Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Microenvironmental regulation of biomacromolecular therapies

Key Points

  • Protein-based and nucleotide-based biomacromolecular therapies are being increasingly used in various clinical interventions, but the importance of the cellular microenvironment of the target cells has been not been carefully or broadly examined.

  • In vitro studies demonstrate that the cellular response to protein therapies (for example, growth factors and antibodies) is mediated by aspects of the cellular microenvironment that regulate adhesion and other cell functions, including the composition and structure of the extracellular matrix (ECM).

  • At least three strategies that modulate the cellular microenvironment in vivo may potentially be employed to enhance local protein therapies: regulating cell viability in the target tissue, controlling cell adhesion to the native ECM and providing an artificial microenvironment (for example, synthetic ECM) designed to amplify the desired response. These strategies have been used to create new tissues and to destroy diseased tissues.

  • Several in vitro studies demonstrate that the ability of a cell to take up and express nucleotides is mediated by variables of the cellular microenvironment, including the presentation of cell-adhesion molecules, the architecture and mechanical properties of the ECM, and external mechanical stimulation.

  • Potential strategies to regulate the effectiveness of nucleotide therapies in vivo include the delivery of supplemental signalling factors that modulate cell proliferation, regulation of integrin expression levels, and implantation of nucleotide-releasing engineered ECM. These strategies have been used to mediate tissue regeneration, gene-based vaccination and treatment of chronic and malignant diseases.

  • Careful translation of results from in vitro studies, together with exploitation of recently developed approaches to control nanoscale and microscale ECM signalling, could significantly enhance and broaden the utility of local biomacromolecular therapies.

Abstract

There is currently great interest in molecular therapies to treat various diseases, and this has prompted extensive efforts to achieve target-specific and controlled delivery of bioactive macromolecules (for example, proteins, antibodies, DNA and small interfering RNA) through the design of smart drug carriers. By contrast, the influence of the microenvironment in which the target cell resides and the effect it might have on the success of biomacromolecular therapies has been under-appreciated. The extracellular matrix (ECM) component of the cellular niche may be particularly important, as many diseases and injury disrupt the normal ECM architecture, the cell adhesion to ECM, and the subsequent cellular activities. This Review will discuss the importance of the ECM and the ECM–cell interactions on the cell response to bioactive macromolecules, and suggest how this information could lead to new criteria for the design of novel drug delivery systems.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The cellular microenvironment varies with tissue injury and pathogenesis.
Figure 2: In vitro cell response to protein drugs is modulated by the microenvironment.
Figure 3: Potential strategies to engineer cell microenvironments in vivo to modulate the cellular response to protein drugs.
Figure 4: In vitro efficiency of gene transfer and subsequent level of expression are modulated by properties of the extracellular matrix (ECM).

Similar content being viewed by others

References

  1. Annex, B. H. & Simons, M. Growth factor-induced therapeutic angiogenesis in the heart: protein therapy. Cardiovasc. Res. 65, 649–655 (2005).

    CAS  PubMed  Google Scholar 

  2. Gasparini, G., Longo, R., Toi, M. & Ferrara, N. Angiogenic inhibitors: a new therapeutic strategy in oncology. Nature Clin. Practice Oncol. 2, 562–577 (2005).

    CAS  Google Scholar 

  3. Nakashima, M. & Reddi, A. H. The application of bone morphogenetic proteins to dental tissue engineering. Nature Biotech. 21, 1025–1032 (2003).

    CAS  Google Scholar 

  4. Bickel, U., Yoshikawa, T. & Pardridge, W. M. Delivery of peptides and proteins through the blood–brain barrier. Adv. Drug Deliv. Rev. 46, 247–279 (2001).

    CAS  PubMed  Google Scholar 

  5. Morris, M. C., Depollier, J., Mery, J., Heitz, F. & Divita, G. A peptide carrier for the delivery of biologically active proteins into mammalian cells. Nature Biotech. 19, 1173–1176 (2001).

    CAS  Google Scholar 

  6. Isner, J. M. Arterial gene transfer of naked DNA for therapeutic angiogenesis: early clinical results. Adv. Drug Deliv. Rev. 30, 185–197 (1998).

    CAS  PubMed  Google Scholar 

  7. Behlke, M. A. Progress towards in vivo use of siRNAs. Mol. Ther. 13, 644–670 (2006).

    CAS  PubMed  Google Scholar 

  8. Gleave, M. E. & Monia, B. P. Antisense therapy for cancer. Nature Rev. Cancer 5, 468–479 (2005).

    CAS  Google Scholar 

  9. Gerngross, T. U. Advances in the production of human therapeutic proteins in yeasts and filamentous fungi. Nature Biotech. 22, 1409–1414 (2004).

    CAS  Google Scholar 

  10. Roque, A. C. A., Lowe, C. R. & Taipa, M. A. Antibodies and genetically engineered related molecules: production and purification. Biotechnol. Prog. 20, 639–654 (2004).

    CAS  PubMed  Google Scholar 

  11. Pavlou, A. K. & Reichert, J. M. Recombinant protein therapeutics — success rates, market trends and values to 2010. Nature Biotech. 22, 1513–1519 (2004).

    CAS  Google Scholar 

  12. Tuszynski, M. H. et al. A Phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nature. Med. 11, 551–555 (2005).

    CAS  PubMed  Google Scholar 

  13. Isner, J. M. Myocardial gene therapy. Nature 415, 234–239 (2002).

    CAS  PubMed  Google Scholar 

  14. Mannucci, P. M. & Tuddenham, E. G. D. The hemophilias — from royal genes to gene therapy. N. Eng. J. Med. 344, 1773–1779 (2001).

    CAS  Google Scholar 

  15. Robinson, B. W. S. & Lake, R. A. Advances in malignant mesothelioma. N. Eng. J. Med. 353, 1591–1603 (2005).

    CAS  Google Scholar 

  16. Talmadge, J. E. The pharmaceutics and delivery of therapeutic polypeptides and proteins. Adv. Drug Deliv. Rev. 10, 247–299 (1993).

    CAS  Google Scholar 

  17. Krejsa, C., Rogge, M. & Sadee, W. Protein therapeutics: new applications for pharmacogenetics. Nature Rev. Drug Discov. 5, 507–521 (2006).

    CAS  Google Scholar 

  18. Ng, E. W. M. et al. Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular disease. Nature Rev. Drug Discov. 5, 123–132 (2006).

    CAS  Google Scholar 

  19. Pack, D. W., Hoffman, A. S., Pun, S. & Stayton, P. S. Design and development of polymers for gene delivery. Nature Rev. Drug Discov. 4, 581–593 (2005).

    CAS  Google Scholar 

  20. Schwarze, S. R., Ho, A., Vocero-Akbani, A. & Dowdy, S. F. In vivo protein transduction: delivery of a biologically active protein into the mouse. Science 285, 1569–1572 (1999).

    CAS  PubMed  Google Scholar 

  21. Mastrobattista, E., van der Aa, M., Hennink, W. E. & Crommelin, D. J. A. Artificial viruses: a nanotechnological approach to gene delivery. Nature Rev. Drug Discov. 5, 115–121 (2006).

    Google Scholar 

  22. Mrsny, R. J. Strategies for targeting protein therapeutics to selected tissues and cells. Expert Opin. Biol. Ther. 4, 65–73 (2004).

    CAS  PubMed  Google Scholar 

  23. Theys, J. et al. Specific targeting of cytosine deaminase to solid tumors by engineered Clostridium acetobutylicum. Cancer Gene Ther. 8, 294–297 (2001).

    CAS  PubMed  Google Scholar 

  24. Luo, D. & Saltzman, W. M. Synthetic DNA delivery systems. Nature Biotech. 18, 33–37 (2000).

    CAS  Google Scholar 

  25. Park, T. G., Jeong, J. H. & Kim, S. W. Current status of polymeric gene delivery systems. Adv. Drug Deliv. Rev. 58, 467–486 (2006).

    CAS  PubMed  Google Scholar 

  26. Storrie, H., Mooney, D. J. Sustained delivery of plasmid DNA from polymeric scaffolds for tissue engineering. Adv. Drug Deliv. Rev. 58, 500–514 (2006).

    CAS  PubMed  Google Scholar 

  27. Lutolf, M. P. & Hubbell, J. A. Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering. Nature Biotech. 23, 47–55 (2005).

    CAS  Google Scholar 

  28. Chen, R. R. & Mooney, D. J. Polymeric growth factor delivery strategies for tissue engineering. Pharm. Res. 20, 1103–1112 (2003).

    CAS  PubMed  Google Scholar 

  29. Putney, S. D. & Burke, P. A. Improving protein therapeutics with sustained-release formulations. Nature Biotech. 16, 153–157 (1998).

    CAS  Google Scholar 

  30. Langer, R. Drug delivery and targeting. Nature 392, 5–10 (1998).

    CAS  PubMed  Google Scholar 

  31. Steeg, P. S. Tumor metastasis: mechanistic insights and clinical challenges. Nature Med. 12, 895–904 (2006).

    CAS  PubMed  Google Scholar 

  32. Mellor, H. R. et al. Optimising non-viral gene delivery in a tumour spheroid model. J. Gene Med. 8, 1160–1170 (2006).

    CAS  PubMed  Google Scholar 

  33. Jain, R. K. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307, 58–62 (2005).

    CAS  PubMed  Google Scholar 

  34. Harris, S. S. & Giorgio, T. D. Convective flow increases lipoplex delivery rate to in vitro cellular monolayers. Gene Ther. 12, 512–520 (2005).

    CAS  PubMed  Google Scholar 

  35. Bhadriraju, K. & Chen, C. S. Engineering cellular microenvironments to cell-based drug testing. Drug Discov. Today 7, 612–620 (2002).

    CAS  PubMed  Google Scholar 

  36. Paszek, M. J. et al. Tensional homeostasis and the malignant phenotype. Cancer Cell 8, 241–254 (2005).

    CAS  PubMed  Google Scholar 

  37. Griffith, L. G. & Swartz, M. A. Capturing complex 3D tissue physiology in vitro. Nature Rev. Mol. Cell Biol. 7, 211–224 (2006).

    CAS  Google Scholar 

  38. Discher, D. E., Janmey, P. & Wang, Y. L. Tissue cells feel and respond to the stiffness of their substrate. Science 310, 1139–1143 (2005).

    CAS  PubMed  Google Scholar 

  39. Sakai, T., Larsen, M. & Yamada, K. M. Fibronectin requirement in branching morphogenesis. Nature 423, 876–881 (2003).

    CAS  PubMed  Google Scholar 

  40. Geiger, B., Bershadsky, A., Pankov, R. & Yamada, K. M. Transmembrane extracellular matrix–cytoskeleton crosstalk. Nature Rev. Mol. Cell Biol. 2, 793–805 (2001).

    CAS  Google Scholar 

  41. Giancotti, F. G. & Ruoslahti, E. Integrin signaling. Science 285, 1028–1032 (1999).

    CAS  PubMed  Google Scholar 

  42. Ingber, D. E. & Folkman, J. How does extracellular matrix control capillary morphogenesis? Cell 58, 803–805 (1989).

    CAS  PubMed  Google Scholar 

  43. Kumar, V. A., Abbas, A. & Fausto, N. Robbins & Cotran Pathologic Basis of Disease 7th edn (Elsevier Saunders, Philadelphia, 2005).

    Google Scholar 

  44. Ingber, D. E. Mechanobiology and diseases of mechanotransduction. Ann. Med. 35, 564–577 (2003).

    PubMed  Google Scholar 

  45. Urban J. P., Smith, S. & Fairbank J. C. Nutrition of the intervertebral disc. Spine 29, 2700–2709 (2004).

    PubMed  Google Scholar 

  46. Kovacs, E. J. & Dipietro, L. A. Fibrogenic cytokines and connective-tissue production. FASEB J. 8, 854–861 (1994).

    CAS  PubMed  Google Scholar 

  47. Maghazachi, A. A. & Al-Aoukaty, A. Chemokines activate natural killer cells through heterotrimeric G-proteins: implications for the treatment of AIDS and cancer. FASEB J. 12, 913–924 (1998).

    CAS  PubMed  Google Scholar 

  48. Stadelmann, W. K., Digenis, A. G. & Tobin, G. R. Impediments to wound healing. Am. J. Surg. 176, S39–S47 (1998).

    Google Scholar 

  49. Vincent, T. & Mechti, N. Extracellular matrix in bone marrow can mediate drug resistance in myeloma. Leuk. Lymphoma 46, 803–811 (2005).

    CAS  PubMed  Google Scholar 

  50. Taichman, R. S. Blood and bone: two tissues whose fates are intertwined to create the hematopoietic stem-cell niche. Blood 105, 2631–2639 (2005).

    CAS  PubMed  Google Scholar 

  51. Calvi, L. M. et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 425, 841–846 (2003).

    CAS  PubMed  Google Scholar 

  52. Yang, A. D. et al. Improving delivery of antineoplastic agents with anti-vascular endothelial growth factor therapy. Cancer 103, 1561–1570 (2005). This paper reports that a synergistic improvement in the efficiency of protein therapies can be achieved by addition of supplemental growth factors.

    CAS  PubMed  Google Scholar 

  53. Richardson, T. P., Peters, M. C., Ennett, A. B. & Mooney, D. J. Polymeric system for dual growth factor delivery. Nature Biotech. 19, 1029–1034 (2001).

    CAS  Google Scholar 

  54. Woodward, T. L., Xie, J. W. & Haslam, S. Z. The role of mammary stroma in modulating the proliferative response to ovarian hormones in the normal mammary gland. J. Mammary Gland Biol. Neoplasia 3, 117–131 (1998).

    CAS  PubMed  Google Scholar 

  55. Thannickal, V. J. et al. Myofibroblast differentiation by transforming growth factor-β1 is dependent on cell adhesion and integrin signaling via focal adhesion kinase. J. Biol. Chem. 278, 12384–12389 (2003). This report discusess the importance of cell adhesion in mediating the efficiency of growth factors, which stimulate cell differentiation.

    CAS  PubMed  Google Scholar 

  56. Woodward, T. L., Xie, J. W., Fendrick, J. L. & Haslam, S. Z. Proliferation of mouse mammary epithelial cells in vitro: interactions among epidermal growth factor, insulin-like growth factor I, ovarian hormones, and extracellular matrix proteins. Endocrinology 141, 3578–3586 (2000).

    CAS  PubMed  Google Scholar 

  57. Pearson, R. G. et al. Spatial confinement of neurite regrowth from dorsal root ganglia within nonporous microconduits. Tissue Eng. 9, 201–208 (2003).

    CAS  PubMed  Google Scholar 

  58. Korff, T. & Augustin, H. G. Tensional forces in fibrillar extracellular matrices control directional capillary sprouting. J. Cell Sci. 112, 3249–3258 (1999).

    CAS  PubMed  Google Scholar 

  59. Maheshwari, G., Wells, A., Griffith, L. G. & Lauffenburger, D. A. Biophysical integration of effects of epidermal growth factor and fibronectin on fibroblast migration. Biophys. J. 76, 2814–2823 (1999). A study demonstrating that the efficiency of growth factors that stimulate cell migration is modulated by the extent of cell adhesion.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Maheshwari, G., Brown, G., Lauffenburger, D. A., Wells, A. & Griffith, L. G. Cell adhesion and motility depend on nanoscale RGD clustering. J. Cell Sci. 113, 1677–1686 (2000).

    CAS  PubMed  Google Scholar 

  61. Alobaid, N. et al. Nanocomposite containing bioactive peptides promote endothelialisation by circulating progenitor cells: an in vitro evaluation. Eur. J. Vasc. Surg. 32, 76–83 (2006).

    CAS  Google Scholar 

  62. Addison, C. L. et al. The response of VEGF-stimulated endothelial cells to angiostatic molecules is substrate-dependent. BMC Cell Biol. 6, 38 (2005).

    PubMed  PubMed Central  Google Scholar 

  63. Sutton, A. B., Canfield, A. E., Schor, S. L., Grant, M. E. & Schor, A. M. The response of endothelial-cells to TGF-β-1 is dependent upon cell-shape, proliferative state and the nature of the substratum. J. Cell Sci. 99, 777–787 (1991).

    CAS  PubMed  Google Scholar 

  64. Dye, J. F. et al. Distinct patterns of microvascular endothelial cell morphology are determined by extracellular matrix composition. Endothelium 11, 151–167 (2004).

    CAS  PubMed  Google Scholar 

  65. Chen, C. S., Mrkisich, M., Huang, S., Whitesides, G. M. & Ingber, D. E. Geometric control of cell life and death. Science 276, 1425–1428 (1997).

    CAS  PubMed  Google Scholar 

  66. Lee, K. Y. et al. Nanoscale adhesion ligand organization regulates osteoblast proliferation and differentiation. Nano Lett. 4, 1501–1506 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Kong, H. J., Polte, T., Alsberg, E. & Mooney, D. J. FRET measurements of cell-traction forces and nano-scale clustering of adhesion ligands varied by substrate stiffness. Proc. Natl Acad. Sci. USA 102, 4300–4305 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Ito, Y. et al. Differential control of cellular gene expression by diffusible and non-diffusible EGF. J. Biochem. 129, 733–737 (2001).

    CAS  PubMed  Google Scholar 

  69. Ehrbar, M., Metters, A., Zammaretti, P., Hubbell, J. A. & Zisch, A. H. Endothelial cell proliferation and progenitor maturation by fibrin-bound VEGF variants with differential susceptibilities to local cellular activity. J. Control. Release 101, 93–109 (2005).

    CAS  PubMed  Google Scholar 

  70. Chabut, D. et al. Low molecular weight fucoidan and heparin enhance the basic fibroblast growth factor-induced tube formation of endothelial cells through heparan sulfate-dependent α6 overexpression. Mol. Pharmacol. 64, 696–702 (2003).

    CAS  PubMed  Google Scholar 

  71. Wijelath, E. S. et al. Novel vascular endothelial growth factor binding domains of fibronectin enhance vascular endothelial growth factor biological activity. Circ. Res. 91, 25–31 (2002).

    CAS  PubMed  Google Scholar 

  72. Elicerini, B. P. & Cheresh, D. A. The role of av integrins during angiogenesis: insights into potential mechanisms of action and clinical development. J. Clin. Invest. 103, 1227–1229 (1999).

    Google Scholar 

  73. Brooks, P. C., Clark, R. A. F. & Cheresh, D. A. Requirement of vascular integrin α(v)β(3) for angiogenesis. Science 264, 569–571 (1994). A study showing that cellular integrin expression levels modulate the efficiency of angiogenesis-promoting growth factors.

    CAS  PubMed  Google Scholar 

  74. Nakatsu, M. N. et al. Angiogenic sprouting and capillary lumen formation modeled by human umbilical vein endothelial cells (HUVEC) in fibrin gels: the role of fibroblasts and angiopoietin-1. Microvasc. Res. 66, 102–112 (2003).

    CAS  PubMed  Google Scholar 

  75. Koblizek, T. I., Weiss, C., Yancopoulos, G. D., Deutsch, U. & Risau, W. Angiopoietin-1 induces sprouting angiogenesis in vitro. Curr. Biol. 8, 529–532 (1998).

    CAS  PubMed  Google Scholar 

  76. Weaver, V. M. et al. β4 Integrin-dependent formation of polarized three-dimensional architecture confers resistance to apoptosis in normal and malignant mammary epithelium. Cancer Cell 2, 205–216 (2002). A resport on cells that are cultured in a 3D microenvironment respond to therapeutic proteins in a distinct manner compared with cells cultured on 2D substrates.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. dit Faute, M. A. et al. Distinctive alterations of invasiveness, drug resistance and cell–cell organization in 3D-cultures of MCF-7, a human breast cancer cell line, and its multidrug resistant variant. Clin. Exp. Metastasis 19, 161–167 (2002).

    PubMed  Google Scholar 

  78. Wang, F. et al. Reciprocal interactions between β1-integrin and epidermal growth factor receptor in three-dimensional basement membrane breast cultures: a different perspective in epithelial biology. Proc. Natl Acad. Sci. USA 95, 14821–14826 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Minchinton, A. I. & Tannock, I. F. Drug penetration in solid tumors. Nature Rev. Cancer 6, 583–592 (2006).

    CAS  Google Scholar 

  80. Herodin, F., Bourin, P., Mayol, J. F., Lataillade, J. J. & Drouet, M. Short-term injection of antiapoptotic cytokine combinations soon after lethal γ-irradiation promotes survival. Blood 101, 2609–2616 (2003). A paper reporting that the cellular microenvironment can mediate the resistance of cells to apoptosis induced by irradiation.

    CAS  PubMed  Google Scholar 

  81. Bender, J. G., Cooney, E. M., Kandel, J. J. & Yamashiro, D. J. Vascular remodeling and clinical resistance to antiangiogenic cancer therapy. Drug Resist. Updat. 7, 289–300 (2004).

    Google Scholar 

  82. Jo, N. et al. Inhibition of platelet-derived growth factor B signaling enhances the efficacy of anti-vascular enclothelial growth factor therapy in multiple models of ocular neovascularization. Am. J. Pathol. 168, 2036–2053 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Lee, J. Y. et al. Enhanced bone formation by controlled growth factor delivery from chitosan-based biomaterials. J. Control. Release 78, 187–197 (2002).

    CAS  PubMed  Google Scholar 

  84. Boontheekul, T. & Mooney, D. J. Protein-based signaling systems in tissue engineering. Curr. Opin. Biotechnol. 14, 559–565 (2003).

    CAS  PubMed  Google Scholar 

  85. Schmoekel, H. G. et al. Bone repair with a form of BMP-2 engineered for incorporation into fibrin cell ingrowth matrices. Biotechnol. Bioeng. 89, 253–262 (2005).

    CAS  PubMed  Google Scholar 

  86. Zisch, A. H. et al. Cell-demanded release of VEGF from synthetic, biointeractive cell-ingrowth matrices for vascularized tissue growth. FASEB J. 17, 2260–2262 (2003).

    CAS  PubMed  Google Scholar 

  87. Lutolf, M. R. et al. Repair of bone defects using synthetic mimetics of collagenous extracellular matrices. Nature Biotech. 21, 513–518 (2003). A study demonstrating that growth-factor release from synthetic ECM can be regulated by cell-mediated degradation of the ECM.

    CAS  Google Scholar 

  88. Murphy, W. L., Peters, M. C., Kohn, D. H. & Mooney, D. J. Sustained release of vascular endothelial growth factor from mineralized poly(lactide-co-glycolide) scaffolds for tissue engineering. Biomaterials 21, 2521–2527 (2000).

    CAS  PubMed  Google Scholar 

  89. Leach, J. K., Kaigler, D., Wang, Z., Krebsbach, P. H. & Mooney, D. J. Coating of VEGF-releasing scaffolds with bioactive glass for angiogenesis and bone regeneration. Biomaterials 27, 3249–3255 (2006).

    CAS  PubMed  Google Scholar 

  90. Gwak, S. J. et al. Synergistic effect of keratinocyte transplantation and epidermal growth factor delivery on epidermal regeneration. Cell Transplant. 14, 809–817 (2005).

    PubMed  Google Scholar 

  91. Davis, M. E. et al. Local myocardial insulin-like growth factor 1 (IGF-1) delivery with biotinylated peptide nanofibers improves cell therapy for myocardial infarction. Proc. Natl Acad. Sci. USA 103, 8155–8160 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Park, M. S., Kim, S. S., Cho, S. W., Choi, C. Y. & Kim, B. S. Enhancement of the osteogenic efficacy of osteoblast transplantation by the sustained delivery of basic fibroblast growth factor. J. Biomed. Mater. Res. 79, 353–359 (2006).

    Google Scholar 

  93. Simmons, C. A., Alsberg, E., Hsiong, S., Kim, W. J. & Mooney, D. J. Dual growth factor delivery and controlled scaffold degradation enhance in vivo bone formation by transplanted bone marrow stromal cells. Bone 35, 562–569 (2004).

    CAS  PubMed  Google Scholar 

  94. Um, S. H. et al. Enzyme-catalysed assembly of DNA hydrogel. Nature Mater. 5, 797–801 (2006).

    CAS  Google Scholar 

  95. Gates, B. D. et al. New approaches to nanofabrication: molding, printing, and other techniques. Chem. Rev. 105, 1171–1196 (2005).

    CAS  PubMed  Google Scholar 

  96. Sia, S. K. & Whitesides, G. M. Microfluidic devices fabricated in poly(dimethylsiloxane) for biological studies. Electrophoresis 24, 3563–3576 (2003).

    CAS  PubMed  Google Scholar 

  97. Hanania, E. G. et al. Recent advances in the application of gene-therapy to human disease. Am. J. Med. 99, 537–552 (1995).

    CAS  PubMed  Google Scholar 

  98. Tuszynski, M. H. & Blesch, A. Nerve growth factor: from animal models of cholinergic neuronal degeneration to gene therapy in Alzheimer's disease. Prog. Brain Res. 146, 441–449 (2004).

    CAS  PubMed  Google Scholar 

  99. Yu, M., Poeschla, E. & Wongstaal, F. Progress towards gene therapy for HIV infection. Gene Ther. 1, 13–26 (1994).

    CAS  PubMed  Google Scholar 

  100. Shi, F. S., Weber, S., Gan, J., Rakhmilevich, A. L. & Mahvi, D. M. Granulocyte-macrophage colony-stimulating factor (GM-CSF) secreted by cDNA-transfected tumor cells induces a more potent antitumor response than exogenous GM-CSF. Cancer Gene Ther. 6, 81–88 (1999).

    CAS  PubMed  Google Scholar 

  101. Yamamoto, M. & Tabata, Y. Tissue engineering by modulated gene delivery. Adv. Drug Deliv. Rev. 58, 535–554 (2006).

    CAS  PubMed  Google Scholar 

  102. Ikeda, Y. & Taira, K. Ligand-targeted delivery of therapeutic siRNA. Pharm. Res. 23, 1631–1640 (2006).

    CAS  PubMed  Google Scholar 

  103. Kasid, U. & Dritschilo, A. RAF antisense oligonucleotide as a tumor radiosensitizer. Oncogene 22, 5876–5884 (2003).

    CAS  PubMed  Google Scholar 

  104. Ledley, F. D. Nonviral gene therapy: the promise of genes as pharmaceutical products. Hum. Gene Ther. 6, 1129–1144 (1995).

    CAS  PubMed  Google Scholar 

  105. Ge, Q. et al. Inhibition of influenza virus production in virus-infected mice by RNA interference. Proc. Natl Acad. Sci. USA 101, 8676–8681 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Lawrie, A. et al. Ultrasound enhances reporter gene expression after transfection of vascular cells in vitro. Circulation 99, 2617–2620 (1999). A study demonstrating that external physical stimulation can regulate the efficiency of gene delivery.

    CAS  PubMed  Google Scholar 

  107. Satkauskas, S. et al. Mechanisms of in vivo DNA electrotransfer: respective contributions of cell electropermeabilization and DNA electrophoresis. Mol. Ther. 5, 133–140 (2002).

    CAS  PubMed  Google Scholar 

  108. Escriou, V., Carriere, M., Bussone, F., Wils, P. & Scherman, D. Critical assessment of the nuclear import of plasmid during cationic lipid-mediated gene transfer. J. Gene Med. 3, 179–187 (2001).

    CAS  PubMed  Google Scholar 

  109. Tseng, W. C., Haselton, F. R. & Giorgio, T. D. Mitosis enhances transgene expression of plasmid delivered by cationic liposomes. Biochim. Biophys. Acta 1445, 53–64 (1999).

    CAS  PubMed  Google Scholar 

  110. Hanenberg, H. et al. Colocalization of retrovirus and target cells on specific fibronectin fragments increases genetic transduction of mammalian cells. Nature Med. 2, 876–882 (1996).

    CAS  PubMed  Google Scholar 

  111. Dao, M. A., Hashino, K., Kato, I. & Nolta, J. A. Adhesion to fibronectin maintains regenerative capacity during ex vivo culture and transduction of human hematopoietic stem and progenitor cells. Blood 92, 4612–4621 (1998).

    CAS  PubMed  Google Scholar 

  112. MacNeill, E. C. et al. Simultaneous infection with retroviruses pseudotyped with different envelope proteins bypasses viral receptor interference associated with colocalization of gp70 and target cells on fibronectin CH-296. J. Virol. 73, 3960–3967 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Goerner, M. et al. The use of granulocyte colony-stimulating factor during retroviral transduction on fibronectin fragment CH-296 enhances gene transfer into hematopoietic repopulating cells in dogs. Blood 94, 2287–2292 (1999).

    CAS  PubMed  Google Scholar 

  114. Kong, H. J., Hsiong, S. & Mooney, D. J. Nanoscale cell adhesion ligands presentation regulates non-viral gene delivery and expression. Nano Lett. 7, 161–166 (2007).

    CAS  PubMed  Google Scholar 

  115. Keselowsky, B. G., Collard, D. M. & Garcia, A. J. Surface chemistry modulates fibronectin conformation and directs integrin binding and specificity to control cell adhesion. J. Biomed. Mater. Res. A. 66, 247–259 (2003).

    PubMed  Google Scholar 

  116. Shen, H., Tan, J. & Saltzman, W. M. Surface-mediated gene transfer from nanocomposites of controlled texture. Nature Mater. 3, 569–574 (2004).

    CAS  Google Scholar 

  117. Kong, H. J. et al. Non-viral gene delivery regulated by stiffness of cell adhesion substrates. Nature Mater. 4, 460–464 (2005). A report demonstrating that the physical properties of a cell-adhesion matrix regulate the efficiency of non-viral gene uptake and expression.

    CAS  Google Scholar 

  118. Hosseinkhani, H. et al. Combination of 3D tissue engineered scaffold and non-viral gene carrier enhance in vitro DNA expression of mesenchymal stem cells. Biomaterials 27, 4269–4278 (2006).

    CAS  PubMed  Google Scholar 

  119. Tidball, J. G. Mechanical signal transduction in skeletal muscle growth and adaptation. J. Appl. Physiol. 98, 1900–1908 (2005).

    CAS  PubMed  Google Scholar 

  120. Huang, H. D., Kamm, R. D. & Lee, R. T. Cell mechanics and mechanotransduction: pathways, probes, and physiology. Am. J. Physiol. 287, C1–C11 (2004).

    CAS  Google Scholar 

  121. Brown, T. D. Techniques for mechanical stimulation of cells in vitro: a review. J. Biomech. 33, 3–14 (2000).

    CAS  PubMed  Google Scholar 

  122. Griese, D. P. et al. Vascular gene delivery of anticoagulants by transplantation of retrovirally-transduced endothelial progenitor cells. Cardiovasc. Res. 58, 469–477 (2003).

    CAS  PubMed  Google Scholar 

  123. Grove, J. E. et al. Marrow-derived cells as vehicles for delivery of gene therapy to pulmonary epithelium. Am. J. Respir. Cell Mol. Biol. 27, 645–651 (2002).

    CAS  PubMed  Google Scholar 

  124. Cheung, A. T. et al. Glucose-dependent insulin release from genetically engineered K cells. Science 290, 1959–1962 (2000).

    CAS  PubMed  Google Scholar 

  125. Lu, Y. X. et al. Recombinant vascular endothelial growth factor secreted from tissue-engineered bioartificial muscles promotes localized angiogenesis. Circulation 104, 594–599 (2001).

    CAS  PubMed  Google Scholar 

  126. Shansky, J., Creswick, B., Lee, P., Wang, X. & Vandenburgh, H. Paracrine release of insulin-like growth factor 1 from a bioengineered tissue stimulates skeletal muscle growth in vitro. Tissue Eng. 12, 1833–1841 (2006).

    CAS  PubMed  Google Scholar 

  127. Cartier, R. & Reszka, R. Utilization of synthetic peptides containing nuclear localization signals for nonviral gene transfer systems. Gene Ther. 9, 157–167 (2002).

    CAS  PubMed  Google Scholar 

  128. Andre, F. M., Cournil-Henrionnet, C., Vernerey, D., Opolon, P. & Mir, L. M. Variability of naked DNA expression after direct local injection: the influence of the injection speed. Gene Ther. 13, 1619–1627 (2006).

    CAS  PubMed  Google Scholar 

  129. Riddle, K. W. et al. Modifying the proliferative state of target cells to control DNA expression and identifying cell types transfected in vivo. Mol. Ther. 15, 361–368 (2006).

    Google Scholar 

  130. Plopper, G. E., McNamee, H. P., Dike, L. E., Bojanowski, K. & Ingber, D. E. Convergence of integrin and growth-factor receptor signaling pathways within the focal adhesion complex. Mol. Biol. Cell 6, 1349–1365 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Nayak, B. P., Sailaja, G. & Jabbar, A. M. Augmenting the immunogenecity of DNA vaccines: role of plasmid-encoded Flt-3 ligand. as a molecular adjuvant in genetic vaccination. Virology 348, 277–288 (2006).

    CAS  PubMed  Google Scholar 

  132. Li, J. M., Fan, L. M., Shah, A. & Brooks, G. Targeting αvβ3 and α5β1 for gene delivery to proliferating VSMCs: synergistic effect of TGF-β1. Am. J. Physiol. 285, H1123–H1131 (2003).

    CAS  Google Scholar 

  133. Shea, L. D., Smiley, E., Bonadio, J. & Mooney, D. J. DNA delivery from polymer matrices for tissue engineering. Nature Biotech. 17, 551–554 (1999).

    CAS  Google Scholar 

  134. Bonadio, J., Smiley, E., Patil, P. & Goldstein, S. Localized, direct plasmid gene delivery in vivo: prolonged therapy results in reproducible tissue regeneration. Nature Med. 5, 753–759 (1999). This paper suggests that tissue regeneration can be promoted by the sustained release of plasmid DNA from a synthetic matrix to inwardly migrating host cells.

    CAS  PubMed  Google Scholar 

  135. Huang, Y. C., Kaigler, D., Rice, K. G., Krebsbach, P. H. & Mooney, D. J. Combined angiogenic and osteogenic factor delivery enhances bone marrow stromal cell-driven bone regeneration. J. Bone Miner. Res. 20, 848–857 (2005).

    CAS  PubMed  Google Scholar 

  136. Endo, M. et al. Bone regeneration by modified gene-activated matrix: effectiveness in segmental tibial defects in rats. Tissue Eng. 12, 489–497 (2006).

    CAS  PubMed  Google Scholar 

  137. Joki, T. et al. Continuous release of endostatin from microencapsulated engineered cells for tumor therapy. Nature Biotech. 19, 35–39 (2001).

    CAS  Google Scholar 

  138. Hill, E., Boontheekul, T. & Mooney, D. J. Regulating activation of transplanted cells controls tissue regeneration. Proc. Natl Acad. Sci. USA 103, 2494–2499 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Khademhosseini, A., Langer, R., Borenstein, J. & Vacanti, J. P. Microscale technologies for tissue engineering and biology. Proc. Natl Acad. Sci. USA 103, 2480–2487 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Langer, R. & Tirrell, D. A. Designing materials for biology and medicine. Nature 428, 487–492 (2004).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank the National Institutes of Health (R37 DE013033, R01 DE13349 and R01 HL069957) and the US Army Research Laboratories and Research Office (DAAD-19-03-1-0168) for financial support.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to David J. Mooney.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

David J. Mooney's homepage

Human Genome Project

Hyun Joon Kong's homepage

Glossary

Biomacromolecule

A high molecular mass molecule formed from biological building blocks such as nucleic acids, amino acids and sugars.

Plasmid

A small circular piece of DNA that can replicate itself independently of chromosomal DNA; frequently used to introduce potentially therapeutic genes into cells.

Extracellular matrix

(ECM). A complex mixture of non-living material surrounding cells within tissues. The ECM provides space for tissue formation, mechanically supports the tissue and provides cues to regulate cell function.

Integrin

A specific type of cell-surface receptor that mediates adhesion to the extracellular matrix and subsequent signalling to the cell.

Silencing RNA

20–25 nucleotide-long double-stranded RNA molecules, referred to as small interfering RNA (siRNA), used to interfere with the expression of a specific gene. siRNA treatments are also termed interference therapies.

Aptamer

Short nucleic-acid- or amino-acid-based molecules that bind a specific target molecule. They are often used as macromolecular drugs.

Matrix metalloproteinase

(MMP). A specific type of enzyme that is capable of degrading extracellular-matrix proteins.

Antisense therapy

A treatment that uses a single strand of nucleic acid (antisense oligonucleotide) that is designed to bind a specific mRNA and inactivate its expression.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kong, H., Mooney, D. Microenvironmental regulation of biomacromolecular therapies. Nat Rev Drug Discov 6, 455–463 (2007). https://doi.org/10.1038/nrd2309

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd2309

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing