The Masquelet Technique: Can Disposable Polypropylene Syringes be an Alternative to Standard PMMA Spacers? A Rat Bone Defect Model

Abstract Background Usually, the two-stage Masquelet induced-membrane technique for extremity reconstruction begins with a polymethylmethacrylate (PMMA) cement spacer–driven membrane, followed by an autologous cancellous bone graft implanted into the membrane cavity to promote healing of large bone defects. In exceptional cases, spacers made of polypropylene disposable syringes were successfully used instead of the usual PMMA spacers because of a PMMA cement shortage caused by a lack of resources. However, this approach lacks clinical evidence and requires experimental validation before being recommended as an alternative to the conventional technique. Questions/purposes To (1) develop and (2) validate a critical-sized femoral defect model in rats for two stages of the Masquelet technique and to (3) compare the biological and bone healing properties of polypropylene-induced membranes and PMMA-induced membranes in this model. Methods Fifty male Sprague Dawley rats aged 8 weeks old received a 6-mm femur defect, which was stabilized with an external fixator that was converted into an internal device. In the development phase, the defect was filled with PMMA in 16 rats to determine the most favorable timing for bone grafting. Two rats were excluded since they died of anesthetic complications. The other 14 were successively euthanized after 2 weeks (n = 3), 4 weeks (n = 4), 6 weeks (n = 4), and 8 weeks (n = 3) for induced membrane analyses. In the validation phase, 12 rats underwent both stages of the procedure using a PMMA spacer and were randomly assigned to two groups, whether the induced membrane was preserved or removed before grafting. To address our final objective, we implanted either polypropylene or PMMA spacers into the defect (Masquelet technique Stage 1; n = 11 rats per group) for the period established by the development phase. In each group, 6 of 11 rats were euthanized to compare the biological properties of polypropylene-induced membranes and PMMA-induced membranes using histological qualitative analysis, semiquantitative assessment of the bone morphogenic protein-2 content by immunostaining, and qualitative assessment of the mesenchymal stromal cell (MSC; CD31-, CD45-, CD90+, and CD73+ phenotypes) content by flow cytometry. Quantitative measurements from serum bone turnover markers were also performed. The five remaining rats of each group were used for Masquelet technique Stage 2, in which rat bone allografts were implanted in the induced membrane cavity after the polypropylene or PMMA spacers were removed. These rats recovered for 10 weeks before being euthanized for microCT quantitative measurements and bone histology qualitative assessment to evaluate and compare the extent of bone regeneration between groups. Results Induced membrane analyses together with serum bone turnover measurements indicated that a 4-week interval time between stages was the most favorable. Removal of the induced membrane before grafting led to almost constant early implant failures with poor bone formation. Four-week-old rats with polypropylene-triggered induced membranes displayed similar histologic organization as rats with PMMA-driven induced membranes, without any difference in the cell density of the extracellular matrix (4933 ± 916 cells per mm2 for polypropylene versus 4923 ± 1284 cells per mm2 for PMMA; p = 0.98). Induced membrane-derived MSCs were found in both groups with no difference (4 of 5 with polypropylene versus 3 of 3 with PMMA; p > 0.99). Induced membrane bone morphogenic protein-2 immunolabeling and serum bone turnover marker levels were comparable between the polypropylene and PMMA groups. MicroCT analysis found that bone regeneration in the polypropylene group seemed comparable with that in the PMMA group (29 ± 26 mm3 for polypropylene versus 24 ± 18 mm3 for PMMA; p > 0.99). Finally, qualitative histological assessment revealed a satisfactory endochondral ossification maturation in both groups. Conclusion Using a critical-sized femoral defect model in rats, we demonstrated that polypropylene spacers could induce membrane encapsulation with histologic characteristics and bone regenerative capacities that seem like those of PMMA spacers. Clinical Relevance In a same bone site, polymers with close physical properties seem to lead to similar foreign body reactions and induce encapsulating membranes with comparable bone healing properties. Polypropylene spacers made from disposable syringes could be a valuable alternative to PMMA. These results support the possibility of a cementless Masquelet technique in cases of PMMA shortage caused by a lack of resources.

[1]  A. Masquelet,et al.  Induced membrane technique with sequential internal fixation: use of a reinforced spacer for reconstruction of infected bone defects , 2020, International Orthopaedics.

[2]  A. Masquelet,et al.  The Masquelet technique: Current concepts, animal models, and perspectives , 2020, Journal of tissue engineering and regenerative medicine.

[3]  A. Masquelet,et al.  [The wrapping induced membrane technique for treating recalcitrant non unions]. , 2020, Annales de chirurgie plastique et esthetique.

[4]  Jens Pietzsch,et al.  Adjuvant Drug-Assisted Bone Healing: Advances and Challenges in Drug Delivery Approaches , 2020, Pharmaceutics.

[5]  P. Giannoudis,et al.  Mixed results with the Masquelet technique: A fact or a myth? , 2019, Injury.

[6]  J. Scimeca,et al.  Fibrin as a Multipurpose Physiological Platform for Bone Tissue Engineering and Targeted Delivery of Bioactive Compounds , 2019, Pharmaceutics.

[7]  S. Amar,et al.  Metacarpal bone reconstruction by a cementless induced membrane technique. , 2019, Hand surgery & rehabilitation.

[8]  A. Masquelet,et al.  Use of the induced membrane technique for long bone reconstruction in low-resource settings. , 2019 .

[9]  J. Watson,et al.  Masquelet Technique: Effects of Spacer Material and Micro-topography on Factor Expression and Bone Regeneration , 2018, Annals of Biomedical Engineering.

[10]  L. De Lucca,et al.  Guided bone regeneration with polypropylene barrier in rabbit's calvaria: A preliminary experimental study , 2018, Heliyon.

[11]  J. Watson,et al.  Masquelet technique: The effect of altering implant material and topography on membrane matrix composition, mechanical and barrier properties in a rat defect model. , 2018, Journal of biomechanics.

[12]  J. Watson,et al.  Altering spacer material affects bone regeneration in the Masquelet technique in a rat femoral defect , 2018, Journal of orthopaedic research : official publication of the Orthopaedic Research Society.

[13]  R. Bareille,et al.  Influence of External Beam Radiotherapy on the Properties of Polymethyl Methacrylate-Versus Silicone-Induced Membranes in a Bilateral Segmental Bone Defect in Rats. , 2017, Tissue engineering. Part A.

[14]  Gang Zheng,et al.  Masquelet's induced membrane promotes the osteogenic differentiation of bone marrow mesenchymal stem cells by activating the Smad and MAPK pathways. , 2018, American journal of translational research.

[15]  Bin Yu,et al.  Calcium sulfate induced versus PMMA-induced membrane in a critical-sized femoral defect in a rat model , 2018, Scientific Reports.

[16]  A. Masquelet Induced Membrane Technique: Pearls and Pitfalls. , 2017, Journal of orthopaedic trauma.

[17]  Yin Xiao,et al.  Structural properties of fracture haematoma: current status and future clinical implications , 2017, Journal of tissue engineering and regenerative medicine.

[18]  G. Muschler,et al.  The Effect of Surgical Technique and Spacer Texture on Bone Regeneration: A Caprine Study Using the Masquelet Technique , 2017, Clinical orthopaedics and related research.

[19]  C. Boudot,et al.  Osteoclasts and their precursors are present in the induced‐membrane during bone reconstruction using the Masquelet technique , 2017, Journal of tissue engineering and regenerative medicine.

[20]  L. Drago,et al.  Masquelet technique: myth or reality? A systematic review and meta-analysis. , 2016, Injury.

[21]  I. Marzi,et al.  Establishment and characterization of the Masquelet induced membrane technique in a rat femur critical‐sized defect model , 2016, Journal of tissue engineering and regenerative medicine.

[22]  Michelle Kelly,et al.  In vivo response to polypropylene following implantation in animal models: a review of biocompatibility , 2016, International Urogynecology Journal.

[23]  A. Lädermann,et al.  The Nice knot as an improvement on current knot options: A mechanical analysis. , 2016, Orthopaedics & traumatology, surgery & research : OTSR.

[24]  I. Marzi,et al.  Alteration of Masquelet's induced membrane characteristics by different kinds of antibiotic enriched bone cement in a critical size defect model in the rat's femur. , 2016, Injury.

[25]  F. Wei,et al.  Induction of granulation tissue for the secretion of growth factors and the promotion of bone defect repair , 2015, Journal of Orthopaedic Surgery and Research.

[26]  R. Bareille,et al.  Comparative study of membranes induced by PMMA or silicone in rats, and influence of external radiotherapy. , 2015, Acta biomaterialia.

[27]  H. Isaksson,et al.  The masquelet induced membrane technique with BMP and a synthetic scaffold can heal a rat femoral critical size defect , 2015, Journal of orthopaedic research : official publication of the Orthopaedic Research Society.

[28]  Y. Shen,et al.  Histological characteristics of induced membranes in subcutaneous, intramuscular sites and bone defect. , 2013, Orthopaedics & traumatology, surgery & research : OTSR.

[29]  Rozalia Dimitriou,et al.  Masquelet technique for the treatment of bone defects: tips-tricks and future directions. , 2011, Injury.

[30]  G. Duda,et al.  Influence of Gender and Fixation Stability on Bone Defect Healing in Middle-aged Rats: A Pilot Study , 2011, Clinical orthopaedics and related research.

[31]  Zhenxing Si,et al.  Successful repair of a critical-sized bone defect in the rat femur with a newly developed external fixator. , 2009, The Tohoku journal of experimental medicine.

[32]  J. van den Dolder,et al.  Bone regenerative properties of rat, goat and human platelet-rich plasma. , 2009, International journal of oral and maxillofacial surgery.

[33]  R. Civitelli,et al.  Bone turnover markers: understanding their value in clinical trials and clinical practice , 2009, Osteoporosis International.

[34]  Hermann Seitz,et al.  Validation of a femoral critical size defect model for orthotopic evaluation of bone healing: a biomechanical, veterinary and trauma surgical perspective. , 2008, Tissue engineering. Part C, Methods.

[35]  V. Bousson,et al.  Long‐bone critical‐size defects treated with tissue‐engineered grafts: A study on sheep , 2007, Journal of orthopaedic research : official publication of the Orthopaedic Research Society.

[36]  V. Bousson,et al.  Induction of a barrier membrane to facilitate reconstruction of massive segmental diaphyseal bone defects: an ovine model. , 2006, Veterinary surgery : VS.

[37]  A C Masquelet,et al.  Induced membranes secrete growth factors including vascular and osteoinductive factors and could stimulate bone regeneration , 2004, Journal of orthopaedic research : official publication of the Orthopaedic Research Society.

[38]  F. Fitoussi,et al.  [Reconstruction of the long bones by the induced membrane and spongy autograft]. , 2000, Annales de chirurgie plastique et esthetique.