Skip to main content

Advertisement

Log in

Clinical translation of controlled protein delivery systems for tissue engineering

  • Review Article
  • Published:
Drug Delivery and Translational Research Aims and scope Submit manuscript

Abstract

Strategies that utilize controlled release of drugs and proteins for tissue engineering have enormous potential to regenerate damaged organs and tissues. The multiple advantages of controlled release strategies merit overcoming the significant challenges to translation, including high costs and long, difficult regulatory pathways. This review highlights the potential of controlled release of proteins for tissue engineering and regenerative medicine. We specifically discuss treatment modalities that have reached preclinical and clinical trials, with emphasis on controlled release systems for bone tissue engineering, the most advanced application with several products already in clinic. Possible strategies to address translational and regulatory concerns are also discussed.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Jaklenec A et al. Progress in the tissue engineering and stem cell industry "are we there yet?". Tissue Eng Part B Rev. 2012;18(3):155–66.

    Article  PubMed  Google Scholar 

  2. Medtronic. Medtronic INFUSE® Bone Graft + LT-CAGE® lumbar tapered fusion device fact sheet. 2012. http://wwwp.medtronic.com/Newsroom/LinkedItemDetails.do?itemId=1101769224707&itemType=fact_sheet&lang=en_US.

  3. Betz VM et al. Bone tissue engineering and repair by gene therapy. Front Biosci. 2008;13:833–41.

    Article  CAS  PubMed  Google Scholar 

  4. Dang JM, Leong KW. Natural polymers for gene delivery and tissue engineering. Adv Drug Deliv Rev. 2006;58(4):487–99.

    Article  CAS  PubMed  Google Scholar 

  5. Place ES, Evans ND, Stevens MM. Complexity in biomaterials for tissue engineering. Nat Mater. 2009;8(6):457–70.

    Article  CAS  PubMed  Google Scholar 

  6. Tessmar JK, Gopferich AM. Matrices and scaffolds for protein delivery in tissue engineering. Adv Drug Deliv Rev. 2007;59(4–5):274–91.

    Article  CAS  PubMed  Google Scholar 

  7. Puleo DA, Kissling RA, Sheu MS. A technique to immobilize bioactive proteins, including bone morphogenetic protein-4 (BMP-4), on titanium alloy. Biomaterials. 2002;23(9):2079–87.

    Article  CAS  PubMed  Google Scholar 

  8. Lee M et al. Modulation of protein delivery from modular polymer scaffolds. Biomaterials. 2007;28(10):1862–70.

    Article  CAS  PubMed  Google Scholar 

  9. Uludag H et al. Characterization of rhBMP-2 pharmacokinetics implanted with biomaterial carriers in the rat ectopic model. J Biomed Mater Res. 1999;46(2):193–202.

    Article  CAS  PubMed  Google Scholar 

  10. Jeon O et al. Enhancement of ectopic bone formation by bone morphogenetic protein-2 released from a heparin-conjugated poly(L-lactic-co-glycolic acid) scaffold. Biomaterials. 2007;28(17):2763–71.

    Article  CAS  PubMed  Google Scholar 

  11. Miller RE et al. Intraarticular injection of heparin-binding insulin-like growth factor 1 sustains delivery of insulin-like growth factor 1 to cartilage through binding to chondroitin sulfate. Arthritis Rheum. 2010;62(12):3686–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Vidal G et al. Enhanced cellular adhesion on titanium by silk functionalized with titanium binding and RGD peptides. Acta Biomater. 2013;9(1):4935–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kanczler JM et al. The effect of mesenchymal populations and vascular endothelial growth factor delivered from biodegradable polymer scaffolds on bone formation. Biomaterials. 2008;29(12):1892–900.

    Article  CAS  PubMed  Google Scholar 

  14. Kanczler JM et al. The effect of the delivery of vascular endothelial growth factor and bone morphogenic protein-2 to osteoprogenitor cell populations on bone formation. Biomaterials. 2010;31(6):1242–50.

    Article  CAS  PubMed  Google Scholar 

  15. Siepmann F et al. Polymer blends for controlled release coatings. J Control Release. 2008;125(1):1–15.

    Article  CAS  PubMed  Google Scholar 

  16. Wang X et al. Controlled release from multilayer silk biomaterial coatings to modulate vascular cell responses. Biomaterials. 2008;29(7):894–903.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wang X et al. Silk coatings on PLGA and alginate microspheres for protein delivery. Biomaterials. 2007;28(28):4161–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Xue W, Bandyopadhyay A, Bose S. Polycaprolactone coated porous tricalcium phosphate scaffolds for controlled release of protein for tissue engineering. J Biomed Mater Res B Appl Biomater. 2009;91(2):831–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Wu G et al. Biomimetic coating of organic polymers with a protein-functionalized layer of calcium phosphate: the surface properties of the carrier influence neither the coating characteristics nor the incorporation mechanism or release kinetics of the protein. Tissue Eng Part C Methods. 2010;16(6):1255–65.

    Article  CAS  PubMed  Google Scholar 

  20. Lee JS, Suarez-Gonzalez D, Murphy WL. Mineral coatings for temporally controlled delivery of multiple proteins. Adv Mater. 2011;23(37):4279–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Rezwan K et al. Biodegradable and bioactive porous polymer/inorganic composite scaffolds for bone tissue engineering. Biomaterials. 2006;27(18):3413–31.

    Article  CAS  PubMed  Google Scholar 

  22. Ariga K et al. Layer-by-layer assembly for drug delivery and related applications. Expert Opin Drug Deliv. 2011;8(5):633–44.

    Article  CAS  PubMed  Google Scholar 

  23. Macdonald ML et al. Tissue integration of growth factor-eluting layer-by-layer polyelectrolyte multilayer coated implants. Biomaterials. 2011;32(5):1446–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wenk E et al. Microporous silk fibroin scaffolds embedding PLGA microparticles for controlled growth factor delivery in tissue engineering. Biomaterials. 2009;30(13):2571–81.

    Article  CAS  PubMed  Google Scholar 

  25. Kempen DH et al. Retention of in vitro and in vivo BMP-2 bioactivities in sustained delivery vehicles for bone tissue engineering. Biomaterials. 2008;29(22):3245–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bian LM et al. Enhanced MSC chondrogenesis following delivery of TGF-beta 3 from alginate microspheres within hyaluronic acid hydrogels in vitro and in vivo. Biomaterials. 2011;32(27):6425–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Saif J et al. Combination of injectable multiple growth factor-releasing scaffolds and cell therapy as an advanced modality to enhance tissue neovascularization. Arterioscler Thromb Vasc Biol. 2010;30(10):1897–904.

    Article  CAS  PubMed  Google Scholar 

  28. Jaklenec A et al. Sequential release of bioactive IGF-I and TGF-beta 1 from PLGA microsphere-based scaffolds. Biomaterials. 2008;29(10):1518–25.

    Article  CAS  PubMed  Google Scholar 

  29. Jabbarzadeh E et al. VEGF-incorporated biomimetic poly(lactide-co-glycolide) sintered microsphere scaffolds for bone tissue engineering. J Biomed Mater Res B-Appl Biomater. 2012;100B(8):2187–96.

    Article  CAS  Google Scholar 

  30. Johnson PJ et al. Maintaining bioactivity of NGF for controlled release from PLGA using PEG. J Biomed Mater Res A. 2008;86(2):420–7.

    Article  PubMed  CAS  Google Scholar 

  31. Park H et al. Delivery of TGF-beta1 and chondrocytes via injectable, biodegradable hydrogels for cartilage tissue engineering applications. Biomaterials. 2005;26(34):7095–103.

    Article  CAS  PubMed  Google Scholar 

  32. Lowman A, Peppas N. Hydrogels. In: Mathiowitz E, editor. Encyclopedia of controlled drug delivery. New York: Wiley; 1999. p. 397–406.

  33. Fortier LA et al. Insulin-like growth factor-I enhances cell-based repair of articular cartilage. J Bone Joint Surg Br. 2002;84(2):276–88.

    Article  CAS  PubMed  Google Scholar 

  34. Nixon AJ et al. Enhanced repair of extensive articular defects by insulin-like growth factor-I-laden fibrin composites. J Orthop Res. 1999;17:475–87.

    Article  CAS  PubMed  Google Scholar 

  35. Gratz KR et al. Biomechanical assessment of tissue retrieved after in vivo cartilage defect repair: tensile modulus of repair tissue and integration with host cartilage. J Biomech. 2006;39(1):138–46.

    Article  PubMed  Google Scholar 

  36. Borselli C et al. Functional muscle regeneration with combined delivery of angiogenesis and myogenesis factors. Proc Natl Acad Sci U S A. 2010;107(8):3287–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Jeon O et al. Long-term delivery enhances in vivo osteogenic efficacy of bone morphogenetic protein-2 compared to short-term delivery. Biochem Biophys Res Commun. 2008;369(2):774–80.

    Article  CAS  PubMed  Google Scholar 

  38. Lo, KW, et al., Studies of bone morphogenetic protein based surgical repair. Adv Drug Deliv Rev. 2012;64:1277–91.

    Google Scholar 

  39. Orthopaedic and Rehabilitation Devices Advisory Panel, F, Executive Summary for P050036 Medtronic’s AMPLIFY rhBMP-2 Matrix, 2010. p. 69.

  40. FDA. Approval letter of INFUSE bone graft—P050053. Silver Spring: FDA; 2007.

  41. FDA. Approval letter for INFUSE bone graft—P000054. Silver Spring: FDA; 2004.

  42. McKay WF, Peckham SM, Badura JM. A comprehensive clinical review of recombinant human bone morphogenetic protein-2 (INFUSE Bone Graft). Int Orthop. 2007;31(6):729–34.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Glassman SD et al. The efficacy of rhBMP-2 for posterolateral lumbar fusion in smokers. Spine (Phila Pa 1976). 2007;32(15):1693–8.

    Article  Google Scholar 

  44. FDA. Approval letter for OP-1 Implant—H010002. Silver Spring: FDA; 2001.

  45. Carragee EJ, Hurwitz EL, Weiner BK. A critical review of recombinant human bone morphogenetic protein-2 trials in spinal surgery: emerging safety concerns and lessons learned. Spine J. 2011;11(6):471–91.

    Article  PubMed  Google Scholar 

  46. Boerckel JD et al. Effects of protein dose and delivery system on BMP-mediated bone regeneration. Biomaterials. 2011;32(22):?>5241–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Brown KV et al. Improving bone formation in a rat femur segmental defect by controlling bone morphogenetic protein-2 release. Tissue Eng Part A. 2011;17(13–14):1735–46.

    Article  CAS  PubMed  Google Scholar 

  48. Bessa PC, Casal M, Reis RL. Bone morphogenetic proteins in tissue engineering: the road from laboratory to clinic, part II (BMP delivery). J Tissue Eng Regen Med. 2008;2(2–3):81–96.

    Article  CAS  PubMed  Google Scholar 

  49. Vo, TN, FK Kasper, and AG Mikos, Strategies for controlled delivery of growth factors and cells for bone regeneration. Adv Drug Deliv Rev. 2012:64;1292–309.

  50. Edelman ER et al. Controlled and modulated release of basic fibroblast growth factor. Biomaterials. 1991;12(7):619–26.

    Article  CAS  PubMed  Google Scholar 

  51. Laham RJ et al. Local perivascular delivery of basic fibroblast growth factor in patients undergoing coronary bypass surgery: results of a phase I randomized, double-blind, placebo-controlled trial. Circulation. 1999;100(18):1865–71.

    Article  CAS  PubMed  Google Scholar 

  52. Ruel M et al. Long-term effects of surgical angiogenic therapy with fibroblast growth factor 2 protein. J Thorac Cardiovasc Surg. 2002;124(1):28–34.

    Article  CAS  PubMed  Google Scholar 

  53. Le KN et al. Vascular regeneration by local growth factor release is self-limited by microvascular clearance. Circulation. 2009;119(22):2928–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Roh JD et al. Tissue-engineered vascular grafts transform into mature blood vessels via an inflammation-mediated process of vascular remodeling. Proc Natl Acad Sci U S A. 2010;107(10):4669–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Matsumura G et al. Successful application of tissue engineered vascular autografts: clinical experience. Biomaterials. 2003;24(13):2303–8.

    Article  CAS  PubMed  Google Scholar 

  56. Shin'oka T et al. Midterm clinical result of tissue-engineered vascular autografts seeded with autologous bone marrow cells. J Thorac Cardiovasc Surg. 2005;129(6):1330–8.

    Article  PubMed  Google Scholar 

  57. Richardson TP, Murphy WL, Mooney DJ. Polymeric delivery of proteins and plasmid DNA for tissue engineering and gene therapy. Crit Rev Eukaryot Gene Expr. 2001;11(1–3):47–58.

    CAS  PubMed  Google Scholar 

  58. Panyam J et al. Polymer degradation and in vitro release of a model protein from poly(D, L-lactide-co-glycolide) nano- and microparticles. J Control Release. 2003;92(1–2):173–87.

    Article  CAS  PubMed  Google Scholar 

  59. Cohen S et al. Controlled delivery systems for proteins based on poly(lactic/glycolic acid) microspheres. Pharm Res. 1991;8(6):713–20.

    Article  CAS  PubMed  Google Scholar 

  60. Bae SE et al. Effect of temporally controlled release of dexamethasone on in vivo chondrogenic differentiation of mesenchymal stromal cells. J Control Release. 2010;143(1):23–30.

    Article  CAS  PubMed  Google Scholar 

  61. Shi X et al. Sintered microsphere scaffolds for controlled release and tissue engineering. Pharm Res. 2011;28(5):1224–8.

    Article  CAS  PubMed  Google Scholar 

  62. Spiller KL et al. A novel method for the direct fabrication of growth factor-loaded microspheres within porous nondegradable hydrogels: controlled release for cartilage tissue engineering. J Control Release. 2012;157(1):39–45.

    Article  CAS  PubMed  Google Scholar 

  63. DeFail AJ et al. Controlled release of bioactive TGF-beta 1 from microspheres embedded within biodegradable hydrogels. Biomaterials. 2006;27(8):1579–85.

    Article  CAS  PubMed  Google Scholar 

  64. Holland TA, Tabata Y, Mikos AG. In vitro release of transforming growth factor-beta 1 from gelatin microparticles encapsulated in biodegradable, injectable oligo(poly(ethylene glycol) fumarate) hydrogels. J Control Release. 2003;91(3):299–313.

    Article  CAS  PubMed  Google Scholar 

  65. Holland TA, Tabata Y, Mikos AG. Dual growth factor delivery from degradable oligo(poly(ethylene glycol) fumarate) hydrogel scaffolds for cartilage tissue engineering. J Control Release. 2005;101(1–3):111–25.

    Article  CAS  PubMed  Google Scholar 

  66. Geuze, R, et al., A differential effect of BMP-2 and VEGF release timing on osteogenesis at ectopic and orthotopic sites in a large animal model. Tissue Eng Part A. 2012;18:2052–62.

    Google Scholar 

  67. Freeman I, Cohen S. The influence of the sequential delivery of angiogenic factors from affinity-binding alginate scaffolds on vascularization. Biomaterials. 2009;30(11):2122–31.

    Article  CAS  PubMed  Google Scholar 

  68. Sun Q et al. Sustained release of multiple growth factors from injectable polymeric system as a novel therapeutic approach towards angiogenesis. Pharm Res. 2010;27(2):264–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Hao X et al. Angiogenic effects of sequential release of VEGF-A165 and PDGF-BB with alginate hydrogels after myocardial infarction. Cardiovasc Res. 2007;75(1):178–85.

    Article  CAS  PubMed  Google Scholar 

  70. Ruvinov E, Leor J, Cohen S. The promotion of myocardial repair by the sequential delivery of IGF-1 and HGF from an injectable alginate biomaterial in a model of acute myocardial infarction. Biomaterials. 2011;32(2):565–78.

    Article  CAS  PubMed  Google Scholar 

  71. Simons M et al. Pharmacological treatment of coronary artery disease with recombinant fibroblast growth factor-2: double-blind, randomized, controlled clinical trial. Circulation. 2002;105(7):788–93.

    Article  CAS  PubMed  Google Scholar 

  72. Henry TD et al. The VIVA trial: vascular endothelial growth factor in ischemia for vascular angiogenesis. Circulation. 2003;107(10):1359–65.

    Article  CAS  PubMed  Google Scholar 

  73. Zachary I, Morgan RD. Therapeutic angiogenesis for cardiovascular disease: biological context, challenges, prospects. Heart. 2011;97(3):181–9.

    Article  CAS  PubMed  Google Scholar 

  74. Obradovic B et al. Integration of engineered cartilage. J Orthop Res. 2001;19(6):1089–97.

    Article  CAS  PubMed  Google Scholar 

  75. Fan H et al. Porous gelatin-chondroitin-hyaluronate tri-copolymer scaffold containing microspheres loaded with TGF-beta1 induces differentiation of mesenchymal stem cells in vivo for enhancing cartilage repair. J Biomed Mater Res A. 2006;77(4):785–94.

    Article  PubMed  CAS  Google Scholar 

  76. Garbern JC et al. Delivery of basic fibroblast growth factor with a pH-responsive, injectable hydrogel to improve angiogenesis in infarcted myocardium. Biomaterials. 2011;32(9):2407–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Zisch AH et al. Cell-demanded release of VEGF from synthetic, biointeractive cell ingrowth matrices for vascularized tissue growth. FASEB J. 2003;17(15):2260–2.

    CAS  PubMed  Google Scholar 

  78. Ehrbar M et al. Cell-demanded liberation of VEGF121 from fibrin implants induces local and controlled blood vessel growth. Circ Res. 2004;94(8):1124–32.

    Article  CAS  PubMed  Google Scholar 

  79. Phelps EA et al. Bioartificial matrices for therapeutic vascularization. Proc Natl Acad Sci U S A. 2010;107(8):3323–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Park J et al. Nerve regeneration following spinal cord injury using matrix metalloproteinase-sensitive, hyaluronic acid-based biomimetic hydrogel scaffold containing brain-derived neurotrophic factor. J Biomed Mater Res A. 2010;93(3):1091–9.

    PubMed  Google Scholar 

  81. Terella A et al. Repair of a calvarial defect with biofactor and stem cell-embedded polyethylene glycol scaffold. Arch Facial Plast Surg. 2010;12(3):166–71.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Kim J et al. In vivo evaluation of MMP sensitive high-molecular weight HA-based hydrogels for bone tissue engineering. J Biomed Mater Res A. 2010;95(3):673–81.

    Article  PubMed  CAS  Google Scholar 

  83. Liu Y, de Groot K, Hunziker EB. BMP-2 liberated from biomimetic implant coatings induces and sustains direct ossification in an ectopic rat model. Bone. 2005;36(5):745–57.

    Article  CAS  PubMed  Google Scholar 

  84. Liu Y et al. The influence of BMP-2 and its mode of delivery on the osteoconductivity of implant surfaces during the early phase of osseointegration. Biomaterials. 2007;28(16):2677–86.

    Article  CAS  PubMed  Google Scholar 

  85. Hagi TT et al. Cell-mediated BMP-2 liberation promotes bone formation in a mechanically unstable implant environment. Bone. 2010;46(5):1322–7.

    Article  PubMed  CAS  Google Scholar 

  86. Lee K, Silva EA, Mooney DJ. Growth factor delivery-based tissue engineering: general approaches and a review of recent developments. J R Soc Interface. 2011;8(55):153–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Kloxin AM et al. Photodegradable hydrogels for dynamic tuning of physical and chemical properties. Science. 2009;324(5923):59–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Yavuz MS et al. Gold nanocages covered by smart polymers for controlled release with near-infrared light. Nat Mater. 2009;8(12):935–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Uesugi Y et al. An ultrasound-responsive nano delivery system of tissue-type plasminogen activator for thrombolytic therapy. J Contr Release. 2010;147(2):269–77.

    Article  CAS  Google Scholar 

  90. Deckers R, Moonen CTW. Ultrasound triggered, image guided, local drug delivery. J Contr Release. 2010;148(1):25–33.

    Article  CAS  Google Scholar 

  91. El-Bialy T et al. In vivo ultrasound-assisted tissue-engineered mandibular condyle: a pilot study in rabbits. Tissue Eng C-Methods. 2010;16(6):1315–23.

    Article  Google Scholar 

  92. Giannoni P et al. Species variability in the differentiation potential of in vitro-expanded articular chondrocytes restricts predictive studies on cartilage repair using animal models. Tissue Eng. 2005;11(1–2):237–48.

    Article  CAS  PubMed  Google Scholar 

  93. Loeser RF et al. Reduction in the chondrocyte response to insulin-like growth factor 1 in aging and osteoarthritis: studies in a non-human primate model of naturally occurring disease. Arthritis Rheum. 2000;43(9):2110–20.

    Article  CAS  PubMed  Google Scholar 

  94. Yasuda A et al. In vitro culture of chondrocytes in a novel thermoreversible gelation polymer scaffold containing growth factors. Tissue Eng. 2006;12(5):1237–45.

    Article  CAS  PubMed  Google Scholar 

  95. Elisseeff J et al. Controlled-release of IGF-I and TGF-beta1 in a photopolymerizing hydrogel for cartilage tissue engineering. J Orthop Res. 2001;19(6):1098–104.

    Article  CAS  PubMed  Google Scholar 

  96. Holland TA et al. Degradable hydrogel scaffolds for in vivo delivery of single and dual growth factors in cartilage repair. Osteoarthr Cartil. 2007;15(2):187–97.

    Article  CAS  PubMed  Google Scholar 

  97. Gadjanski I, Spiller K, Vunjak-Novakovic G. Time-dependent processes in stem cell-based tissue engineering of articular cartilage. Stem Cell Rev. 2012;8(3):863–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Kempen DH et al. Effect of local sequential VEGF and BMP-2 delivery on ectopic and orthotopic bone regeneration. Biomaterials. 2009;30(14):2816–25.

    Article  CAS  PubMed  Google Scholar 

  99. Patel ZS et al. Dual delivery of an angiogenic and an osteogenic growth factor for bone regeneration in a critical size defect model. Bone. 2008;43(5):931–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Young S et al. Dose effect of dual delivery of vascular endothelial growth factor and bone morphogenetic protein-2 on bone regeneration in a rat critical-size defect model. Tissue Eng Part A. 2009;15(9):2347–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Street J et al. Vascular endothelial growth factor stimulates bone repair by promoting angiogenesis and bone turnover. Proc Natl Acad Sci U S A. 2002;99(15):9656–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Lee JY et al. Enhanced bone formation by controlled growth factor delivery from chitosan-based biomaterials. J Contr Release. 2002;78(1–3):187–97.

    Article  CAS  Google Scholar 

  103. Peng H et al. VEGF improves, whereas sFlt1 inhibits, BMP2-induced bone formation and bone healing through modulation of angiogenesis. J Bone Miner Res. 2005;20(11):2017–27.

    Article  CAS  PubMed  Google Scholar 

  104. Hunziker E et al. Translation from research to applications. Tissue Eng. 2006;12(12):3341–64.

    Article  CAS  PubMed  Google Scholar 

  105. Moya ML et al. The effect of FGF-1 loaded alginate microbeads on neovascularization and adipogenesis in a vascular pedicle model of adipose tissue engineering. Biomaterials. 2010;31(10):2816–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Simons M. Angiogenesis: where do we stand now? Circulation. 2005;111(12):1556–66.

    Article  PubMed  Google Scholar 

  107. Burkhoff D, Jones JW, Becker LC. Variability of myocardial perfusion defects assessed by thallium-201 scintigraphy in patients with coronary artery disease not amenable to angioplasty or bypass surgery. J Am Coll Cardiol. 2001;38(4):1033–9.

    Article  CAS  PubMed  Google Scholar 

  108. Wallace H. Coulter Foundation. 2012. http://www.whcf.org/.

  109. Hollister SJ, Murphy WL. Scaffold translation: barriers between concept and clinic. Tissue Eng B Rev. 2011;17(6):459–74.

    Article  Google Scholar 

  110. FDA. Overview of medical devices and their regulatory pathways. 2011. http://www.fda.gov/AboutFDA/CentersOffices/OfficeofMedicalProductsandTobacco/CDRH/CDRHTransparency/ucm203018.htm.

  111. Lee MH et al. Considerations for tissue-engineered and regenerative medicine product development prior to clinical trials in the United States. Tissue Eng B Rev. 2010;16(1):41–54.

    Article  CAS  Google Scholar 

  112. FDA. About combination products. 2009. http://www.fda.gov/CombinationProducts/AboutCombinationProducts/default.htm.

  113. FDA. Tissue Engineered Medical Products Standards (TEMPS). 2009. http://www.fda.gov/MedicalDevices/DeviceRegulationandGuidance/Standards/ucm135369.htm.

  114. FDA. Device advice: investigation device exemption. Silver Spring: FDA; 2012.

  115. FDA. PMA clinical studies. Silver Spring: FDA; 2010.

  116. FDA. Tissue guidance documents. http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/Tissue/default.htm. Accessed 22 Dec 2012.

  117. Ramazanoglu M et al. The effect of combined delivery of recombinant human bone morphogenetic protein-2 and recombinant human vascular endothelial growth factor 165 from biomimetic calcium-phosphate-coated implants on osseointegration. Clin Oral Implants Res. 2011;22(12):1433–9.

    Article  PubMed  Google Scholar 

  118. Schmitt, CM, et al., Histological results after maxillary sinus augmentation with Straumann(R) BoneCeramic, Bio-Oss(R), Puros(R), and autologous bone. A randomized controlled clinical trial. Clin Oral Implants Res. 2013. doi:10.1111/j.1600-0501.2012.02431.x.

  119. Li J et al. Repair of rat cranial bone defects with nHAC/PLLA and BMP-2-related peptide or rhBMP-2. J Orthop Res. 2011;29(11):1745–52.

    Article  CAS  PubMed  Google Scholar 

  120. Li M et al. Calcium phosphate cement with BMP-2-loaded gelatin microspheres enhances bone healing in osteoporosis: a pilot study. Clin Orthop Relat Res. 2010;468(7):1978–85.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Kolambkar YM et al. An alginate-based hybrid system for growth factor delivery in the functional repair of large bone defects. Biomaterials. 2011;32(1):65–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Kaipel, M, et al., BMP-2 but not VEGF or PDGF in fibrin matrix supports bone healing in a delayed-union rat model. J Orthop Res. 2012;30:1563–9.

    Google Scholar 

  123. Fujioka-Kobayashi, M, et al., Cholesteryl group-and acryloyl group-bearing pullulan nanogel to deliver BMP2 and FGF18 for bone tissue engineering. Biomaterials. 2012;33:7613–20.

    Google Scholar 

  124. Yamamoto M, Takahashi Y, Tabata Y. Enhanced bone regeneration at a segmental bone defect by controlled release of bone morphogenetic protein-2 from a biodegradable hydrogel. Tissue Eng. 2006;12(5):1305–11.

    Article  CAS  PubMed  Google Scholar 

  125. Phillips FM et al. In vivo BMP-7 (OP-1) enhancement of osteoporotic vertebral bodies in an ovine model. Spine J. 2006;6(5):500–6.

    Article  PubMed  Google Scholar 

  126. De la Riva B et al. Local controlled release of VEGF and PDGF from a combined brushite-chitosan system enhances bone regeneration. J Control Release. 2010;143(1):45–52.

    Article  PubMed  CAS  Google Scholar 

  127. Nakahara T et al. In situ tissue engineering of periodontal tissues by seeding with periodontal ligament-derived cells. Tissue Eng. 2004;10(3–4):537–44.

    Article  CAS  PubMed  Google Scholar 

  128. Liu XW et al. Insulin-like growth factor-1 suspended in hyaluronan improves cartilage and subchondral cancellous bone repair in osteoarthritis of temporomandibular joint. Int J Oral Maxillofac Surg. 2011;40(2):184–90.

    Article  PubMed  Google Scholar 

  129. Batten ML, Hansen JC, Dahners LE. Influence of dosage and timing of application of platelet-derived growth factor on early healing of the rat medial collateral ligament. J Orthop Res. 1996;14(5):736–41.

    Article  CAS  PubMed  Google Scholar 

  130. Conova L et al. A pilot study of poly(N-isopropylacrylamide)-g-polyethylene glycol and poly(N-isopropylacrylamide)-g-methylcellulose branched copolymers as injectable scaffolds for local delivery of neurotrophins and cellular transplants into the injured spinal cord. J Neurosurg Spine. 2011;15(6):594–604.

    Article  PubMed  Google Scholar 

  131. Hiraoka Y et al. In situ regeneration of adipose tissue in rat fat pad by combining a collagen scaffold with gelatin microspheres containing basic fibroblast growth factor. Tissue Eng. 2006;12(6):1475–87.

    Article  CAS  PubMed  Google Scholar 

  132. Vashi AV et al. Adipose tissue engineering based on the controlled release of fibroblast growth factor-2 in a collagen matrix. Tissue Eng. 2006;12(11):3035–43.

    Article  CAS  PubMed  Google Scholar 

  133. Huang Q et al. In Vivo mesenchymal cell recruitment by a scaffold loaded with transforming growth factor b1 and the potential for in situ chondrogenesis. Tissue Eng. 2002;8(3):469–82.

    Article  CAS  PubMed  Google Scholar 

  134. Zhang G et al. Controlled release of stromal cell-derived factor-1 alpha in situ increases c-kit + cell homing to the infarcted heart. Tissue Eng. 2007;13(8):2063–71.

    Article  CAS  PubMed  Google Scholar 

  135. Thevenot PT et al. The pivotal role of fibrocytes and mast cells in mediating fibrotic reactions to biomaterials. Biomaterials. 2011;32(33):8394–403.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Schantz JT, Chim H, Whiteman M. Cell guidance in tissue engineering: SDF-1 mediates site-directed homing of mesenchymal stem cells within three-dimensional polycaprolactone scaffolds. Tissue Eng. 2007;13(11):2615–24.

    Article  CAS  PubMed  Google Scholar 

  137. Chen RR et al. Spatio-temporal VEGF and PDGF delivery patterns blood vessel formation and maturation. Pharm Res. 2007;24(2):258–64.

    Article  PubMed  CAS  Google Scholar 

  138. Holland TA et al. Osteochondral repair in the rabbit model utilizing bilayered, degradable oligo(poly(ethylene glycol) fumarate) hydrogel scaffolds. J Biomed Mater Res A. 2005;75(1):156–67.

    Article  PubMed  CAS  Google Scholar 

  139. Yilgor, P, et al., An in vivo study on the effect of scaffold geometry and growth factor release on the healing of bone defects. J Tissue Eng Regen Med. 2013. doi:10.1002/term.1456.

  140. Matsuse D et al. Combined transplantation of bone marrow stromal cell-derived neural progenitor cells with a collagen sponge and basic fibroblast growth factor releasing microspheres enhances recovery after cerebral ischemia in rats. Tissue Eng Part A. 2011;17(15–16):1993–2004.

    Article  CAS  PubMed  Google Scholar 

  141. Lutolf MP et al. Repair of bone defects using synthetic mimetics of collagenous extracellular matrices. Nat Biotechnol. 2003;21(5):513–8.

    Article  CAS  PubMed  Google Scholar 

  142. Wu G et al. Functionalization of deproteinized bovine bone with a coating-incorporated depot of BMP-2 renders the material efficiently osteoinductive and suppresses foreign-body reactivity. Bone. 2011;49(6):1323–30.

    Article  CAS  PubMed  Google Scholar 

  143. Klenke FM et al. Sustained release of VEGF from CaP ceramics promotes biomaterial vascularization in vivo. J Bone Joint Surg Orthop Proc. 2010;92(IV):617.

    Google Scholar 

  144. Tengood JE et al. Sequential delivery of vascular endothelial growth factor and sphingosine 1-phosphate for angiogenesis. Biomaterials. 2010;31(30):7805–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Govender S et al. Recombinant human bone morphogenetic protein-2 for treatment of open tibial fractures: a prospective, controlled, randomized study of four hundred and fifty patients. J Bone Joint Surg Am. 2002;84-A(12):2123–34.

    PubMed  Google Scholar 

  146. Swiontkowski MF et al. Recombinant human bone morphogenetic protein-2 in open tibial fractures. A subgroup analysis of data combined from two prospective randomized studies. J Bone Joint Surg Am. 2006;88(6):1258–65.

    Article  PubMed  Google Scholar 

  147. Alt V et al. A health economic analysis of the use of rhBMP-2 in Gustilo–Anderson grade III open tibial fractures for the UK, Germany, and France. Injury. 2009;40(12):1269–75.

    Article  PubMed  Google Scholar 

  148. Aro HT et al. Recombinant human bone morphogenetic protein-2: a randomized trial in open tibial fractures treated with reamed nail fixation. J Bone Joint Surg Am. 2011;93(9):801–8.

    Article  PubMed  Google Scholar 

  149. Burkus JK et al. Clinical and radiographic outcomes of anterior lumbar interbody fusion using recombinant human bone morphogenetic protein-2. Spine (Phila Pa 1976). 2002;27(21):2396–408.

    Article  Google Scholar 

  150. Baskin DS et al. A prospective, randomized, controlled cervical fusion study using recombinant human bone morphogenetic protein-2 with the CORNERSTONE-SR allograft ring and the ATLANTIS anterior cervical plate. Spine (Phila Pa 1976). 2003;28(12):1219–24. discussion 1225.

    Google Scholar 

  151. Shields LB et al. Adverse effects associated with high-dose recombinant human bone morphogenetic protein-2 use in anterior cervical spine fusion. Spine (Phila Pa 1976). 2006;31(5):542–7.

    Article  Google Scholar 

  152. Burkus JK et al. Use of rhBMP-2 in combination with structural cortical allografts: clinical and radiographic outcomes in anterior lumbar spinal surgery. J Bone Joint Surg Am. 2005;87(6):1205–12.

    Article  PubMed  Google Scholar 

  153. Glassman SD et al. RhBMP-2 versus iliac crest bone graft for lumbar spine fusion: a randomized, controlled trial in patients over sixty years of age. Spine (Phila Pa 1976). 2008;33(26):2843–9.

    Article  Google Scholar 

  154. FDA. InFUSE™ Bone Graft/LT-CAGE™ Lumbar Tapered Fusion Devices—P000058. 2002. http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfTopic/pma/pma.cfm?num=P000058.

  155. Boden SD et al. Use of recombinant human bone morphogenetic protein-2 to achieve posterolateral lumbar spine fusion in humans: a prospective, randomized clinical pilot trial: 2002 Volvo Award in clinical studies. Spine (Phila Pa 1976). 2002;27(23):2662–73.

    Article  Google Scholar 

  156. Glassman SD et al. Initial fusion rates with recombinant human bone morphogenetic protein-2/compression resistant matrix and a hydroxyapatite and tricalcium phosphate/collagen carrier in posterolateral spinal fusion. Spine (Phila Pa 1976). 2005;30(15):1694–8.

    Article  Google Scholar 

  157. Dimar JR et al. Clinical outcomes and fusion success at 2 years of single-level instrumented posterolateral fusions with recombinant human bone morphogenetic protein-2/compression resistant matrix versus iliac crest bone graft. Spine (Phila Pa 1976). 2006;31(22):2534–9. discussion 2540.

    Article  Google Scholar 

  158. Mulconrey DS et al. Bone morphogenetic protein (RhBMP-2) as a substitute for iliac crest bone graft in multilevel adult spinal deformity surgery: minimum two-year evaluation of fusion. Spine (Phila Pa 1976). 2008;33(20):2153–9.

    Article  Google Scholar 

  159. Dawson E et al. Recombinant human bone morphogenetic protein-2 on an absorbable collagen sponge with an osteoconductive bulking agent in posterolateral arthrodesis with instrumentation. A prospective randomized trial. J Bone Joint Surg Am. 2009;91(7):1604–13.

    Article  PubMed  Google Scholar 

  160. Friedlaender GE. OP-1 clinical studies. J Bone Joint Surg Am. 2001;83(1 2):S160–1.

    PubMed  Google Scholar 

  161. Friedlaender GE et al. Osteogenic protein-1 (bone morphogenetic protein-7) in the treatment of tibial nonunions. J Bone Joint Surg Am. 2001;83(1 2):S151–8.

    PubMed  Google Scholar 

  162. Geesink RG, Hoefnagels NH, Bulstra SK. Osteogenic activity of OP-1 bone morphogenetic protein (BMP-7) in a human fibular defect. J Bone Joint Surg Br. 1999;81(4):710–8.

    Article  CAS  PubMed  Google Scholar 

  163. Ristiniemi J et al. RhBMP-7 accelerates the healing in distal tibial fractures treated by external fixation. J Bone Joint Surg Br. 2007;89(2):265–72.

    Article  CAS  PubMed  Google Scholar 

  164. Calori GM et al. Application of rhBMP-7 and platelet-rich plasma in the treatment of long bone non-unions: a prospective randomised clinical study on 120 patients. Injury. 2008;39(12):1391–402.

    Article  CAS  PubMed  Google Scholar 

  165. Johnsson R, Stromqvist B, Aspenberg P. Randomized radiostereometric study comparing osteogenic protein-1 (BMP-7) and autograft bone in human noninstrumented posterolateral lumbar fusion: 2002 Volvo Award in clinical studies. Spine (Phila Pa 1976). 2002;27(23):2654–61.

    Article  Google Scholar 

  166. Vaccaro AR et al. A pilot safety and efficacy study of OP-1 putty (rhBMP-7) as an adjunct to iliac crest autograft in posterolateral lumbar fusions. Eur Spine J. 2003;12(5):495–500.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Furlan JC et al. Use of osteogenic protein-1 in patients at high risk for spinal pseudarthrosis: a prospective cohort study assessing safety, health-related quality of life, and radiographic fusion. Invited submission from the Joint Section on Disorders of the Spine and Peripheral Nerves, March 2007. J Neurosurg Spine. 2007;7(5):486–95.

    Article  PubMed  Google Scholar 

  168. Leach J, Bittar RG. BMP-7 (OP-1) safety in anterior cervical fusion surgery. J Clin Neurosci. 2009;16(11):1417–20.

    Article  CAS  PubMed  Google Scholar 

  169. Kanayama M et al. A prospective randomized study of posterolateral lumbar fusion using osteogenic protein-1 (OP-1) versus local autograft with ceramic bone substitute: emphasis of surgical exploration and histologic assessment. Spine (Phila Pa 1976). 2006;31(10):1067–74.

    Article  Google Scholar 

  170. FDA. OP-1 Putty—H020008. 2004. http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cftopic/pma/pma.cfm?num=H020008.

  171. Irokawa D et al. Effect of beta tricalcium phosphate particle size on recombinant human platelet-derived growth factor-BB-induced regeneration of periodontal tissue in dog. Dent Mater J. 2010;29(6):721–30.

    Article  CAS  PubMed  Google Scholar 

  172. FDA. Approval letter for GEM 21S (growth-factor enhanced matrix)—P040013. Silver Spring: FDA; 2005.

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gordana Vunjak-Novakovic.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Spiller, K.L., Vunjak-Novakovic, G. Clinical translation of controlled protein delivery systems for tissue engineering. Drug Deliv. and Transl. Res. 5, 101–115 (2015). https://doi.org/10.1007/s13346-013-0135-1

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13346-013-0135-1

Keywords

Navigation