiation capacity of tendon-derived stem cells (TDSCs) by interfering with the Smad2/3 signaling pathway, blocking the phosphorylation of Smad2/3 translocated from cytoplasm into the nucleus to regulate the expression of both osteogenesis-related transcription factors and related proteins. Results from in vivo experiments further validated Galunisertib's effect on HO attenuation, by intercepting the TGF-ß/Smad2/3 signaling pathway. In conclusion, our findings demonstrated Galunisertib's potential as a prophylactic drug for the treatment of traumatic HO or other related diseases triggered by over-expressed TGF-ß.


Subject(s)
Ossification, Heterotopic , Osteogenesis , Animals , Mice , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/pathology , Pyrazoles , Quinolines , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/pharmacology
15.
Expert Opin Pharmacother ; 23(10): 1195-1203, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35698796

ABSTRACT

INTRODUCTION: Heterotopic ossification (HO) of the hip joint may happen accompanying skeletal muscle trauma or surgical procedures. The pharmacological prophylaxis of heterotopic ossification (HO) following total hip arthroplasty (THA) is debated. AREAS COVERED: This expert opinion aims to systematically investigate the efficacy of current pharmacological options as prophylaxis for HO following THA. EXPERT OPINION: The current evidence identified celecoxib, naproxen, and diclofenac as best option for the prevention of HO in patients who undergo primary THA. The most appropriate pharmacotherapy for the prevention of HO is still debated and should be customized according to patients' comorbidities and medical history. For patients with cardiovascular comorbidities, naproxen, or diclofenac should be considered along with proton pump inhibitors to prevent gastrointestinal complications. For patients with history of gastrointestinal disease, celecoxib can be recommended. These conclusions must be considered within the limitations of the present investigation. Between studies, heterogeneities in the administration protocols were evident. In some RCTs, the length of the follow-up was shorter than 12 months. The current clinical practice would benefit of high-quality recommendations and the development of the shared official guidelines.


Subject(s)
Arthroplasty, Replacement, Hip , Ossification, Heterotopic , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthroplasty, Replacement, Hip/adverse effects , Celecoxib/therapeutic use , Diclofenac/therapeutic use , Expert Testimony , Humans , Naproxen/therapeutic use , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/etiology , Ossification, Heterotopic/prevention & control , Postoperative Complications/drug therapy
16.
J Biomater Appl ; 36(8): 1458-1468, 2022 03.
Article in English | MEDLINE | ID: mdl-35043696

ABSTRACT

Traumatic heterotopic ossification (THO) is a serious and common clinical post-traumatic complication for which there is no effective and safe drug treatment. Routine administration of nonsteroidal anti-inflammatory drugs (NSAIDs) after injury is extensively used approach for THO. However, serious adverse events can occur in the event of an overdose of NSAIDs. In our study, we have developed a poly(lactic acid-co-glycolic acid) (PLGA) microsphere by emulsifying solvent volatilization for the prolonged slow delivery of celecoxib (CLX). Three groups of celecoxib-poly(lactic acid-co-glycolic acid) microspheres (CLX-PLGA MPs) were prepared with particle sizes of 3.75±1.28 µm, 49.56±17.15 µm, and 94.98±42.53 µm. Meanwhile, related parameters of microspheres in each group were studied: drug loading (DL), encapsulation rate (EE), and slow-release behavior. The DL and EE of the 3 CLX-PLGA MPs did not vary significantly, and subsequently, we selected the second drug loading microspheres with a retardation period of about 70 days for subsequent experiments. Moreover, cellular and animal experiments suggest that the microspheres are biocompatible and can be safely applied to localized trauma tissue. Finally, it is demonstrated that CLX-PLGA MPs have an effect on inhibiting the osteogenic differentiation of bone marrow mesenchymal stem cells and have the potential to inhibit ectopic bone formation of the THO model in Sprague-Dawley rat. Therefore, this study suggests that CLX-PLGA MPs are expected to be applied topically in the early post-traumatic period to prevent the development of THO.


Subject(s)
Ossification, Heterotopic , Polyglycolic Acid , Animals , Celecoxib/therapeutic use , Glycolates , Lactic Acid , Microspheres , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/etiology , Osteogenesis , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley
17.
J Oral Maxillofac Surg ; 80(5): 869-888, 2022 05.
Article in English | MEDLINE | ID: mdl-35032438

ABSTRACT

PURPOSE: The purpose of this study was to create a treatment protocol for cases of heterotopic ossification (HO) of the temporomandibular joint (TMJ), particularly those refractory to current TMJ HO protocols. In addition, we demonstrate the success of this protocol on a unique case of recurrent HO that failed multiple TMJ HO protocols in the setting of an improvised explosive device (IED) blast in a wounded warrior. METHODS: An electronic literature review was conducted via PubMed and Web of Science. Twenty-five studies were identified to provide supporting evidence for a proposed, up-to-date protocol for the treatment of refractory TMJ HO. The authors present a case report of a wounded warrior with HO ankylosis of bilateral TMJs in the setting of IED blast and demonstrate successful use of our surgical and pharmacotherapeutic protocol. RESULTS: Based on the literature review, our proposed protocol consists of pharmacotherapy with celecoxib and etidronate, with weekly forced dilation (brisement) and home physical therapy with the TheraBite Jaw Motion Rehab System. Surgically, the TMJ should be treated with two-stage reconstruction using initial polymethyl methacrylate spacers and subsequent total joint replacement with custom prostheses, fat grafting, and 3-dimensional-navigated total resection of HO. This protocol was successfully utilized in our patient's refractory HO ankylosed TMJ secondary to IED blast, and the patient's maximal incisal opening was regained and has remained stable 2 years after surgery without recurrent HO. CONCLUSIONS: Our method for treatment in this case deviated from the standard TMJ Concepts HO protocol in that it included multimodal pharmacotherapy with celecoxib and etidronate. Based on our literature review and experience, we advise that clinicians utilize our protocol for the management of all craniofacial HO cases, particularly in cases of recurrent HO that fail conventional therapies and/or involving high-order blast trauma.


Subject(s)
Ossification, Heterotopic , Temporomandibular Joint Disorders , Celecoxib/therapeutic use , Combined Modality Therapy , Etidronic Acid/therapeutic use , Humans , Joint Prosthesis , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/surgery , Temporomandibular Joint Disorders/drug therapy , Temporomandibular Joint Disorders/surgery
18.
Wounds ; 34(11): E112-E114, 2022 11.
Article in English | MEDLINE | ID: mdl-36608841

ABSTRACT

INTRODUCTION: HO with concurrent chronic osteomyelitis is extremely rare. To the authors' knowledge, this is the first case in the English-language literature with wound infection and mature HO with chronic osteomyelitis caused by mixed infection of Pasteurella canis, Peptoniphilus coxii, Peptostreptococcus canis, and Fusobacterium nucleatum following licking of a wound by a domesticated dog. CASE REPORT: A 49-year-old female with a painful, swollen, and purulent wound with bone exposure, measuring 2.5 cm × 1.5 cm, on the right leg was referred after an unsuccessful 3-month treatment regimen for an open wound resulting from a motorcycle accident. The patient's dog licked the wound several times 1 week after the accident. Sequestrectomy and debridement were performed after a 3-week OPD treatment. Postoperative treatment included NPWT applied for 6 days, 1 week of open wound care, STSG 2 weeks after the first operation, and IV antibiotics for 3 weeks. Pathologic examination was positive for HO with chronic osteomyelitis. The patient was discharged 3 weeks after admission under stable condition followed by OPD treatment. Wound healing was achieved 2 months after discharge. CONCLUSIONS: Repeated licking of the patient's wound by her dog caused the colonization of pathogens from the dog's saliva, and inappropriate wound care by the patient herself resulted in HO with chronic osteomyelitis, which was successfully treated with a regimen of NPWT, open wound care, STSG, and IV antibiotics.


Subject(s)
Ossification, Heterotopic , Osteomyelitis , Wound Infection , Female , Dogs , Animals , Anti-Bacterial Agents/therapeutic use , Wound Healing , Osteomyelitis/complications , Osteomyelitis/therapy , Wound Infection/drug therapy , Ossification, Heterotopic/complications , Ossification, Heterotopic/drug therapy
19.
Cells ; 10(11)2021 11 19.
Article in English | MEDLINE | ID: mdl-34831466

ABSTRACT

Retinoids are metabolic derivatives of vitamin A and regulate the function of many tissues and organs both prenatally and postnatally. Active retinoids, such as all trans-retinoic acid, are produced in the cytoplasm and then interact with nuclear retinoic acid receptors (RARs) to up-regulate the transcription of target genes. The RARs can also interact with target gene response elements in the absence of retinoids and exert a transcriptional repression function. Studies from several labs, including ours, showed that chondrogenic cell differentiation and cartilage maturation require (i) the absence of retinoid signaling and (ii) the repression function by unliganded RARs. These and related insights led to the proposition that synthetic retinoid agonists could thus represent pharmacological agents to inhibit heterotopic ossification (HO), a process that recapitulates developmental skeletogenesis and involves chondrogenesis, cartilage maturation, and endochondral ossification. One form of HO is acquired and is caused by injury, and another severe and often fatal form of it is genetic and occurs in patients with fibrodysplasia ossificans progressiva (FOP). Mouse models of FOP bearing mutant ACVR1R206H, characteristic of most FOP patients, were used to test the ability of the retinoid agonists selective for RARα and RARγ against spontaneous and injury-induced HO. The RARγ agonists were found to be most effective, and one such compound, palovarotene, was selected for testing in FOP patients. The safety and effectiveness data from recent and ongoing phase II and phase III clinical trials support the notion that palovarotene may represent a disease-modifying treatment for patients with FOP. The post hoc analyses showed substantial efficacy but also revealed side effects and complications, including premature growth plate closure in some patients. Skeletally immature patients will need to be carefully weighed in any future regulatory indications of palovarotene as an important therapeutic option in FOP.


Subject(s)
Ossification, Heterotopic/drug therapy , Retinoids/agonists , Retinoids/therapeutic use , Animals , Chondrogenesis/drug effects , Clinical Trials as Topic , Humans , Ossification, Heterotopic/metabolism , Receptors, Retinoic Acid/metabolism , Retinoids/pharmacology , Signal Transduction/drug effects
20.
Biochem Biophys Res Commun ; 573: 171-178, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34419763

ABSTRACT

Rapamycin (RAPA), which was first described as an anti-fungal agent, is a potent immunosuppressant that suppresses tumors and inhibits the mTOR signaling pathway. Heterotopic ossification (HO) is abnormal bone formation outside the skeletal system (e.g., in muscles, tendons, articular capsules and other soft tissues), often due to trauma or injury. There are currently no drugs available to treat traumatic HO, largely due to limited understanding of the disease. In this study, we focused on the role of oxidative stress (OS) in the early stage of traumatic HO, and explored the underlying mechanism of traumatic HO by using RAPA to specifically inhibit the mTOR pathway, which is known to play a role in the pathogenesis of HO. To assess the effects of RAPA in traumatic HO, we used an NSE-BMP4 transgenic mouse model that develops ossification in response to traumatic injury and intramuscular injection of cardiotoxin to initiate injury. These mice were then treated with RAPA or vehicle intraperitoneally every other day for 2 weeks. Our results demonstrate that RAPA can inhibit HO through a number of different mechanisms. We show that OS and a strong inflammatory response contribute to the hypoxia associated with the early stages of HO, and that RAPA inhibits these responses. Furthermore, RAPA reduces the vascularization triggered by mTOR signaling that leads to HO formation. Therefore, we believe that RAPA could be an effective treatment for the early stages of HO.


Subject(s)
Immunosuppressive Agents/pharmacology , Ossification, Heterotopic/drug therapy , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/pathology , Oxidative Stress/drug effects , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...