Percutaneous Autologous Bone Marrow-Derived Mesenchymal Stromal Cell Implantation Is Safe for Reconstruction of Human Lower Limb Long Bone Atrophic Nonunion

Mohsen Emadedin, Narges Labibzadeh, Roghayeh Fazeli, Fatemeh Mohseni, Seyedeh Esmat Hosseini, Reza Moghadasali, Soura Mardpour, Vajiheh Azimian, Alireza Goodarzi, Maede Ghorbani Liastani, Ali Mirazimi Bafghi, Mohamadreza Baghaban Eslaminejad, Nasser Aghdami, Mohsen Emadedin, Narges Labibzadeh, Roghayeh Fazeli, Fatemeh Mohseni, Seyedeh Esmat Hosseini, Reza Moghadasali, Soura Mardpour, Vajiheh Azimian, Alireza Goodarzi, Maede Ghorbani Liastani, Ali Mirazimi Bafghi, Mohamadreza Baghaban Eslaminejad, Nasser Aghdami

Abstract

Objective: Nonunion is defined as a minimum of a 9-month period of time since an injury with no visibly progressive signs of healing for 3 months. Recent studies show that application of mesenchymal stromal cells (MSCs) in the laboratory setting is effective for bone regeneration. Animal studies have shown that MSCs can be used to treat nonunions. For the first time in an Iranian population, the present study investigated the safety of MSC implantation to treat human lower limb long bone nonunion.

Materials and methods: It is a prospective clinical trial for evaluating the safety of using autologus bone marrow derived mesenchymal stromal cells for treating nonunion. Orthopedic surgeons evaluated 12 patients with lower limb long bone nonunion for participation in this study. From these, 5 complied with the eligibility criteria and received MSCs. Under fluoroscopic guidance, patients received a one-time implantation of 20-50×106 MSCs into the nonunion site. All patients were followed by anterior-posterior and lateral X-rays from the affected limb, in addition to hematological, biochemical, and serological laboratory tests obtained before and 1, 3, 6, and 12 months after the implantation. Possible adverse effects that included local or systemic, serious or non-serious, and related or unrelated effects were recorded during this time period.

Results: From a safety perspective, all patients tolerated the MSCs implantation during the 12 months of the trial. Three patients had evidence of bony union based on the after implantation Xrays.

Conclusion: The results have suggested that implantation of bone marrow-derived MSCs is a safe treatment for nonunion. A double-blind, controlled clinical trial is required to assess the efficacy of this treatment (Registration Number: NCT01206179).

Keywords: Autologous; Bone Marrow; Mesenchymal Stromal Cells; Nonunion.

Figures

Fig.1
Fig.1
Flowchart of patients. HDL; High density lipoprotein, LDL; Low density lipoprotein, ALT; Alanin aminotransferase, AST; Aspartate aminoternsferase, BUN; Blood urea nitrogen, TSH; Thyroid stimulating hormone, T4; Thyroxine, PT; Prothrombine time, PTT; Partial thromboplastine time, INR; International ration, HBS Ag; Hepatitis B surface Ag, HIV; Human immunodeficiency virus, HTLV; Human T lymphtropic virus, BM; Bone marrow, and MSCs; Mesenchymal stromal cells.
Fig.2
Fig.2
Characterization of passage-1 human bone marrow mesenchymal stromal cells (MSCs). A. Phenotypic appearance of fibroblast-like MSCs after one passage in MSCs injected in patients with nonunion and B. Representative flow cytometric analysis using WinMDI software confirmed the expressions of CD90, CD105, CD73, CD44, and CD45/CD34 surface markers (red lines) on MSCs compared to isotype controls (black lines).
Fig.3
Fig.3
Patients radiographies. A. Anteroposterior and lateral radiographs of a non-united open tibial and fibular fracture of a 32-year-old patient, B. 2 months after using mesenchymal stromal cells (MSCs) with demonstrated radiological signs of healing, C. Lateral radiograph of a non-united femural fracture of a 23-year-old patient before implantation of mesenchymal stromal cells, and D. 6 months after transcutaneous implantation of MSCs with radiological signs of healing.

References

    1. Einhorn TA. Enhancement of fracture-healing. J Bone Joint Surg Am. 1995;77(6):940–956.
    1. Giannoudis PV, Einhorn TA, Marsh D. Fracture healing: the diamond concept. Injury. 2007;38(Suppl 4):S3–6.
    1. Assiotis A, Sachinis NP, Chalidis BE. Pulsed electromagnetic fields for the treatment of tibial delayed unions and nonunions.A prospective clinical study and review of the literature. J Orthop Surg Res. 2012;7:24–24.
    1. Tressler MA, Richards JE, Sofianos D, Comrie FK, Kregor PJ, Obremskey WT. Bone morphogenetic protein-2 compared to autologous iliac crest bone graft in the treatment of long bone nonunion. Orthopedics. 2011;34(12):e877–884.
    1. Khan Y, Laurencin CT. Fracture repair with ultrasound: clinical and cell-based evaluation. J Bone Joint Surg Am. 2008;90(Suppl 1):138–144.
    1. Sugaya H, Mishima H, Aoto K, Li M, Shimizu Y, Yoshioka T, et al. Percutaneous autologous concentrated bone marrow grafting in the treatment for nonunion. Eur J Orthop Surg Traumatol. 2014;24(5):671–678.
    1. Pountos I, Giannoudis PV. Biology of mesenchymal stem cells. Injury. 2005;36(Suppl 3):S8–S12.
    1. Dreger T, Watson JT, Akers W, Molligan J, Achilefu S, Schon LC, et al. Intravenous application of CD271-selected mesenchymal stem cells during fracture healing. J Orthop Trauma. 2014;28(Suppl 1):S15–19.
    1. Qi Y, Zhao T, Yan W, Xu K, Shi Z, Wang J. Mesenchymal stem cell sheet transplantation combined with locally released simvastatin enhances bone formation in a rat tibia osteotomy model. Cytotherapy. 2013;15(1):44–56.
    1. Cheung WH, Chin WC, Wei FY, Li G, Leung KS. Applications of exogenous mesenchymal stem cells and low intensity pulsed ultrasound enhance fracture healing in rat model. Ultrasound Med Biol. 2013;39(1):117–125.
    1. Obermeyer TS, Yonick D, Lauing K, Stock SR, Nauer R, Strotman P, et al. Mesenchymal stem cells facilitate fracture repair in an alcohol-induced impaired healing model. J Orthop Trauma. 2012;26(12):712–718.
    1. Xiang L, Liang C, Zhen-Yong K, Liang-Jun Y, Zhong-Liang D. BMP9-induced osteogenetic differentiation and bone formation of muscle-derived stem cells. J Biomed Biotechnol. 2012;2012:610952–610952.
    1. Zhu L, Liu W, Cui L, Cao Y. Tissue-engineered bone repair of goat-femur defects with osteogenically induced bone marrow stromal cells. Tissue Eng. 2006;12(3):423–433.
    1. Xue G, He M, Zhao J, Chen Y, Tian Y, Zhao B, et al. Intravenous umbilical cord mesenchymal stem cell infusion for the treatment of combined malnutrition nonunion of the humerus and radial nerve injury. Regen Med. 2011;6(6):733–741.
    1. Fayaz HC, Giannoudis PV, Vrahas MS, Smith RM, Moran C, Pape HC, et al. The role of stem cells in fracture healing and nonunion. Int Orthop. 2011;35(11):1587–1597.
    1. Horwitz EM, Prockop DJ, Fitzpatrick LA, Koo WW, Gordon PL, Neel M, et al. Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat Med. 1999;5(3):309–313.
    1. Bone RC, Balk RA, Cerra FB, Dellinger RP, Fein AM, Knaus WA, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis.The ACCP/SCCM Consensus Conference Committee.American College of Chest Physicians/Society of Critical Care Medicine. Chest. 1992;101(6):1644–1655.
    1. Younger EM, Chapman MW. Morbidity at bone graft donor sites. J Orthop Trauma. 1989;3(3):192–195.
    1. Marsell R, Einhorn TA. Emerging bone healing therapies. J Orthop Trauma. 2010;24(Suppl 1):S4–8.
    1. Lynch JR, Taitsman LA, Barei DP, Nork SE. Femoral nonunion: risk factors and treatment options. J Am Acad Orthop Surg. 2008;16(2):88–97.
    1. Kwong FN, Harris MB. Recent developments in the biology of fracture repair. J Am Acad Orthop Surg. 2008;16(11):619–625.
    1. Mahendra A, Maclean AD. Available biological treatments for complex non-unions. Injury. 2007;38(Suppl 4):S7–12.
    1. Wang X, Qiu Y, Triffitt J, Carr A, Xia Z, Sabokbar A. Proliferation and differentiation of human tenocytes in response to platelet rich plasma: an in vitro and in vivo study. J Orthop Res. 2012;30(6):982–990.
    1. Starr AJ. Fracture repair: successful advances, persistent problems, and the psychological burden of trauma. J Bone Joint Surg Am. 2008;90(Suppl 1):132–137.
    1. Müller ME. Treatment of nonunions by compression. Clin Orthop Relat Res. 1965;43:83–92.
    1. Pape HC, Evans A, Kobbe P. Autologous bone graft: properties and techniques. J Orthop Trauma. 2010;24(Suppl 1):S36–40.
    1. De Long WG Jr, Einhorn TA, Koval K, McKee M, Smith W, Sanders R, et al. Bone grafts and bone graft substitutes in orthopaedic trauma surgery: a critical analysis. J Bone Joint Surg Am. 2007;89(3):649–658.
    1. Giannoudis PV, Dinopoulos H, Tsiridis E. Bone substitutes: an update. Injury. 2005;36(Suppl 3):S20–27.
    1. Gazdag AR, Lane JM, Glaser D, Forster RA. Alternatives to autogenous bone graft: efficacy and indications. J Am Acad Orthop Surg. 1995;3(1):1–8.
    1. Sen MK, Miclau T. Autologous iliac crest bone graft: should it still be the gold standard for treating nonunions? Injury. 2007;38(Suppl 1):S75–80.
    1. Chan BP, Hui TY, Wong MY, Yip KH, Chan GC. Mesenchymal stem cell-encapsulated collagen microspheres for bone tissue engineering. Tissue Eng Part C Methods. 2010;16(2):225–235.

Source: PubMed

3
購読する