Advertisement for orthosearch.org.uk
Results 1 - 20 of 160
Results per page:
Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_8 | Pages 68 - 68
11 Apr 2023
Turnbull G Picard F Clarke J Shu W
Full Access

As arthroplasty demand grows worldwide, the need for a novel cost-effective treatment option for articular cartilage (AC) defects tailored to individual patients has never been greater. 3D bioprinting can deposit patient cells and other biomaterials in user-defined patterns to build tissue constructs from the “bottom-up,” potentially offering a new treatment for AC defects. The aim of this research was to create bioinks that can be injected or 3D bioprinted to aid osteochondral defect repair using human cells. Novel composite bioinks were created by mixing different ratios of methacrylated alginate (AlgMA) with methacrylated gelatin (GelMA). Chondrocytes or mesenchymal stem cells (MSCs) were then encapsulated in the bioinks and 3D bioprinted using a custom-built extrusion bioprinter. UV and double-ionic (BaCl2 and CaCl2) crosslinking was deployed following bioprinting to strengthen bioink stability in culture. Chondrocyte and MSC spheroids were also produced via 3D culture and then bioprinted to accelerate cell growth and development of ECM in bioprinted constructs. Excellent viability of chondrocytes and MSCs was seen following bioprinting (>95%) and maintained in culture over 28 days, with accelerated cell growth seen with inclusion of MSC or chondrocyte spheroids in bioinks (p<0.05). Bioprinted 10mm diameter constructs maintained shape in culture over 28 days, whilst construct degradation rates and mechanical properties were improved with addition of AlgMA (p<0.05). Composite bioinks were also injected into in vitro osteochondral defects (OCDs) and crosslinked in situ, with maintained cell viability and repair of osteochondral defects seen over a 14-day period. In conclusion we developed novel composite AlgMA/GelMA bioinks that can be triple-crosslinked, facilitating dense chondrocyte and MSC growth in constructs following 3D bioprinting. The bioink can be injected or 3D bioprinted to successfully repair in vitro OCDs, offering hope for a new approach to treating AC defects


Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_8 | Pages 108 - 108
11 Apr 2023
Turnbull G Picard F Clarke J Li B Shu W
Full Access

As arthroplasty demand grows worldwide, the need for a novel cost-effective treatment option for articular cartilage (AC) defects tailored to individual patients has never been greater. 3D bioprinting can deposit patient cells and other biomaterials in user-defined patterns to build tissue constructs from the “bottom-up,” potentially offering a new treatment for AC defects. The aim of this research was to create bioinks that can be injected or 3D bioprinted to aid osteochondral defect repair using human cells. Novel composite bioinks were created by mixing different ratios of methacrylated alginate (AlgMA) with methacrylated gelatin (GelMA). Chondrocytes or mesenchymal stem cells (MSCs) were then encapsulated in the bioinks and 3D bioprinted using a custom-built extrusion bioprinter. UV and double-ionic (BaCl2 and CaCl2) crosslinking was deployed following bioprinting to strengthen bioink stability in culture. Chondrocyte and MSC spheroids were also bioprinted to accelerate cell growth and development of ECM in bioprinted constructs. Excellent viability of chondrocytes and MSCs was seen following bioprinting (>95%) and maintained in culture over 28 days, with accelerated cell growth seen with inclusion of MSC or chondrocyte spheroids in bioinks (p<0.05). Bioprinted 10mm diameter constructs maintained shape in culture over 28 days, whilst construct degradation rates and mechanical properties were improved with addition of AlgMA (p<0.05). Composite bioinks were also injected into in vitro osteochondral defects (OCDs) and crosslinked in situ, with maintained cell viability and repair of osteochondral defects seen over a 14-day period. In conclusion we developed novel composite AlgMA/GelMA bioinks that can be triple-crosslinked, facilitating dense chondrocyte and MSC growth in constructs following 3D bioprinting. The bioink can be injected or 3D bioprinted to successfully repair in vitro OCDs, offering hope for a new approach to treating AC defects


Orthopaedic Proceedings
Vol. 103-B, Issue SUPP_16 | Pages 23 - 23
1 Dec 2021
Boyd A Rodzen K Morton M Acheson J McIlhagger A Morgan R Tormey D Dave F Sherlock R Meenan B
Full Access

Abstract. INTRODUCTION. Polyetheretherketone (PEEK) is a high-performance thermoplastic polymer which has found increasing application in orthopaedic implant devices and has a lot of promise for ‘made-to-measure’ implants produced through additive manufacturing [1]. However, a key limitation of PEEK is that it is bioinert and there is a requirement to functionalise its surface to make the material osteoconductive to ensure a more rapid, improved and stable fixation, in vivo. One approach to solving this issue is to modify PEEK with bioactive materials, such as hydroxyapatite (HA). OBJECTIVE. To 3D PEEK/HA composite materials using a Fused Filament Fabrication (FFF) approach to enhance the properties of the PEEK matrix. METHODS. PEEK/HA composites (0–30% w/w HA/PEEK) were 3D printed using a modified Ultimaker 2+ 3D printer. The mechanical, thermal, physical, chemical and in vitro properties of the 3D printed samples were all studied as part of this work. RESULTS. The CT images of both the filament and the 3D printed samples showed that the HA material was evenly dispersed throughout the bulk all the samples. SEM/EDX measurements highlighted that HA was homogenously distributed across the surface. As the HA content of the samples increases, so does the tensile modulus, ranging from 4.2 GPa (PEEK) to 6.1 GPa (30% HA/PEEK) and are significantly higher than datasheet information of injected molded PEEK samples. All materials supported the growth of osteoblast cells on their surface. CONCLUSIONS. The results clearly show that we can successfully and easily 3D print HA/PEEK composite materials up to 30% w/w HA/PEEK. The samples produced have a homogeneous distribution of HA in both the bulk and surface of all the samples, and their mechanical performance of the PEEK is enhanced by the addition of HA


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_11 | Pages 86 - 86
1 Dec 2020
Bal Z Kaito T Ishiguro H Okada R Kanayama S Kushioka J Kodama J Chijimatsu R Korkusuz P Dede EÇ Gizer M Yoshikawa H Korkusuz F
Full Access

To prevent the reported side effects of rhBMP-2, an important cytokine with bone forming capacity, the sustained release of rhBMP-2 is highly important. Synthetic copolymer polylactic acid-polyethylene glycol (PLA-PEG) is already shown to be a good carrier for rhBMP-2. The nano-sized hydroxyapatite (nHAp) is mentioned to be superior to conventional hydroxyapatite due to its decreased particle size which increases the surface area, so protein-cell adhesion and mechanical properties concomitantly. In the literature no study is reported with PLA-PEG / rhBMP-2/ nHAp for bone regeneration. In this study, we assessed the controlled release profile of rhBMP-2 from the novel biomaterial of PLA-PEG / rhBMP-2 / nHAp in vitro and evaluated the bone forming capacity of the composite in rat posterolateral spinal fusion (PSF) model in vivo. Composites were prepared via addition of rhBMP-2 (0µg, 3µg or 10µg) and nHAp (12.5mg) into PLA-PEG (5mg) + acetone solution and shaping. The release kinetics of the cytokine from the composites with 5µg BMP-2 was investigated by ELISA. The effect of nHAp and nHAp with rhBMP-2 on cell differentiation (rat BMSC cells, passage 3) was tested with ALP staining. In vivo bone formation was investigated by PSF on L4-L5 in a total of 36 male SD rats and weekly µCT results and histology at 8. th. weeks post operation were used for assessment of the bone formation. All animal experiments was approved by the institutional review board confirming to the laws and regulations of Japan. The composite showed an initial burst release in the first 24 hours (51.7% of the total released rhBMP-2), but the release was continued for the following 21 days. Thus, the sustained release of rhBMP-2 from the composite was verified. ALP staining results showed nHAp with rhBMP-2 contributed better on differentiation than nHAp itself. µCT and histology demonstrated that spinal fusion was achieved either one or both transverse processes in almost all BMP 3µg and BMP 10µg treated animals. On the contrary, only small or no bone formation was observed in the BMP0µg group (bilateral non-union / unilateral fusion/ bilateral fusion, BMP0µg group; 9/0/0, BMP3µg group; 1/0/11, BMP10µg group; 0/1/11). We developed a new technology for bone regeneration with BMP-2/PLA-PEG/nHAp composite. With this composite, the required dose of BMP-2 for spinal fusion in rats (10µg) was decreased to 1/3 (3µg) which can be explained by the superior properties of nano-sized hydroxyapatite and by the achievement of sustainable release of rhBMP-2 from the composite. This study is supported by Japanese Society of the Promotion of Science (JSPS) and Scientific and Technological Research Council of Turkey (TUBITAK). [Project No: 215S834]


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_1 | Pages 66 - 66
2 Jan 2024
Nikody M Li J Koper D Balmayor E Habibovic P Moroni L
Full Access

Critical-sized bone defects remain challenging in the clinical setting. Autologous bone grafting remains preferred by clinicians. However, the use of autologous tissue is associated with donor-site morbidity and limited accessibility to the graft tissue. Advances in the development of synthetic bone substitutes focus on improving their osteoinductive properties. Whereas osteoinductivity has been demonstrated with ceramics, it is still a challenge in case of polymeric composites. One of the approaches to improve the regenerative properties of biomaterials, without changing their synthetic character, is the addition of inorganic ions with known osteogenic and angiogenic properties. We have previously reported that the use of a bioactive composite with high ceramic content composed of poly(ethyleneoxide terephthalate)/poly(butylene terephthalate) (1000PEOT70PBT30, PolyActive, PA) and 50% beta-tricalcium phosphate (β-TCP) with the addition of zinc in a form of a coating of the TCP particles can enhance the osteogenic differentiation of human mesenchymal stromal cells (hMSCs) (3). To further support the regenerative properties of these scaffolds, inorganic ions with known angiogenic properties, copper or cobalt, were added to the coating solution. β-TCP particles were immersed in a zinc and copper or zinc and cobalt solution with a concentration of 15 or 45 mM. 3D porous scaffolds composed of 1000PEOT70PBT30 and pure or coated β-TCP were additively manufactured by 3D fibre deposition. The osteogenic and angiogenic properties of the fabricated scaffolds were tested in vitro through culture with hMSCs and human umbilical vein endothelial cells, respectively. The materials were further evaluated through ectopic implantation in an in vivo mini-pig model. The early expression of relevant osteogenic gene markers (collagen-1, osteocalcin) of hMSCs was upregulated in the presence of lower concentration of inorganic ions. Further analysis will focus on the evaluation of ectopic bone formation and vascularisation of these scaffolds after implantation in a mini-pig ectopic intramuscular model


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_2 | Pages 44 - 44
1 Jan 2017
Inyang A Vaughan C
Full Access

Medical grade polyurethanes have been widely promoted for biomedical applications. In particular, the use of polycarbonate-urethanes (PCU) has drawn considerable attention in the orthopaedic device industry as a result of their excellent mechanical properties, biostability and biocompatibility. PCUs have been extensively utilized in vascular grafts, stents and artificial heart valves. Specifically, bionate thermoplastic PCU, commercially produced by DSM PTG (Berkeley, California), has been of great interest in the field of orthopaedics because of its outstanding load-bearing properties and excellent wear resistance. Also, it is characterized by its long-term durability and resistance to hydrolytic degradation making it a good candidate for in-vivo orthopaedic applications. PCUs have been considered for meniscal replacement because of its unique weight-bearing capabilities, ability to withstand intense forces within the knee joint and ease of lubrication due to its hydrophilic nature. In addition, the low frictional properties essential for a meniscal replacement is obtainable with PCUs. Materials used for this study were a commercial polycarbonate-urethanes, Bionate PCU 80A (B8) and 90A (B9) pellets, and polyethylene continuous strands fibres (PE) obtained from DSM Polymer Technology Group, USA. Some quantity of the B8 and B9 pellets were dried separately in a vacuum oven at 100°C for 14 hours. A custom mould was designed for the production of the mechanical test samples. The quantity of the constituent materials was determined using composite theory known as the Rule of Mixtures. E. c. =. E. m. V. m. +. E. f. E. f. where V. m. and V. f. are the volume fraction of the matrix and fibre respectively. Three specimens each of the prepared composites were tested for tensile and compression strength and at a crosshead speed of 12 mm/min using a Zwick/Roell 1484 Material Testing Machine. The PCUs were not as stiff as their fibre-reinforced composites, which indicate that the stiffness of the PCU composite materials is a function of both the stiffness of the PCU matrix and the interspersed fibres. The tensile moduli of composites of B8 and B9 increased appreciably with PE. An increase of 227% was obtained for the B8 with the incorporation of PE fibres while percentage increase in stiffness for B9 was 148% for PE reinforcement fibres. The compressive modulus dropped with the inclusion of the PE fibres in the B9, a reduction of 55% was recorded while an increment of 4% was obtained with PE added to the B8. The results from this study demonstrate that the tensile and compressive properties of PCU can be custom-tailored to that of the meniscal tissue by systematically embedding reinforcement fibres into the PCU matrix such that a composite with desirable mechanical properties is obtained. The results of both tensile and compressive results visibly revealed the reinforcing effect of the fibres used in this study. However, additional studies are required to completely describe the PCU composite as a candidate meniscal substitute capable of gaining its full functionality


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_14 | Pages 130 - 130
1 Nov 2018
Leeuwenburgh S
Full Access

Calcium phosphate ceramics and bioactive glasses are frequently used in orthopedic surgery to stimulate the regeneration of bone tissue due to their superior compatibility to bone tissue. Nevertheless, the brittleness and lack of self-healing behavior of bioceramics are still considered as serious drawbacks. Therefore, these bioceramics have been combined with organic biomaterials for several decades. Since the 1990s, the emergence of nanotechnology has accelerated the progress with respect to the development of organic-inorganic nanocomposites of improved functionality compared to conventional composite biomaterials. This presentation focuses on the development of injectable (nano)composites with self-healing and/or load-bearing capacity. To this end, the affinity between polymeric and inorganic components was tuned by modifying non-covalent interactions between both composite components. Specifically, we exploited reversible interactions between hydrogel matrices and inorganic nanoparticles (traditional nanocomposites), hydrogel nanoparticles and inorganic nanoparticles (colloidal nanocomposites), as well as fibers and bioceramic matrices (fiber-reinforced cement composites). The resulting composite biomaterials were mechanically strong and self-healing, which may open up new avenues of research on the applicability of self-healing and load-bearing composite biomaterials for regenerative medicine


Orthopaedic Proceedings
Vol. 103-B, Issue SUPP_16 | Pages 74 - 74
1 Dec 2021
Chen H Khong J Huang J
Full Access

Abstract. Objectives. Direct ink writing (DIW) has gained considerable attention in production of personalized medical implants. Laponite nanoclay is added in polycaprolactone (PCL) to improve printability and bioactivity for bone implants. The 3D structure of DIW printed PCL/Laponite products was qualitatively evaluated using micro-CT. Methods. PCL/LP composite ink was formulated by dissolving 50% m/v PCL in dichloromethane with Laponite loading of up to 30%. The rheological properties of the inks were determined using Discovery HR-2 rheometer. A custom-made direct ink writer was used to fabricate both porous scaffold with 0°/90° lay-down pattern, and solid dumbbell-shaped specimens (ASTM D638 Type IV) with two printing orientations, 0° and 90° to the loading direction in tensile testing. The 3D structure of specimens was assessed using a micro-CT. Independent t-tests were performed with significance level at p<0.05. Results. The addition of Laponite in PCL ink has significantly enhanced viscosity for shape fidelity and shear-thinning property facilitating extrusion for DIW. Uniform distribution of Laponite was illustrated by micro-CT. For the 32-layer scaffold, interconnectivity of pores is observed at all 3 planes. The variation of height and width of layers is within 6% except the bottom 2 layers which are significantly lower and wider than other layers for mechanical support. For solid specimens, no ditches/interfaces between filaments are observed in 90° orientation while they are distinctive in 0° orientation because deposited filaments contact each other sooner in 90° orientation. 90° specimens also have lower air gap fraction (0.8 vs 5.4 %) and significantly higher Young's modulus (235 vs 195 MPa) and tensile strength (12.0 vs 9.5 MPa). Conclusions. The mechanical properties and printability of PCL/Laponite composites can be improved by controlling printing parameters; Micro-CT is an important tool to investigate the structure and properties of 3D printed products for bone tissue engineering


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_15 | Pages 28 - 28
1 Nov 2018
Bal Z Kaito T Ishiguro H Korkusuz P Dede E Korkusuz F
Full Access

Sustained release of BMP-2 is reported to be able to reduce the required dose of BMP-2 for bone induction. Nanohydroxyapatite (nHAp) has an osteoinduction capability which is lack in conventional hydroxyapatite. In this study, we combined PLA-PEG with nHAp and investigated the bone regenerative capacity of the newly established composite material of rhBMP-2/PLA-PEG/nHAp in a rat model of spinal fusion. The PLA-PEG was liquidized in acetone and mixed with nHAp and rhBMP-2. The sheet-shaped BMP-2/PLA-PEG (5mg)/nHAp (12.5mg) composites were prepared while evaporating the acetone. The release kinetics of rhBMP-2 from the composite was investigated by ELISA. In vivo bone formation was investigated by posterolateral spinal fusion in rats (the dosage of rhBMP-2; 0µg/ 0.5µg / 3µg). Bone formation was assessed by µCT and histology at post-op. 8 weeks. The composite showed the burst-release in the initial 24 hours (69% of total release) and the subsequent sustained-release for 25 days. According to µCT and histology of the spinal fusion experiment for all groups the bone formation was observed. While no bony bridging was observed in 0 µg and 0.5 µg BMP groups; in 3 µg group bony bridging and fusion were achieved. We developed a new technology for bone regeneration with rhBMP-2/PLA-PEG/nHAp composite. The reduction in the required dose of BMP-2 for bone induction was achieved. This result can be explained by the high bone induction ability of nHAp and sustainable release of BMP from PLA-PEG in the composite


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_14 | Pages 129 - 129
1 Nov 2018
Sá MJC Segundo FAS Freitas VML Azevedo AS Silva ACF de Lima GG Devine DM
Full Access

The aim of this study was to evaluate the trochlear bone and cartilaginous regeneration of rabbits using a composite based on platelet rich plasma (PRP), chitosan and hydroxyapatite. The study was approved by the ethics committee of the Federal University of Campina Grande under number 72/2017. Surgical holes measuring four millimetres in diameter were performed in rabbit trochleae, one surgical hole in each animal remained empty and another one was filled with the composite. Clinical-orthopaedic and radiographic evaluations were carried out for 60 days, after which the animals were euthanized for histomorphometric evaluations. Clinical-evaluations exhibited lameness of two members of the treatment (T) group and one member of control (C) group. The radiographic evaluation of T group exhibited absence of subchondral bone reaction (33%); nonetheless, presence of moderate subchondral bone reaction was more frequently reported in group C with 67%. Microscopic evaluation revealed the presence of tissue neoformation, composed of dense connective tissue. Microscopic findings were similar in both groups, with a difference in the amount of neoformed tissue, which was confirmed after the morphometric analysis, revealing a significant difference in the quantity of newly formed tissue at the bone / cartilage / implant interface in the T group. The results indicate that the composite based on chitosan, hydroxyapatite and PRP enhanced bone and cartilage healing


Bone & Joint Research
Vol. 4, Issue 4 | Pages 56 - 64
1 Apr 2015
Lv YM Yu QS

Objectives. The major problem with repair of an articular cartilage injury is the extensive difference in the structure and function of regenerated, compared with normal cartilage. Our work investigates the feasibility of repairing articular osteochondral defects in the canine knee joint using a composite lamellar scaffold of nano-ß-tricalcium phosphate (ß-TCP)/collagen (col) I and II with bone marrow stromal stem cells (BMSCs) and assesses its biological compatibility. Methods. The bone–cartilage scaffold was prepared as a laminated composite, using hydroxyapatite nanoparticles (nano-HAP)/collagen I/copolymer of polylactic acid–hydroxyacetic acid as the bony scaffold, and sodium hyaluronate/poly(lactic-co-glycolic acid) as the cartilaginous scaffold. Ten-to 12-month-old hybrid canines were randomly divided into an experimental group and a control group. BMSCs were obtained from the iliac crest of each animal, and only those of the third generation were used in experiments. An articular osteochondral defect was created in the right knee of dogs in both groups. Those in the experimental group were treated by implanting the composites consisting of the lamellar scaffold of ß-TCP/col I/col II/BMSCs. Those in the control group were left untreated. Results. After 12 weeks of implantation, defects in the experimental group were filled with white semi-translucent tissue, protruding slightly over the peripheral cartilage surface. After 24 weeks, the defect space in the experimental group was filled with new cartilage tissues, finely integrated into surrounding normal cartilage. The lamellar scaffold of ß-TCP/col I/col II was gradually degraded and absorbed, while new cartilage tissue formed. In the control group, the defects were not repaired. Conclusion. This method can be used as a suitable scaffold material for the tissue-engineered repair of articular cartilage defects. Cite this article: Bone Joint Res 2015;4:56–64


Orthopaedic Proceedings
Vol. 98-B, Issue SUPP_16 | Pages 35 - 35
1 Oct 2016
Asif I Williams S Fisher J Al-Hajjar M Anderson J Tipper J
Full Access

Wear particles produced by alumina ceramic-on-ceramic (CoC) bearings cause a minimal immunological response with low cytotoxicity and inflammatory potential. 1, 2. However, more comprehensive immunological studies are yet to be completed for the composite CoC (zirconia-toughened, platelet reinforced alumina) hip replacements due to difficulties in isolating the very low volume of clinically relevant wear debris generated by such materials in vitro. The aim of this study was to compare the cytotoxic effects of clinically relevant cobalt chromium (CoCr) nano-particles with commercial composite ceramic particles. Composite ceramic particles (commercial BIOLOX® delta powder) were obtained from CeramTec, Germany and clinically relevant CoCr wear particles were generated using a six station pin-on-plate wear simulator. L929 fibroblast cells were cultured with 50µm. 3. of CoCr wear debris or composite ceramic particles at low to high volumes ranging from 500µm. 3. –0.5µm. 3. per cell and the cyctotoxic effects of the particles were assessed over a period of 6 days using the ATP-Lite™ cell viability assay. The composite ceramic particles were bimodal in size (0.1–2µm & 30–100nm) and showed mild cytotoxic effects when compared with equivalent particle volumes (50µm. 3. ) of clinically relevant CoCr nano-particles (10–120nm). The CoCr nano-particles had significant cytotoxic effects from day 1, whereas the composite ceramic particles only showed cytotoxic effects at particle concentrations of 50 and 500µm. 3. after 6 days. The increased cytotoxicity of the clinically relevant CoCr nano-particles may have been attributed to the release of Co and Cr ions. This study demonstrated the potential cytotoxic effects of model ceramic particles at very high volume concentrations, but it is unlikely that such high particle volumes will be experienced routinely in vivo in such low wearing bearing materials. Future work will investigate the longer-term effects on genotoxicity and oxidative stress of low volumes of clinically-relevant generated BIOLOX® delta ceramic wear particles


Orthopaedic Proceedings
Vol. 103-B, Issue SUPP_13 | Pages 144 - 144
1 Nov 2021
García-Rey E Saldaña-Quero L Sedel L
Full Access

Introduction and Objective. Despite pure alumina have shown excellent long-term results in patients undergoing total hip arthroplasty (THA), alumina matrix composites (AMCs) composed of alumina and zirconium oxide are more commonly used. There are no comparative studies between these two different ceramics. We performed a retrospective case-control study to compare results and associated complications between AMC from two manufacturers and those with pure alumina from another manufacturer. Materials and Methods. 480 uncemented THAs with ceramic on ceramic (CoC) bearing surfaces (288 men and 192 women; mean age of 54.1 ± 12.4 years), were implanted from 2010 to 2015. Group 1: 281 THAs with pure alumina; Group 2A: 142 with AMC bearing in a trabecular titanium cup. Group 2B: 57 hips with AMC bearing with a porous-coated cup. Results. The mean follow-up was 7.3 years. There was one late infection in group 1, eight dislocations, three in group 1 (1.1%), three in group 2A (2.1%), all with a 36 mm femoral head, and two in group 2C (3.5%). Liner malseating was found in one hip in group 1, and in five hips in group 2C, of these, there were four liner fractures (7.0%). Four cups were revised for iliopsoas impingement (three in group 1 and one in group 2B). Two cups were revised for aseptic loosening, one in group 1 and one in group 2A, and four revised femoral stems in group 2A, three for subsidence and another for postoperative periprosthetic B. 2. fracture. The mean preoperative Harris Hip Score was 48.6 ± 3.3 in the whole series and 93.9 ± 7.2 at the end of follow-up. The survival rate of revision for any cause was 98.2% (95% Confidence Interval: 96.6–99.8) at ten years for group 1, 95.8% (95% CI: 92.1–99.5) for group 2A, and 91.1% (95% CI: 83.7–98.5) for group 2B (log-rank 0.030). Conclusions. Outcome of uncemented CoC THA in young patients was satisfactory at mid-term in all three groups. However, liner fractures were frequent in group 2B. All dislocated hips in group 2A had a 36 mm femoral head diameter, and revision due to any cause was less frequent in group 1. Pure alumina CoC THA can be used as a benchmark for comparison with newer CoC THAs


Abstract. Objective. To compare the periprosthetic fracture mechanics between a collared and collarless fully coated cementless femoral stem in a composite femur. Methods. Two groups of six composite femurs (‘Osteoporotic femur’, SawBones, WA USA) were implanted with either a collared (collared group) or collarless (collarless group) cementless femoral stem which was otherwise identical by a single experienced surgeon. Periprosthetic fractures of the femur were simulated using a previously published technique. High speed video recording was used to identify fracture mechanism. Fracture torque and angular displacement were measured and rotational work and system stiffness were estimated for each trial. Results were compared between collared and collarless group and the comparison was evaluated against previously published work using fresh frozen femurs and the same protocol. Results. In composite femur testing median fracture torque (IQR) was greater with a collared versus collarless implant (48.41 [42.60 to 50.27] Nm versus 45.12 [39.13 to 48.09] Nm, p= 0.4). Median rotational displacement (IQR) was less with a collared versus collarless implant (0.29 [0.27 to 0.31] radians versus 0.33 [0.32 to 0.34] radians, p= 0.07). Estimated rotary work was similar between groups (5.76 [4.92 to 6.64] J versus 5.21 [4.25 to 6.04] J, p= 0.4). Torsional stiffness was greater with a collared versus collarless implant (158.36 [152.61, 163.54] Nm per radian versus 138.79 [122.53, 140.59] Nm per radian, p= 0.5). Collarless stems were seen to move independently of the femur and fracture patterns originated at the calcar. Conclusions. Testing with composite femurs using an established protocol produced similar results to previously published studies using human femurs, but the difference between collared and collarless stems was smaller. The internal homogenous foam material in composite femurs does not accurately represent the heterogeneous cancellous bone which supports a femoral stem in vivo and may lead to overestimation of implant stability. Declaration of Interest. (a) fully declare any financial or other potential conflict of interest


Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_7 | Pages 138 - 138
4 Apr 2023
Markel D Dietz P Wu B Bou-Akl T Ren W
Full Access

The efficacy of saline irrigation for the treatment of periprosthetic infection (PJI) is limited in the presence of infected implants. This study evaluated the efficacy of vancomycin/tobramycin-doped polyvinyl alcohol (PVA)/ceramic composites (PVA-VAN/TOB-P) after saline irrigation in a mouse pouch infection model. 3D printed porous titanium (Ti) cylinders (400, 700 and 100 µm in pore size) were implanted into mice pouches, then inoculated with S. aureus at the amounts of 1X10. 3. CFU and 1X10. 6. CFU per pouch, respectively. Mice were randomized into 4 groups (n=6 for each group): (1) no bacteria; (2) bacteria without saline wash; 3) saline wash only, and (4) saline wash+PVA-VAN/TOB-P. After seven days, pouches were washed out alone or with additional injection of 0.2 ml of PVA-VAN/TOB-P. Mice were sacrificed 14 days after pouch wash. Bacteria cultures of collected Ti cylinders and washout fluid and histology of pouch tissues were performed. The low-grade infection (1X10. 3. CFU) was more significant in 400 µm Ti cylinders than that in Ti cylinders with larger pore sizes (700 and 1000 µm (p<0.05). A similar pattern of high-grade infection (1X10. 6. CFU) was observed (p<0.05). For the end wash, the bacteria burden (0.49±0.02) in saline wash group was completely eradicated by the addition of PVA-VAN/TOB-P (0.005±0.001, p<0.05). We noticed that 400 µm Ti cylinders have the highest risk of implant infection. Our data supported that the effect of saline irrigation was very limited in the presence of contaminated porous Ti cylinders. PVA-VAN/TOB-P was biodegradable, biocompatible, and was effective in eradicating bacteria retention after saline irrigation in a mouse model of low grade and high-grade infection. We believe that PVA-VAN/TOB-P represents an alternative to reduce the risk of PJI by providing a sustained local delivery of antibiotics


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_2 | Pages 45 - 45
1 Jan 2017
Manjubala I Ganesan P Narendrakumar U Madhan B
Full Access

Current strategy for orthopedic tissue engineering mainly focusses on the regeneration of the damaged tissue using cell-seeded three-dimensional scaffolds. Biocompatible scaffolds with controllable degradation and suitable mechanical property are required to support new tissue in-growth and regeneration . [1]. Porous composite scaffolds made from organic and inorganic materials are highly preferred, which can mimic the natural bone in their composition as well can enhance tissue repair . [2]. Scaffolds with optimum mechanical strength in both dry and wet state are more suitable for in vivo orthopedic application. Biphasic calcium phosphate (BCP), a biocompatible ceramic and carboxymethyl cellulose (CMC), a semi-natural polymer are used in the study to prepare composite scaffolds. Citric acid is used as a crosslinking agent for the polymer to improve its stability . [3]. Stability, mechanical property in dry and wet conditions and cytocompatibility of the scaffolds were investigated. Cellulose-BCP (BC25) and crosslinked cellulose-BCP (BC25CA) scaffolds are fabricated by freeze-drying method. The stability of the scaffolds was assessed in phosphate buffered saline (PBS) and compressive modulus was measured in dry and wet condition. Cytocompatibility was assessed by culturing pre-osteoblast cells at a density of 2.5×10. 4. on crosslinked scaffold and cell proliferation was measured by performing MTT assay on day 4 and 7. Crosslinked scaffold was more stable than non-crosslinked scaffold in aqueous environment as the latter disintegrated within few hours in the solution. Non-crosslinked scaffold showed higher compressive modulus of 116.3±14.8 kPa in dry condition but is reduced to 1.2±0.7 kPa in hydrated state. Though the crosslinked scaffold shows low compressive modulus of 37.67±6.7 kPa in dry state, it exhibited appreciable compressive moduli of 17.15±1.3 kPa in hydrated state. Thus, the crosslinking of the scaffolds improved the stability as well as the mechanical strength in wet condition. Cytocompatibility was assessed by culturing pre-osteoblast cells and from the MTT assay, it is shown that the cells are proliferating on the crosslinked scaffolds with time which indicates that the scaffolds are non-toxic and cytocompatible. Stability and optimum mechanical property for scaffold in aqueous environment are highly crucial for in vivo hard tissue regeneration. This study demonstrated the preparation of crosslinked scaffolds which exhibited good stability and mechanical strength in wet condition along with a porous architecture, controlled degradability and cytocompatibility, hence, crosslinked cellulose-BCP scaffold can be used for orthopedic application


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_2 | Pages 38 - 38
1 Jan 2017
Ehret C Sagardoy T Siadous R Bareille R De Mones E Amedee J Aid R Letourneur D Pechev S Etienne L
Full Access

Worldwide 500,000 cases of maxillofacial cancer are diagnosed each year. After surgery, the reconstruction of large bone defect is often required. The induced membrane approach (Masquelet, 2000) is one of the strategies, but exhibits limitations in an oncological context (use of autografts with or without autologous cells and Bone Morphogenetic Proteins). The objectives of this work are to develop an injectable osteoinductive and osteoconductive composite matrix composed of doped strontium (Sr) hydroxyapatite (HA) particles dispersed within a polysaccharide scaffold, to evaluate in vitro their ability to stimulate osteoblastic differentiation of human mesenchymal stem cells (hMSC) and to stimulate in vivo bone tissue regeneration. HA particles were synthesized with different ratios of Sr. X-ray diffraction (XRD), Inductively Coupled Plasma (ICP), and particle size analysis (Nanosizer™) were used to characterize these particles. HA and Sr-doped HA were dispersed at different ratios within a pullulan-dextran based matrices (Autissier, 2010), Electronic scanning microscopy Back Scattering Electron microscopy (ESEM-BSE) and ICP were used to characterize the composite scaffolds. In vitro assays were performed using hMSC (cell viability using Live/Dead assay, expression of osteoblastic markers by quantitative Polymerase Chain Reaction). Matrices containing these different particles were implanted subcutaneously in mice and analyzed by Micro-Computed Tomography (micro-CT) and histologically (Masson's trichrome staining) after 2 and 4 weeks of implantation. XRD analysis was compatible with a carbonated hydroxyapatite and patterns of Sr-doped HA are consistent of Sr substitution on HA particles. Morphological evaluation (TEM and Nanosizer™) showed that HA and Sr-doped HA particles form agglomerates (150 nm to 4 µm). Matrices composed with different ratios of HA or Sr-doped-HA, exhibit a homogenous distribution of the particles (ESEM-BSE), whatever the conditions of substitution. In vitro studies revealed that Sr-doped HA particles within the matrix stimulates the expression of osteoblastic markers, compared to non-doped HA matrices. Subcutaneous implantation of the matrices demonstrated the formation of a mineralized tissue. Quantitative analyses show that the mineralization of the implants is dependent of the amount of HA particles dispersed, with an optimal ratio of 5% of particles. Histological analysis revealed osteoid tissue in contact to the matrix. In conclusion, the ability of this injectable composite scaffold to promote ectopically tissue mineralization is promising for bone tissue engineering. Osseous implantation in a femoral bone defect in rats is now in progress. 5% of doped HA particles were implanted within the induced membranes in a context of radiotherapy procedure. Micro-CT analyses are ongoing. This new matrix could represent an alternative to the autografts for the regeneration of large bone defects in an oncological context


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_14 | Pages 96 - 96
1 Nov 2018
Frayssinet A Petta D Eglin D D'Este M Mosser G Helary C
Full Access

Collagen and hyaluronic acid are two major components of intervertebral disc (IVD). They give resistance and hydration to Nucleus Pulposus. In this study, we assessed the impact of Collagen (COLL) and Hyaluronic acid-Tyramine (THA) contents on the mechanical properties and the structure of composite hydrogels. For this purpose, a range of composites were obtained using a 4 mg/mL collagen concentration and different COLL/THA ratios from 8:1 to 1:5 (w/w). Composite gelling was performed by pH increase, triggering collagen fibrillogenesis and oxidative coupling of tyramine moieties in THA catalyzed by H. 2. O. 2. and horseradish peroxidase (HRP). To modulate the THA gelling kinetic, different HRP concentrations (0.05; 0.1 and 0.5 U/mL) were used. Composites with a low THA content exhibited a fibrillar structure and possessed mechanical properties close to those of pure collagen hydrogels (200 Pa). From the ratio 1:1, the storage modulus increased to reach c.a 1200 Pa for the ratio 1:5. From the ratio 1:2, the fibrillar structure disappeared and sheets, characteristic of THA hydrogels, were observed. The HRP activity dramatically impacted the physical properties. A rapid THA gelling associated with a high THA content tended to destabilize collagen fibrils and promoted the formation of covalent bond between collagen and THA. On the opposite a slow gelling kinetic favored collagen fibril formation up to the COLL/THA ratio 1:2. Taken together, these results show that a slow gelling and an 8 mg/mL THA concentration are the appropriate conditions to obtain biomimetic biomaterials for the treatment of Nucleus Pulposus


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_1 | Pages 20 - 20
1 Jan 2017
Mohammad H Pillai A
Full Access

We describe a case series using calcium sulphate bio composite with antibiotics (Cerament/Stimulan) in treating infected metalwork in the lower limb. Eight patients aged 22–74 (7 males, 1 female) presented with clinical evidence of infected limb metal work from previous orthopaedic surgery. Metal work removal with application of either cerement in 5 cases (10–20ml including 175mg–350mg gentamycin) or stimulan in 3 cases (10–20ml including either 1g vancomycin or clindamycin 1.2g or 100mg tigecycline) into the site was performed. Supplemental systemic antibiotic therapy (oral/intravenous) was instituted based on intraoperative tissue culture and sensitivity. Four patients had infected ankle metalwork, 2 patients infected distal tibial metalwork and 2 had infected external fixators. Metal work was removed in all cases. The mean pre operative CRP was 15.8mg/l (range 1–56mg/l). The mean postoperative CRP at 1 month was 20.5mg/l (range 2–98mg/l). The mean pre op WCC was 7.9×10. 9. (range 4.7–10.5 ×10. 9. ). Mean post op WCC at 1 month was 7.1×10. 9. (range 5.0–9.2×10. 9. ). The organisms cultured included enterobacter, staphylococcus aureus, staphylococcus epidermidis, staphylococcus cohnii, stenotrophomonas, acinetobacter, group B streptococcus, enterococcus and escherichia coli. No additional procedures were required in any case. All surgical wounds went on to heal uneventfully. Infection control and union was achieved both clinically and radiologically in all cases. Our results support the use of a calcium sulphate bio composite with antibiotic as an adjuvant for effective local infection control in cases with implant related bone sepsis. The technique is well tolerated with no systemic or local side effects. We believe that implant removal, debridement and local antibiotic delivery can minimise the need for prolonged systemic antibiotic therapy in such cases


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_1 | Pages 106 - 106
1 Jan 2017
Maisani M Bareille R Levesque L Amédée J Mantovani D Chassande O
Full Access

First works focuses on the characterization (physical and biological) of this biomaterial. Current work had studied osteoinductive and osteoconductive capacity of these hydrogels. In vivoresults highlight a significant bone reconstruction two months after implantations on bone lesions in mice. Bone is a dynamic and vascularized tissue that has the ability of naturally healing upon damage. Nevertheless, in the case of critical size defects this potential is impaired. Present approaches mainly consider autografts and allografts, which presents several limitations. Bone Tissue Engineering (BTE) is based on the use of 3D matrices to guide both cellular growth, differentiation to promote bone regeneration. Hence, matrices can contain biological materials such as cells and growth factors. Our project aims to design a hydrogel for BTE, particularly for bone lesion filling. We previously showed that a porous 3D hydrogel, Glycosyl-Nucleoside-Fluorinated (GNF) is: 1) non-cytotoxic to clustered human Adipose Mesenchymal Stem Cells (hASCs), 2) bioinjectable and 3) biodegradable. Therefore, this novel class of hydrogels show promise for the development of therapeutic solutions for BTE [1]. The hypothesis of this research was that improving the capacity to promote the adhesion of cells by adding collagen gel matrices and bone morphogenic protein 2 (BMP-2) to improve the bone regenerative potential of this gel. Collagen is a protein matrix well known for its cytocompatibility [2]. BMP-2, have been shown ability to induce bone formation in combination with an adequate matrix [3]. Thereby, the overall aim of this work was to design, develop and validate a new composite hydrogel for BTE. GNF was prepared as previously described in detail[1], at a concentration of 3% (w/v). Type I-collagen gel was prepared from rat-tail tendons at a concentration of 4 g/L [2]. hASCs were isolated from human adipose tissue in our laboratory. To establish a suitable microenvironment for cell proliferation and differentiation cells were seeded in collagen and then GNF gel was added and the resulting mixture was blended, BMP-2 (InductOs ® Kit) is added to this preparation (5µm BMP-2/ml). Fluorometry was used to follow BMP2 release in vitro andin vivo(NOG mices;n=6), orthotopic calvariumbone critical defect (3.3 mm) has been selected to challenge the bone repair. Adding collagen hydrogel improve cell adhesion, survivals and proliferation rather than simple GNF hydrogel. This novel gel composite has the ability to sustain hASCs adhesion and differentiation towards the osteoblastic lineage (positive ALP cells). Fluorometry showed the ability of our hydrogel to prolong the residence of BMP-2 (in vitro and in vivo) compared to collagen hydrogel sponges. Implantation of hydrogel containing hASC and BMP-2 has shown encouraging results in bone reconstruction: 2 months after implantation of biomaterials a significant bone reconstruction can be observed using X-Ray imaging. Adding collagen to GNF allowed to obtain gels showing satisfactory cell-behaviour. In parallel, the presence of GNF hydrogel helps to improve mechanical properties of the biomaterial (hydrogel stability and controlled release of BMP-2). The first in vivostudies have shown encouraging bone regeneration capacity of these hydrogels. The implantation performed on a larger number of animals and quantitative microCT analysis will enable us to judge the effectiveness of this hydrogel as a new injectable biomaterial for BTE. This work was partially supported by NSERC-Canada, FRQ-NT-Quebec, FRQ-S- Quebec, and CFI-Canada. Mathieu Maisani was awarded of a NSERC CREATE Program in Regenerative Medicine (www.ncprm.ulaval.ca)