Safety of autologous bone marrow aspiration concentrate transplantation: initial experiences in 101 patients

Christian Hendrich, Engelmaier Franz, Gerhart Waertel, Rolf Krebs, Marcus Jäger, Christian Hendrich, Engelmaier Franz, Gerhart Waertel, Rolf Krebs, Marcus Jäger

Abstract

The clinical application of cellular based therapies with ex vivo cultivation for the treatment of diseases of the musculoskeletal system has until now been limited. In particular, the advanced laboratory and technical effort necessary, regulatory issues as well as high costs are major obstacles. On the other hand, newly developed cell therapy systems permit intra-operative enrichment and application of mesenchymal and progenitor stem cells from bone marrow aspirate concentrate (BMAC) in one single operative session. The objective of the present clinical surveillance study was to evaluate new bone formation after the application of BMAC as well as to record any possible therapy-specific complicationsFor this purpose, the clinical-radiological progress of a total of 101 patients with various bone healing disturbances was documented (surveillance study). The study included 37 necrosis of the head of the femur, 32 avascular necroses/bone marrow edema of other localization, 12 non-unions, 20 other defects. The application of BMAC was performed in the presence of osteonecrosis via a local injection as part of a core decompression (n=72) or by the local adsorption of intra-operative cellular bone substitution material (scaffold) incubated with BMAC during osteosynthesis (n=17) or in further surgery (n=12).After an average of 14 months (2-24 months), the patients were re-examined clinically and radiologically and interviewed. Further surgery was necessary in 2 patients within the follow-up period. These were due to a progression of a collapsed head of the femur with initial necrosis in ARCO Stage III, as well as inadequate new bone formation with secondary loss of correction after periprosthetic femoral fracture. The latter healed after repeated osteosynthesis plus BMAC application without any consequences. Other than these 2 patients, no further complications were observed. In particular, no infections, no excessive new bone formation, no induction of tumor formation, as well as no morbidity due to the bone marrow aspiration from the iliac crest were seen.There were no specific complications within the short follow-up period and a simple intra-operative use of the system for different forms of bone loss could be demonstrated. In the authors' opinion, the on-site preparation of the bone marrow cells within the operating theater eliminates the specific risk of ex vivo cell proliferation and has a safety advantage in the use of autologous cell therapy for bone regeneration. Additional studies should be completed to determine efficacy.

Keywords: bone marrow aspirate concentrate application..

Figures

Figure 1
Figure 1
Indications in 101 autologous mesenchymal stem cell transplantations. AVN: avascular necrosis.
Figure 2
Figure 2
A 53-year old patient with necrosis of the head of the femur with accompanying large bone marrow edema on the right side. Transcutaneous core decompression and BMAC transplantation. MRI controls after four and eight weeks. In addition to the clearly distinguishable drill channels, an almost complete normalization of the bone marrow signal can be seen.
Figure 3
Figure 3
A 36-year old patient with necrosis of the head of the femur after DHS. BMAC transplantation with incipient subchondral fracture. During the further course of treatment, there was collapse of the femoral head followed by a total joint replacement.
Figure 4
Figure 4
An 86-year old female patient with periprosthetic fracture after total hip revision surgery showed a failure of LISS osteosynthesis. Re-osteosynthesis was combined with application of bone substitute material (CopiOs®) augmented by autologous BMAC. Despite good new bone formation, increasing axial deviation was noted after two months. Additional internal fixation by plate osteosynthesis from the anterior combined with a second CopiOs/BMAC transplantation was performed. Here, some tissue from the initial transplantation site was taken for histology. The patient showed a solid fusion of the fracture after a further three months post-operatively. The histological analysis of the transplantation site showed a significant new formation of woven bone (polarization optics, magnification × 200).

References

    1. Rueger JM. Bone substitution materials. Current status and prospects. Orthopade. 1998;27:72–9.
    1. Carstens MH, Chin M, Li XJ. In situ osteogenesis: regeneration of 10-cm mandibular defect in porcine model using recombinant human bone morphogenetic protein-2 (rhBMP-2) and Helistat absorbable collagen sponge. J Craniofac Surg. 2005;16:1033–42.
    1. Kraus KH, Kirker-Head C. Mesenchymal stem cells and bone regeneration. Vet Surg. 2006;35:232–42.
    1. Hernigou P, Beaujean F. Treatment of osteonecrosis with autologous bone marrow grafting. Clin Orthop Relat Res. 2002:14–23.
    1. Hernigou P, Beaujean F, Lambotte JC. Decrease in the mesenchymal stem-cell pool in the proximal femur in corticosteroid-induced osteonecrosis. J Bone Joint Surg Br. 1999;81:349–55.
    1. Hernigou P, Bernaudin F, Reinert P, et al. Bone-marrow transplantation in sickle-cell disease. Effect on osteonecrosis: a case report with a four-year follow-up. J Bone Joint Surg Am. 1997;79:1726–30.
    1. Hernigou P, Daltro G, Filippini P, et al. Percutaneous implantation of autologous bone marrow osteoprogenitor cells as treatment of bone avascular necrosis related to sickle cell disease. Open Orthop J. 2008;2:62–5.
    1. Hernigou P, Lambotte JC. Volumetric analysis of osteonecrosis of the femur. Anatomical correlation using MRI. J Bone Joint Surg Br. 2001;83:672–5.
    1. Hernigou P, Mathieu G, Poignard A, et al. Percutaneous autologous bone-marrow grafting for nonunions. Surgical technique. J Bone Joint Surg Am. 2006;88(Suppl 1 Pt 2):322–7.
    1. Hernigou P, Poignard A, Beaujean F, Rouard H. Percutaneous autologous bone-marrow grafting for nonunions. Influence of the number and concentration of progenitor cells. J Bone Joint Surg Am. 2005;87:1430–7.
    1. Hernigou P, Poignard A, Manicom O, et al. The use of percutaneous autologous bone marrow transplantation in nonunion and avascular necrosis of bone. J Bone Joint Surg Br. 2005;87:896–902.
    1. Jä ger M, Jelinek EM, Wess KM, et al. Bone marrow concentrate: a novel strategy for bone defect treatment. Curr Stem Cell Res Ther. 2009;4:34–43.
    1. Hermann PC, Huber SL, Herrler T, et al. Concentration of bone marrow total nucleated cells by a point-of-care device provides a high yield and preserves their functional activity. Cell Transplant. 2008;16:1059–69.
    1. Epstein NE. Bone void filler in posterior iliac crest reconstruction and to supplement intertransverse process fusion. Spine J. 2006;6:600–2.
    1. Gisep A, Wieling R, Bohner M, et al. Resorption patterns of calcium-phosphate cements in bone. J Biomed Mater Res A. 2003;66:532–40.
    1. Bhattacharyya T, Chang D, Meigs JB, et al. Mortality after periprosthetic fracture of the femur. J Bone Joint Surg Am. 2007;89:2658–62.
    1. Gangji V, Hauzeur JP. Treatment of osteonecrosis of the femoral head with implantation of autologous bone-marrow cells. Surgical technique. J Bone Joint Surg Am. 2005;87(Suppl 1Pt 1):106–12.
    1. Gangji V, Hauzeur JP. Cellular-based therapy for osteonecrosis. Orthop Clin North Am. 2009;40:213–21.
    1. Gangji V, Toungouz M, Hauzeur JP. Stem cell therapy for osteonecrosis of the femoral head. Expert Opin Biol Ther. 2005;5:437–42.
    1. Mertsching H, Walles T. Europe's advanced therapy medicinal products: chances and challenges. Expert Rev Med Devices. 2009;6:109–10.
    1. Bernstein P, et al. Regenerative therapy of large periprosthetic bone defects - Problems in submitting applications. Expert workshop of the DGOU: stem cell-based therapy. Munich. 2009
    1. Caplan AI. The mesengenic process. Clin Plast Surg. 1994;21:429–35.
    1. Kawate K, Yajima H, Ohgushi H, et al. Tissue-engineered approach for the treatment of steroid-induced osteonecrosis of the femoral head: transplantation of autologous mesenchymal stem cells cultured with beta-tricalcium phosphate ceramics and free vascularized fibula. Artif Organs. 2006;30:960–2.
    1. Nöth U, Reichert J, Reppenhagen S, et al. Cell based therapy for the treatment of femoral head necrosis. Orthopade. 2007;36:466–71.
    1. Quarto R, Mastrogiacomo M, Cancedda R, et al. Repair of large bone defects with the use of autologous bone marrow stromal cells. N Engl J Med. 2001;344:385–6.
    1. Walldorf J, Aurich H, Cai H, et al. Expanding hepatocytes in vitro before cell transplantation: donor age-dependent proliferative capacity of cultured human hepatocytes. Scand J Gastroenterol. 2004;39:584–93.
    1. Kasten P, Beyen I, Egermann M, et al. Instant stem cell therapy: characterization and concentration of human mesenchymal stem cells in vitro. Eur Cell Mater. 2008;16:47–55.
    1. Wongchuensoontorn C, Liebehenschel N, Schwarz U, et al. Application of a new chair-side method for the harvest of mesenchymal stem cells in a patient with nonunion of a fracture of the atrophic mandible-a case report. J Cranioma xillofac Surg. 2009;37:155–61.

Source: PubMed

3
Suscribir