Efficacy of autologous stem cell-based therapy for osteonecrosis of the femoral head in sickle cell disease: a five-year follow-up study

Gildasio Cerqueira Daltro, Vitor Fortuna, Eliane Silva de Souza, Marcela Miranda Salles, Ana Claudia Carreira, Roberto Meyer, Songeli Menezes Freire, Radovan Borojevic, Gildasio Cerqueira Daltro, Vitor Fortuna, Eliane Silva de Souza, Marcela Miranda Salles, Ana Claudia Carreira, Roberto Meyer, Songeli Menezes Freire, Radovan Borojevic

Abstract

Introduction: Stem cell therapy with bone marrow-derived mononuclear cells (BMMCs) is an option for improving joint function in osteonecrosis of the femoral head (ONFH). Bone marrow-derived mesenchymal stromal cell (MSC) numbers and their osteogenic differentiation are decreased in patients with ONFH. However, whether this decrease also extends to the early stages of ONFH in sickle cell disease (SCD) is still unclear.

Methods: We conducted a phase I/II, non-controlled study to determine efficacy and safety of BMMC implantation using a minimally invasive technique in SCD patients with ONFH. Eighty-nine patients were recruited and followed up for 60 months after surgery. Clinical and radiographic findings were assessed, and data were completed by in vitro analysis.

Results: At the final follow-up (60 months) there was a significant improvement in clinical joint symptoms and pain relief as measured by the Harris Hip Score (P = 0.0005). In addition, after the BMMC implantation procedure, radiographic assessment showed disease stabilization and only 3.7 % of the treated patients did not achieve a satisfactory clinical result. The amount of fibroblast colony-forming units was 28.2 ± 13.9 per 1 million BMMCs after concentration. Flow cytometry analysis showed a significantly higher number of hematopoietic stem/endothelial progenitor cell markers in concentrated BMMCs when compared with bone marrow aspirate, indicating an enrichment of these cell types. Isolated MSCs from SCD patients with pre-collapse ONFH maintained the replicative capacity without significant loss of their specific biomolecular characteristics, multi-differentiation potential, and osteogenic differentiation activities. Cytokines and growth factors (interleukin-8, transforming growth factor-beta, stromal cell-derived factor-1alpha and vascular endothelial growth factor) that mediate endogenous bone regeneration were also produced by expanded MSCs from SCD patients.

Conclusion: The autologous BMMC implantation with a minimally invasive technique resulted in significant pain relief and halted the progression of early stages of ONFH in SCD patients. MSCs from SCD patients display biological properties that may add to the efficiency of surgical treatment in ONFH. In summary, our results indicate that infusion of BMMCs enriched with stem/progenitor cells is a safe and effective treatment for the early stages of ONFH in SCD patients.

Trial registration: ClinicalTrials.gov NCT02448121; registered 15 May 2015.

Figures

Fig. 1
Fig. 1
BMMC treatment significantly improved the outcome of femoral heads at early-stage ONFH in SCD. ae Changes in HHS for femoral heads grouped according to Ficat stage over the entire postoperative period. Floating bars represent maximum and minimum range of variation, with lines at mean HHS. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, versus preoperative HHS values
Fig. 2
Fig. 2
Radiographic sequence from a representative case of a symptomatic 44-year-old man with ONFH successfully treated with BMMC implantation. a Coronal T1-weighted MRI scans of the right hip before surgery show osteonecrotic lesion in Ficat IIB stage. The osteonecrotic lesion is surrounded by sclerotic low-signal margin (arrow) with subjacent marrow edema. b Anteroposterior X-ray radiograph showing minimally invasive decompression. The trephine was introduced under fluoroscopy through the greater trochanter into the femoral head. The BMMCs were injected through the trephine into the necrotic zone. c, d Twelve-month postoperative coronal (c) and axial (d) MRI (T1-weighted) scans after autologous BMMC implantation showing the channel, decreased bone edema and changes of signal in the superior part of the femur. Axial MR image (d) shows well-delimited marrow region of increased signal intensity (arrow) within the necrotic area and hypointense line around them due to reparative new bone. e, f Preoperative (e) and 60-month postoperative anteroposterior (f) radiographs of the same hip showing the femoral head has maintained the sphericity (arrow)
Fig. 3
Fig. 3
Increased number of conjunctive and osteogenic progenitor cells in concentrated BMMC. a Scatter plots showing the distribution of total mononuclear cell number in 1 × 106 cells/mL after bone marrow aspiration (BMA) and after concentration of bone marrow-derived mononuclear cells (BMMC) on Ficoll density gradient centrifugation. b, c Flask and morphologic characteristics of colony-forming unit-fibroblasts (CFU-F) from BMMC. d, e Flask with mineralized matrix stained with red Alizarin-S and Crystal Violet of colony-forming unit-osteoblasts (CFU-O) from BMMC. f Frequency of CFU-F/106 cells and CFU-O/106 cells in concentrated BMMC. g Frequency of CFU-F/106 cells isolated in BMMC from individuals with osteonecrosis at a distant site was independent of the disease stage. The high standard deviation indicates the high variability of CFU-F and CFU-O within the SCD bone marrow. Data are represented as individuals values. Horizontal bars indicate mean value. Scale bar: 50 μm. ***P < 0.001. NS, not significant
Fig. 4
Fig. 4
Concentrated number of endothelial and hematopoietic progenitor cell phenotypes in BMMC. a–d The frequency of cell subpopulations was obtained after labeling of total bone marrow-derived mononuclear cells (BMMC) (1 × 106 cells) separately with antibodies (set 1 CD133/CD34/KDR or set 2 CD34/KDR/CD45) and analysis by flow cytometry. Phenotypes CD34+/KDR+(a), CD34+/KDR+/CD133+(b), CD34/CD45dim(c) and CD34+/KDR+/CD45dim (d) were concentrated in the implanted BMMC of SCD patients. e Concentration factor calculated by the quotient of the specified phenotype number in BMMC/BMA. The high standard deviation indicates the high variability of cell phenotypes within the SCD bone marrow. Data are represented as individuals values. Horizontal bars indicate mean value. *P < 0.05, **P < 0.01, ***P < 0.005, ****P < 0.001. BMA bone marrow aspirate
Fig. 5
Fig. 5
Expansion, characterization and pluripotency properties of human MSCs from SCD patients. a Ex vivo expanded mesenchymal stromal cells (MSC) from SCD patients with pre-collapse ONFH at a distant site in control culture conditions. Total viable cell counts of MSCs from donors with progressive disease stage are plotted for MSCs expanded in fetal bovine serum. b Morphologic characteristics of MSCs at passage 3. ce Differentiation of MSCs. Results of staining assays on MSCs induced to osteogenic differentiation (c, mineralized bone nodules stained with Alizarin red), adipogenic differentiation (d, intracellular lipid droplets stained with Oil red O), and chondrogenic differentiation (e, static micromass cell culture stained with Alcian Blue). f Immunophenotyping profile of MSCs isolated from SCD patients. Negative peaks (green line) correspond to cell autofluorescence. MSCs were positively labeled to CD29, CD90, CD105, CD146 and negatively labeled to CD31, CD34, CD45 and CD133. Data are mean ± SD number of cells (n = 4). Data are representative of three independent experiments. Magnification: 400× in b–d. Scale bar: 50 μm in e
Fig. 6
Fig. 6
Osteogenic abilities of bone marrow-derived MSCs are not defective in patients with osteonecrosis. a Quantitative RT-PCR of osteoblast-related genes from control and osteogenic-induced MSCs after 10 days were normalized and compared according to housekeeping genes as control. b Scatter plots (left) showing the distribution of total fluorescence intensity from FACS analysis (right) of cellular osteocalcin expression in MSCs. The relative fluorescence intensity shows significant differences between control and osteogenic differentiation conditions. c Alizarin Red staining (left) indicating mineralized nodule formation of cultured MSCs after treatment with osteogenic media for 21 days. Quantitative analysis of the amount of Alizarin staining in each group is also indicated (right). No significant difference (NS) was found between MSCs from SCD patients with early-stage osteonecrosis and patients with further stages of the disease at a distant site. Alizarin Red staining was independent of the disease stage. d Enzyme-linked immunosorbent assay for interleukin-8 (IL-8), transforming growth factor-beta (TGF-β), stromal cell-derived factor-1alpha (SDF-1α) and vascular endothelial growth factor (VEGF). Concentration (pg/mL) of cytokines and growth factors in supernatants of MSCs of SCD patients after 2 days of culture. MSCs from SCD patients produce distinct levels of IL-8. Horizontal bars represent the mean and SD of data. The triangles represent individual values. *P < 0.05, **P < 0.01, ***P < 0.001

References

    1. Adorno EV, Couto FD, Moura Neto JP, Menezes JF, Rego M, Reis MG, et al. Hemoglobinopathies in newborns from Salvador, Bahia, Northeast Brazil. Cad Saude Publica. 2005;21:292–8. doi: 10.1590/S0102-311X2005000100032.
    1. Adorno EV, Zanette A, Lyra I, Seixas MO, Reis MG, Gonçalves MS. Clinical and molecular characteristics of sickle cell anemia in the northeast of Brazil. Genet Mol Biol. 2008;31:621–5. doi: 10.1590/S1415-47572008000400003.
    1. Gonçalves MS, Queiroz IL, Cardoso SA, Zanetti A, Strapazoni AC, Adorno E, et al. Interleukin 8 as a vaso-occlusive marker in Brazilian patients with sickle cell disease. Braz J Med Biol Res. 2001;34:1309–13. doi: 10.1590/S0100-879X2001001000011.
    1. Acurio MT, Friedman RJ. Hip arthroplasty in patients with sickle-cell haemoglobinopathy. J Bone Joint Surg (Br) 1992;74:367–71.
    1. Kato GJ, Gladwin MT, Steinberg MH. Deconstructing sickle cell disease: reappraisal of the role of hemolysis in the development of clinical subphenotypes. Blood Rev. 2007;21:37–47. doi: 10.1016/j.blre.2006.07.001.
    1. Hernigou P, Habibi A, Bachir D, Galacteros F. The natural history of asymptomatic osteonecrosis of the femoral head in adults with sickle cell disease. J Bone Joint Surg Am. 2006;88:2565–72. doi: 10.2106/JBJS.E.01455.
    1. Mont MA, Zywiel MG, Marker DR, McGrath MS, Delanois RE. The natural history of untreated asymptomatic osteonecrosis of the femoral head: a systematic literature review. J Bone Joint Surg Am. 2010;92:2165–70. doi: 10.2106/JBJS.I.00575.
    1. Osunkwo I. An update on the recent literature on sickle cell bone disease. Curr Opin Endocrinol Diabetes Obes. 2013;20:539–46. doi: 10.1097/01.med.0000436192.25846.0b.
    1. Alonge TO, Shokunbi WA. The choice of arthroplasty for secondary osteoarthritis of the hip joint following avascular necrosis of the femoral head in sicklers. J Natl Med Assoc. 2004;96:678–81.
    1. Ilyas I, Moreau P. Simultaneous bilateral total hip arthroplasty in sickle cell disease. J Arthroplasty. 2002;17:441–5. doi: 10.1054/arth.2002.31084.
    1. Hernigou P, Beaujean F. Treatment of osteonecrosis with autologous bone marrow grafting. Clin Orthop Relat Res. 2002;405:14–23. doi: 10.1097/00003086-200212000-00003.
    1. Hernigou P, Flouzat-Lachaniette CH, Delambre J, Poignard A, Allain J, Chevallier N, et al. Osteonecrosis repair with bone marrow cell therapies: state of the clinical art. Bone. 2015;70:102–9. doi: 10.1016/j.bone.2014.04.034.
    1. Yan Z, Hang D, Guo C, Chen Z. Fate of mesenchymal stem cells transplanted to osteonecrosis of femoral head. J Orthop Res. 2009;27:442–6. doi: 10.1002/jor.20759.
    1. Mao Q, Jina H, Liao F, Xiao L, Chend D, Tong P. The efficacy of targeted intraarterial delivery of concentrated autologous bone marrow containing mononuclear cells in the treatment of osteonecrosis of the femoral head: a five year follow-up study. Bone. 2013;57:509–16. doi: 10.1016/j.bone.2013.08.022.
    1. Zhao D, Cui D, Wang B, Tian F, Guo L, Yang L, et al. Treatment of early stage osteonecrosis of the femoral head with autologous implantation of bone marrow derived and cultured mesenchymal stem cells. Bone. 2012;50:325–30. doi: 10.1016/j.bone.2011.11.002.
    1. Li ZH, Liao W, Cui XL, Zhao Q, Liu M, Chen YH, et al. Intravenous transplantation of allogeneic bone marrow mesenchymal stem cells and its directional migration to the necrotic femoral head. Int J Med Sci. 2011;8:74–83. doi: 10.7150/ijms.8.74.
    1. Gangji V, DeMaertelaer V, Hauzeur JP. Autologous bone marrow cell implantation in the treatment of non-traumatic osteonecrosis of the femoral head: five year follow-up of a prospective controlled study. Bone. 2011;49:1005–9. doi: 10.1016/j.bone.2011.07.032.
    1. Murphy MB, Moncivais K, Caplan AI. Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp Mol Med. 2013;45:1–16. doi: 10.1038/emm.2013.94.
    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. doi: 10.1302/0301-620X.81B2.8818.
    1. Suh KT, Kim SW, Roh HL, Youn MS, Jung JS. Decreased osteogenic differentiation of mesenchymal stem cells in alcohol induced osteonecrosis. Clin Orthop Relat Res. 2005;431:220–5. doi: 10.1097/01.blo.0000150568.16133.3c.
    1. Lee JS, Lee JS, Roh HL, Kim CH, Jung JS, Suh KT. Alterations in the differentiation ability of mesenchymal stem cells in patients with nontraumatic osteonecrosis of the femoral head: comparative analysis according to the risk factor. J Orthop Res. 2006;24:604–9. doi: 10.1002/jor.20078.
    1. Hernigou P, Beaujean F. Abnormalities in the bone marrow of the iliac crest in patients who have osteonecrosis secondary to corticosteroid therapy or alcohol abuse. J Bone Joint Surg Am. 1997;79-A:1047–53.
    1. Gangji V, Hauzeur JP, Schoutens A, Hinsenkamp M, Appelboom T, Egrise D. Abnormalities in the replicative capacity of osteoblastic cells in the proximal femur of patients with osteonecrosis of the femoral head. J Rheumatol. 2003;30:348–51.
    1. Daltro GC, Fortuna VA, de Araújo SAS, Ferraz Lessa PI, Sobrinho UA, Borojevic R. Femoral head necrosis treatment with autologous stem cells in sickle cell disease. Acta Orthop Bras. 2008;16:44–8.
    1. Mitchell MD, Kundel HL, Steinberg ME, Kressel HY, Alavi A, Axel L. Avascular necrosis of the hip: comparison of MR, CT, and scintigraphy. AJR Am J Roentgenol. 1986;147:67–71. doi: 10.2214/ajr.147.1.67.
    1. Steinberg ME, Hayken GD, Steinberg DR. A quantitative system for staging avascular necrosis. J Bone Joint Surg (Br) 1995;77:34–41.
    1. Lyra IM, Goncalves MS, Braga JA, Gesteira Mde F, Carvalho MH, Saad ST, et al. Clinical, hematological, and molecular characterization of sickle cell anemia pediatric patients from two different cities in Brazil. Cad Saude Publica. 2005;21:1287–90. doi: 10.1590/S0102-311X2005000400032.
    1. Yoo MC, Chung DW, Hahn CS. Free vascularized fibula grafting for the treatment of osteonecrosis of the femoral head. Clin Orthop Relat Res. 1992;277:128–38.
    1. Sotereanos DG, Plakseychuk AY, Rubash HE. Free vascularized fibula grafting for the treatment of osteonecrosis of the femoral head. Clin Orthop Relat Res. 1997;344:243–56. doi: 10.1097/00003086-199711000-00024.
    1. Hernigou P, Daltro G, Filippini P, Mukasa MM, Manicom O. 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. doi: 10.2174/1874325000802010062.
    1. Ahrens N, Tormin A, Paulus M, Roosterman D, Salama A, Krenn V, et al. Mesenchymal stem cell content of human vertebral bone marrow. Transplantation. 2004;78:925–9. doi: 10.1097/01.TP.0000133305.81823.2A.
    1. Baksh D, Davies JE, Zandstra PW. Adult human bone marrow-derived mesenchymal progenitor cells are capable of adhesion-independent survival and expansion. Exp Hematol. 2003;31:723–32. doi: 10.1016/S0301-472X(03)00106-1.
    1. Fadini GP, Schiavon M, Rea F, Avogaro A, Agostini C. Depletion of endothelial progenitor cells may link pulmonary fibrosis and pulmonary hypertension. Am J Respir Crit Care Med. 2007;176:724–5. doi: 10.1164/ajrccm.176.7.724a.
    1. Nevskaya T, Bykovskaia S, Lyssuk E, Shakhov I, Zaprjagaeva M, Mach E, et al. Circulating endothelial progenitor cells in systemic sclerosis: relation to impaired angiogenesis and cardiovascular manifestations. Clin Exp Rheumatol. 2008;26:421–9.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 2001;25:402–8. doi: 10.1006/meth.2001.1262.
    1. Shui C, Spelsberg TC, Riggs BL, Khosla S. Changes in Runx2/Cbfa1 expression and activity during osteoblastic differentiation of human bone marrow stromal cells. J Bone Miner Res. 2003;18:213–21. doi: 10.1359/jbmr.2003.18.2.213.
    1. Kulterer B, Friedl G, Jandrositz A, Sanchez-Cabo F, Prokesch A, Paar C, et al. Gene expression profiling of human mesenchymal stem cells derived from bone marrow during expansion and osteoblast differentiation. BMC Genomics. 2007;8:70. doi: 10.1186/1471-2164-8-70.
    1. Hernigou P, Bachir D, Galacteros F. The natural history of symptomatic osteonecrosis in adults with sickle-cell disease. J Bone Joint Surg Am. 2003;85-A:500–4.
    1. Mahadeo KM, Oyeku S, Taragin B, Rajpathak SN, Moody K, Santizo R, et al. Increased prevalence of osteonecrosis of the femoral head in children and adolescents with sickle-cell disease. Am J Hematol. 2011;86:806–8. doi: 10.1002/ajh.22103.
    1. Kim SY, Kim DH, Park IH. Multiple drilling compared with core decompression for the treatment of osteonecrosis of the femoral head. J Bone Joint Surg (Br). 2004;86:149.
    1. Martí-Carvajal AJ, Solà I, Agreda-Pérez LH. Treatment for avascular necrosis of bone in people with sickle cell disease. Cochrane Database Syst Rev. 2014;10:7.
    1. Neumayr LD, Aguilar C, Earles AN, Jergesen HE, Haberkern CM, Kammen BF, et al. Physical therapy alone compared with core decompression and physical therapy for femoral head osteonecrosis in sickle cell disease. Results of a multicenter study at a mean of three years after treatment. J Bone Joint Surg Am. 2006;88:2573–82. doi: 10.2106/JBJS.E.01454.
    1. Mukisi-Mukaza M, Manicom O, Alexis C, Bashoun K, Donkerwolcke M, Burny F. Treatment of sickle cell disease’s hip necrosis by core decompression: a prospective case–control study. Orthop Traumatol Surg Res. 2009;95:498–504. doi: 10.1016/j.otsr.2009.07.009.
    1. Al OA. Multiple drilling compared with standard core decompression for avascular necrosis of the femoral head in sickle cell disease patients. Arch Orthop Trauma Surg. 2013;133:609–13. doi: 10.1007/s00402-013-1714-9.
    1. Sen RK, Tripathy SK, Aggarwal S, Marwaha N, Sharma RR, Khandelwal N. Early results of core decompression and autologous bone marrow mononuclear cells instillation in femoral head osteonecrosis: a randomized control study. J Arthroplasty. 2012;27:679–86. doi: 10.1016/j.arth.2011.08.008.
    1. Wang BL, Sun W, Shi ZC, Zhang NF, Yue DB, Guo WS, et al. Treatment of nontraumatic osteonecrosis of the femoral head with the implantation of core decompression and concentrated autologous bone marrow containing mononuclear cells. Arch Orthop Trauma Surg. 2010;130:859–65. doi: 10.1007/s00402-009-0939-0.
    1. Ma Y, Wang T, Liao J, Gu H, Lin X, Jiang Q, et al. Efficacy of autologous bone marrow buffy coat grafting combined with core decompression in patients with avascular necrosis of femoral head: a prospective, double-blinded, randomized, controlled study. Stem Cell Res Ther. 2014;5:115. doi: 10.1186/scrt505.
    1. Hernigou P, Zilber S, Filippini P, Mathieu G, Poignard A, Galacteros F. Total THA in adult osteonecrosis related to sickle cell disease. Clin Orthop Relat Res. 2008;466:300–8. doi: 10.1007/s11999-007-0069-3.
    1. Vichinsky EP, Neumayr LD, Haberkern C, Earles AN, Eckman J, Koshy M, et al. The perioperative complication rate of orthopedic surgery in sickle cell disease: report of the National Sickle Cell Surgery Study Group. Am J Hematol. 1999;62:129–38. doi: 10.1002/(SICI)1096-8652(199911)62:3<129::AID-AJH1>;2-J.
    1. Schneider W, Knahr K. Total hip replacement in younger patients: survival rate after avascular necrosis of the femoral head. Acta Orthop Scand. 2004;75:142–6. doi: 10.1080/00016470412331294385.
    1. Issa K, Naziri Q, Maheshwari AV, Rasquinha VJ, Delanois RE, Mont MA. Excellent results and minimal complications of total hip arthroplasty in sickle cell hemoglobinopathy at mid-term follow-up using cementless prosthetic components. J Arthroplasty. 2013;28:1693–8. doi: 10.1016/j.arth.2013.03.017.
    1. Yoo JJ, Song WS, Koo KH, Yoon KS, Kim HJ. Osteogenic abilities of bone marrow stromal cells are not defective in patients with osteonecrosis. Int Orthop. 2009;33:867–72. doi: 10.1007/s00264-008-0524-0.
    1. Siddappa R, Licht R, van Blitterswijk C, de Boer J. Donor variation and loss of multipotency during in vitro expansion of human mesenchymal stem cells for bone tissue engineering. J Orthop Res. 2007;25:1029–41. doi: 10.1002/jor.20402.
    1. Bostrom MP, Asnis P. Transforming growth factor beta in fracture repair. Clin Orthop Relat Res. 1998;355:124–31. doi: 10.1097/00003086-199810001-00014.

Source: PubMed

3
Iratkozz fel