We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Therapeutic delivery of nucleic acids for skin wound healing

    Rajalekshmy Girishkumar Padmakumari

    Biosurface Technology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India

    ,
    Caroline Diana Sherly

    Biosurface Technology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India

    &
    Rekha Mannemcherril Ramesan

    *Author for correspondence: Tel.: +91 471 252 0214;

    E-mail Address: rekhamr@sctimst.ac.in

    Biosurface Technology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India

    Published Online:https://doi.org/10.4155/tde-2022-0003

    Though wound care has advanced, treating chronic wounds remains a challenge and there are many clinical issues that must be addressed. Gene therapy is a recent approach to treating chronic wounds that remains in its developmental stage. The limited reports available describe the therapeutic applications of various forms of nucleic acid delivery for treating chronic wounds, including DNA, mRNA, siRNA, miRNA and so on. Though these bioactive molecules represent great therapeutic potential, sustaining their bioactivity in the wound bed is a challenge. To overcome this hurdle, delivery systems are also being widely investigated. In this review, nucleic acid-based therapy and its delivery for treating chronic wounds is discussed in detail.

    Graphical abstract

    References

    • 1. Tobin DJ. Biochemistry of human skin – our brain on the outside. Chem. Soc. Rev. 35(1), 52–67 (2006).
    • 2. Kanitakis Jean. Anatomy, histology and immunohistochemistry of normal human skin. Eur. J. Dermatol. 12(4), 390–401 (2002).
    • 3. Banerjee J, Sen CK. MicroRNAs in skin and wound healing. Methods Mol. Biol. 936, 343–356 (2013).
    • 4. Smyth Templeton N. Gene and Cell Therapy: Therapeutic Mechanisms and Strategies. (4th Edition). CRC Press (2015).
    • 5. Mulholland EJ, Dunne N, McCarthy HO. MicroRNA as therapeutic targets for chronic wound healing. Mol. Ther. Nucleic Acids 8, 46–55 (2017).
    • 6. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature 453(7193), 314–321 (2008).
    • 7. Reinke JM, Sorg H. Wound repair and regeneration. ESR 49(1), 35–43 (2012).
    • 8. Berger AG, Chou JJ, Hammond PT. Approaches to modulate the chronic wound environment using localized nucleic acid delivery. Adv. Wound Care (2020).
    • 9. Schultz GS, Chin GA, Moldawer L, Diegelmann RF. Principles of Wound Healing. University of Adelaide Press, Adelaide, Australia (2011).
    • 10. Gailit J, Clark RAF. Wound repair in the context of extracellular matrix. Curr. Opin. Cell Biol. 6(5), 717–725 (1994).
    • 11. Bennett NT, Schultz GS. Growth factors and wound healing: part II. Role in normal and chronic wound healing. Am. J. Surg. 166(1), 74–81 (1993).
    • 12. Bennett NT, Schultz GS. Growth factors and wound healing: biochemical properties of growth factors and their receptors. Am. J. Surg. 165(6), 728–737 (1993).
    • 13. Rumalla VK, Borah GL. Cytokines, growth factors, and plastic surgery. Plast. Reconstr. Surg. 108(3), 719–733 (2001).
    • 14. Meszaros AJ, Reichner JS, Albina JE. Macrophage-induced neutrophil apoptosis. J. Immunol. 165(1), 435–441 (2000).
    • 15. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol. 8(12), 958–969 (2008).
    • 16. Mustoe TA, O'Shaughnessy K, Kloeters O. Chronic wound pathogenesis and current treatment strategies: a unifying hypothesis. Plast. Reconstr. Surg. 117(Suppl. 7), S35–S41 (2006).
    • 17. El-Sherbiny IM, Yacoub MH. Hydrogel scaffolds for tissue engineering: progress and challenges. Glob. Cardiol. Sci. Pract. 2013(3), 38 (2013).
    • 18. Lipsky BA, Hoey C. Topical antimicrobial therapy for treating chronic wounds. Clin. Infect. Dis. 49(10), 1541–1549 (2009).
    • 19. Draelos ZD, Rizer RL, Trookman NS. A comparison of postprocedural wound care treatments: do antibiotic-based ointments improve outcomes? J. Am. Acad. Dermatol. 64(Suppl. 3), S23–S29 (2011).
    • 20. Newton DJ, Khan F, Belch JJF, Mitchell MR, Leese GP. Blood flow changes in diabetic foot ulcers treated with dermal replacement therapy. J. Foot Ankle Surg. 41(4), 233–237 (2002).
    • 21. Kirby J. Hyperbaric oxygen therapy and negative pressure as advanced wound management. Mol. Med. 116(3), 192–194 (2019).
    • 22. Olsson M, Järbrink K, Divakar U et al. The humanistic and economic burden of chronic wounds: a systematic review. Wound Repair Regen. 27(1), 114–125 (2019).
    • 23. Deonarine K, Panelli MC, Stashower ME et al. Gene expression profiling of cutaneous wound healing. J. Transl. Med. 5(1), 11 (2007).
    • 24. Berger AG, Chou JJ, Hammond PT. Approaches to modulate the chronic wound environment using localized nucleic acid delivery. Adv. Wound Care 10, 503–528 (2020).
    • 25. Aoki M, Miyake K, Ogawa R et al. siRNA knockdown of tissue inhibitor of metalloproteinase-1 in keloid fibroblasts leads to degradation of collagen type I. J. Invest. Dermatol. 134(3), 818–826 (2014).
    • 26. Monaghan M, Browne S, Schenke-Layland K, Pandit A. A collagen-based scaffold delivering exogenous microRNA-29B to modulate extracellular matrix remodeling. Mol. Ther. 22(4), 786–796 (2014).
    • 27. Spies M, Nesic O, Barrow RE, Perez-Polo JR, Herndon DN. Liposomal IGF-1 gene transfer modulates pro- and anti-inflammatory cytokine mRNA expression in the burn wound. Gene Ther. 8(18), 1409–1415 (2001).
    • 28. Wang X, Coradin T, Hélary C. Modulating inflammation in a cutaneous chronic wound model by IL-10 released from collagen–silica nanocomposites via gene delivery. Biomater. Sci. 6(2), 398–406 (2018).
    • 29. Kim HS, Yoo HS. In vitro and in vivo epidermal growth factor gene therapy for diabetic ulcers with electrospun fibrous meshes. Acta Biomater. 9(7), 7371–7380 (2013).
    • 30. Chai J, Baatar D, Tarnawski A. Serum response factor promotes re-epithelialization and muscular structure restoration during gastric ulcer healing. Gastroenterology 126(7), 1809–1818 (2004).
    • 31. Luo J-D, Wang Y-Y, Fu W-L, Wu J, Chen AF. Gene therapy of endothelial nitric oxide synthase and manganese superoxide dismutase restores delayed wound healing in Type 1 diabetic mice. Circulation 110(16), 2484–2493 (2004).
    • 32. Koyama T, Hackl F, Aflaki P et al. A new technique of ex vivo gene delivery of VEGF to wounds using genetically modified skin particles promotes wound angiogenesis. J. Am. Coll. Surg. 212(3), 340–348 (2011).
    • 33. Waddington SN, Crossley R, Sheard V et al. Gene delivery of a mutant TGFβ3 reduces markers of scar tissue formation after cutaneous wounding. Mol. Ther. 18(12), 2104–2111 (2010).
    • 34. Shaterian A, Kao S, Chen L et al. The candidate tumor suppressor gene Ecrg4 as a wound terminating factor in cutaneous injury. Arch. Dermatol. Res. 305(2), 141–149 (2013).
    • 35. Wang C, Ma L, Gao C. Design of gene-activated matrix for the repair of skin and cartilage. Polym. J. 46(8), 476–482 (2014).
    • 36. Sosnowski BK, Engler R, Chandler LA. Developing a gene-activated matrix product for chronic wounds: a biotech's perspective. In: Advances in Wound Care: Volume 1 Mary Ann Liebert, Inc., NY, USA (2010).
    • 37. Sousa F, Passarinha L, Queiroz JA. Biomedical application of plasmid DNA in gene therapy: a new challenge for chromatography. Biotechnol. Genet. Eng. Rev. 26, 83–116 (2010).
    • 38. Sun L, Xu L, Chang H et al. Transfection with aFGF cDNA improves wound healing. J. Invest. Dermatol. 108(3), 313–318 (1997).
    • 39. Wolff JA, Malone RW, Williams P et al. Direct gene transfer into mouse muscle in vivo. Science 247(4949 Pt 1), 1465–1468 (1990).
    • 40. DeWeerdt S. RNA therapies explained. Nature 574(7778), S2–S3 (2019).
    • 41. Khalil AS, Yu X, Umhoefer JM et al. Single-dose mRNA therapy via biomaterial-mediated sequestration of overexpressed proteins. Sci. Adv. 6(27), eaba2422 (2020).
    • 42. Wahid F, Shehzad A, Khan T, Kim YY. MicroRNAs: synthesis, mechanism, function, and recent clinical trials. Biochim. Biophys. Acta Mol. Cell Res. 1803(11), 1231–1243 (2010).
    • 43. Yi R, O'Carroll D, Pasolli HA et al. Morphogenesis in skin is governed by discrete sets of differentially expressed microRNAs. Nat. Genet. 38(3), 356–362 (2006).
    • 44. Sen CK, Roy S. microRNA in cutaneous wound healing. In: Current Perspectives in microRNAs (miRNA). Ying S-Y (Ed.). Springer, Dordrecht, The Netherlands (2008).
    • 45. Dana H, Chalbatani GM, Mahmoodzadeh H et al. Molecular mechanisms and biological functions of siRNA. Int. J. Biomed. Sci. 13(2), 48–57 (2017).
    • 46. Meuli M, Liu Y, Liggitt D et al. Efficient gene expression in skin wound sites following local plasmid injection. J. Invest. Dermatol. 116(1), 131–135 (2001).
    • 47. Préat V, Dujardin N. Topical delivery of nucleic acids in the skin. STP Pharm. Sci. 11(1), 57 (2001).
    • 48. Nayerossadat N, Maedeh T, Ali PA. Viral and nonviral delivery systems for gene delivery. Adv. Biomed. Res. 1, 27 (2012).
    • 49. Cevher E, Sezer AD, Çağlar EŞ. Gene delivery systems: recent progress in viral and non-viral therapy. Recent Advances in Novel Drug Delivery Carrier Systems. Sezer AD (Ed.). IntechOpen, London, UK (2012).
    • 50. Park Jc, Terry Mj, Man Lx et al. 152 retroviral gene therapy with the gene for PDGF-B promotes wound healing in diabetic mice. Wound Repair Regen. 13(2), A28–A48 (2005).
    • 51. Stoff A, Rivera AA, Banerjee NS et al. Strategies to enhance transductional efficiency of adenoviral-based gene transfer to primary human fibroblasts and keratinocytes as a platform in dermal wounds. Wound Repair Regen. 14(5), 608–617 (2006).
    • 52. Vranckx JJ, Yao F, Petrie N et al. In vivo gene delivery of Ad-VEGF121 to full-thickness wounds in aged pigs results in high levels of VEGF expression but not in accelerated healing. Wound Repair Regen. 13(1), 51–60 (2005).
    • 53. Glorioso JC, Goins WF, Schmidt MC et al. Engineering herpes simplex virus vectors for human gene therapy. Adv. Pharmacol. 40, 103–136 (1997).
    • 54. Badillo AT, Chung S, Zhang L, Zoltick P, Liechty KW. Lentiviral gene transfer of SDF-1alpha to wounds improves diabetic wound healing. J. Surg. Res. 143(1), 35–42 (2007).
    • 55. Zhu L, Mahato RI. Lipid and polymeric carrier-mediated nucleic acid delivery. Expert Opin. Drug Deliv. 7(10), 1209–1226 (2010).
    • 56. Chuan D, Jin T, Fan R, Zhou L, Guo G. Chitosan for gene delivery: methods for improvement and applications. Adv. Colloid Interface Sci. 268, 25–38 (2019).
    • 57. Lungwitz U, Breunig M, Blunk T, Göpferich A. Polyethylenimine-based non-viral gene delivery systems. Eur. J. Pharm. Biopharm. 60(2), 247–266 (2005).
    • 58. Chauhan A. Dendrimers for drug delivery. Molecules 23(4), 938 (2018). www.mdpi.com/1420-3049/23/4/938
    • 59. Benizri S, Gissot A, Martin A, Vialet B, Grinstaff MW, Barthélémy P. Bioconjugated Oligonucleotides: Recent Developments and Therapeutic Applications. Bioconjug. Chem. 30(2), 366–383 (2019).
    • 60. Shepard JA, Wesson PJ, Wang CE et al. Gene therapy vectors with enhanced transfection based on hydrogels modified with affinity peptides. Biomaterials 32(22), 5092–5099 (2011).
    • 61. Shin S, Tuinstra HM, Salvay DM, Shea LD. Phosphatidylserine immobilization of lentivirus for localized gene transfer. Biomaterials 31(15), 4353–4359 (2010).
    • 62. Shin S, Salvay DM, Shea LD. Lentivirus delivery by adsorption to tissue engineering scaffolds. J. Biomed. Mater. Res. A 93(4), 1252–1259 (2010).
    • 63. Hengge UR, Chan EF, Foster RA, Walker PS, Vogel JC. Cytokine gene expression in epidermis with biological effects following injection of naked DNA. Nat. Genet. 10(2), 161–166 (1995).
    • 64. Eriksson E, Yao F, Svensjö T et al. In vivo gene transfer to skin and wound by microseeding. J. Surg. Res. 78(2), 85–91 (1998).
    • 65. Eming SA, Whitsitt JS, He L, Krieg T, Morgan JR, Davidson JM. Particle-mediated gene transfer of PDGF isoforms promotes wound repair. J. Invest. Dermatol. 112(3), 297–302 (1999).
    • 66. Jeschke MG, Richter G, Herndon DN et al. Therapeutic success and efficacy of nonviral liposomal cDNA gene transfer to the skin in vivo is dose dependent. Gene Ther. 8(23), 1777–1784 (2001).
    • 67. Doukas J, Chandler LA, Gonzalez AM et al. Matrix immobilization enhances the tissue repair activity of growth factor gene therapy vectors. Hum. Gene Ther. 12(7), 783–798 (2001).
    • 68. Schwarz KW, Murray MT, Sylora R, Sohn RL, Dulchavsky SA. Augmentation of wound healing with translation initiation factor eIF4E mRNA. J. Surg. Res. 103(2), 175–182 (2002).
    • 69. Nguyen PD, Tutela JP, Thanik VD et al. Improved diabetic wound healing through topical silencing of p53 is associated with augmented vasculogenic mediators. Wound Repair Regen. 18(6), 553–559 (2010).
    • 70. Sun N, Ning B, Hansson KM et al. Modified VEGF-A mRNA induces sustained multifaceted microvascular response and accelerates diabetic wound healing. Sci Rep. 8(1), 17509 (2018).
    • 71. Icli B, Nabzdyk CS, Lujan-Hernandez J et al. Regulation of impaired angiogenesis in diabetic dermal wound healing by microRNA-26a. J. Mol. Cell. Cardiol. 91, 151–159 (2016).
    • 72. Khalil AS, Yu X, Umhoefer JM et al. Single-dose mRNA therapy via biomaterial-mediated sequestration of overexpressed proteins. Sci. Adv. 6(27), eaba2422 (2020).
    • 73. Randeria PS, Seeger MA, Wang X-Q et al. siRNA-based spherical nucleic acids reverse impaired wound healing in diabetic mice by ganglioside GM3 synthase knockdown. Proc. Natl Acad. Sci. USA 112(18), 5573–5578 (2015).
    • 74. Martin JR, Nelson CE, Gupta MK et al. Local delivery of PHD2 siRNA from ROS-degradable scaffolds to promote diabetic wound healing. Adv. Healthc. Mater. 5(21), 2751–2757 (2016).
    • 75. Ghatak S, Li J, Chan YC et al. AntihypoxamiR functionalized gramicidin lipid nanoparticles rescue against ischemic memory improving cutaneous wound healing. Nanomedicine 12(7), 1827–1831 (2016).
    • 76. Dang CP, Leelahavanichkul A. Over-expression of miR-223 induces M2 macrophage through glycolysis alteration and attenuates LPS-induced sepsis mouse model, the cell-based therapy in sepsis. PLOS ONE 15(7), e0236038 (2020).
    • 77. Lan B, Zhang L, Yang L et al. Sustained delivery of MMP-9 siRNA via thermosensitive hydrogel accelerates diabetic wound healing. J. Nanobiotechnol. 19(1), 130 (2021).
    • 78. Lin MP, Marti GP, Dieb R et al. Delivery of plasmid DNA expression vector for keratinocyte growth factor-1 using electroporation to improve cutaneous wound healing in a septic rat model. Wound Rep. Regen. 14(5), 618–624 (2006).
    • 79. Kwon MJ, An S, Choi S et al. Effective healing of diabetic skin wounds by using nonviral gene therapy based on minicircle vascular endothelial growth factor DNA and a cationic dendrimer. J. Gene Med. 14(4), 272–278 (2012).
    • 80. Guo R, Xu S, Ma L, Huang A, Gao C. The healing of full-thickness burns treated by using plasmid DNA encoding VEGF-165 activated collagen-chitosan dermal equivalents. Biomaterials 32(4), 1019–1031 (2011).
    • 81. Wang S, Yan C, Zhang X et al. Antimicrobial peptide modification enhances the gene delivery and bactericidal efficiency of gold nanoparticles for accelerating diabetic wound healing. Biomater. Sci. 6(10), 2757–2772 (2018).
    • 82. Wang P, Huang S, Hu Z et al. In situ formed anti-inflammatory hydrogel loading plasmid DNA encoding VEGF for burn wound healing. Acta Biomater. 100, 191–201 (2019).
    • 83. Bitto A, Minutoli L, Galeano MR et al. Angiopoietin-1 gene transfer improves impaired wound healing in genetically diabetic mice without increasing VEGF expression. Clin. Sci. (Lond) 114(12), 707–718 (2008).
    • 84. Chesnoy S, Lee P-Y, Huang L. Intradermal injection of transforming growth factor-beta1 gene enhances wound healing in genetically diabetic mice. Pharm. Res. 20(3), 345–350 (2003).
    • 85. Yamamoto A, Kormann M, Rosenecker J, Rudolph C. Current prospects for mRNA gene delivery. Eur. J. Pharm. Biopharm. 71(3), 484–489 (2009).
    • 86. Gan L-M, Lagerström-Fermér M, Carlsson LG et al. Intradermal delivery of modified mRNA encoding VEGF-A in patients with type 2 diabetes. Nat. Commun. 10(1), 871 (2019).
    • 87. Ohki Y, Heissig B, Sato Y et al. Granulocyte colony-stimulating factor promotes neovascularization by releasing vascular endothelial growth factor from neutrophils. FASEB J. 19(14), 2005–2007 (2005).
    • 88. Cianfarani F, Tommasi R, Failla CM et al. Granulocyte/macrophage colony-stimulating factor treatment of human chronic ulcers promotes angiogenesis associated with de novo vascular endothelial growth factor transcription in the ulcer bed. Br. J. Dermatol. 154(1), 34–41 (2006).
    • 89. Li H, Chang L, Du WW et al. Anti-microRNA-378a enhances wound healing process by upregulating integrin beta-3 and vimentin. Mol. Ther. 22(10), 1839–1850 (2014).
    • 90. Tang H, Wang X, Zhang M et al. MicroRNA-200b/c-3p regulate epithelial plasticity and inhibit cutaneous wound healing by modulating TGF-β-mediated RAC1 signaling. Cell Death Dis. 11(10), 1–17 (2020).
    • 91. Zhou J, Zhao Y, Simonenko V et al. Simultaneous silencing of TGF-β1 and COX-2 reduces human skin hypertrophic scar through activation of fibroblast apoptosis. Oncotarget 8(46), 80651–80665 (2017).
    • 92. Li J, Ghatak S, El Masry MS et al. Topical lyophilized targeted lipid nanoparticles in the restoration of skin barrier function following burn wound. Mol. Ther. 26(9), 2178–2188 (2018).
    • 93. Lucas T, Schäfer F, Müller P, Eming SA, Heckel A, Dimmeler S. Light-inducible antimiR-92a as a therapeutic strategy to promote skin repair in healing-impaired diabetic mice. Nat. Commun. 8(1), 15162 (2017).
    • 94. Icli B, Wu W, Ozdemir D et al. MicroRNA-615-5p regulates angiogenesis and tissue repair by targeting AKT/eNOS (protein kinase B/endothelial nitric oxide synthase) signaling in endothelial cells. Arterioscler. Thromb. Vasc. Biol. 39(7), 1458–1474 (2019).
    • 95. Wang J-M, Tao J, Chen D-D et al. MicroRNA miR-27b rescues bone marrow–derived angiogenic cell function and accelerates wound healing in type 2 diabetes mellitus. Arterioscler. Thromb. Vasc. Biol. 34(1), 99–109 (2014).
    • 96. Lang H, Zhao F, Zhang T et al. MicroRNA-149 contributes to scarless wound healing by attenuating inflammatory response. Mol. Med. Rep. 16(2), 2156–2162 (2017).
    • 97. Saleh B, Dhaliwal HK, Portillo-Lara R et al. Local immunomodulation using an adhesive hydrogel loaded with miRNA-laden nanoparticles promotes wound healing. Small 15(36), 1902232 (2019).
    • 98. Miscianinov V, Martello A, Rose L et al. MicroRNA-148b targets the TGF-β pathway to regulate angiogenesis and endothelial-to-mesenchymal transition during skin wound healing. Mol. Ther. 26(8), 1996–2007 (2018).
    • 99. Wang SY, Kim H, Kwak G et al. Development of microRNA-21 mimic nanocarriers for the treatment of cutaneous wounds. Theranostics 10(7), 3240–3253 (2020).
    • 100. Li N, Luo H-C, Yang C et al. Cationic star-shaped polymer as an siRNA carrier for reducing MMP-9 expression in skin fibroblast cells and promoting wound healing in diabetic rats. Int. J. Nurs. 9(1), 3377–3387 (2014).
    • 101. Ye J, Xie T, Niu Y et al. A potential mechanism for diabetic wound healing: cutaneous environmental disorders. In: Wound Healing - New insights into Ancient Challenges [Internet]. Alexandrescu VA (Ed.). IntechOpen, London (2016).
    • 102. Kasiewicz LN, Whitehead KA. Lipid nanoparticles silence tumor necrosis factor α to improve wound healing in diabetic mice. Bioeng. Transl. Med. 4(1), 75–82 (2018).
    • 103. Kasiewicz LN, Whitehead KA. Silencing TNFα with lipidoid nanoparticles downregulates both TNFα and MCP-1 in an in vitro co-culture model of diabetic foot ulcers. Acta Biomater. 32, 120–128 (2016).
    • 104. O'Rourke BP, Kramer AH, Cao LL et al. Fidgetin-like 2 siRNA enhances the wound healing capability of a surfactant polymer dressing. Adv. Wound Care 8(3), 91–100 (2019).
    • 105. Edson J, Kwon YJ. RNAi for silencing drug resistance in microbes toward development of nanoantibiotics. J.Control. Release 189, 150–157 (2014).
    • 106. Zhao R, Yan Q, Huang H, Lv J, Ma W. Transdermal siRNA-TGFβ1–337 patch for hypertrophic scar treatment. Matrix Biol. 32(5), 265–276 (2013).
    • 107. Tissue Repair Company. Growth factor gene therapy for wound healing. clinicaltrials.gov. https://clinicaltrials.gov/ct2/show/NCT00065663
    • 108. University of Pennsylvania. Phase I trial to evaluate the safety of platelet-derived growth factor B (PDGF-B) and a limb compression bandage in venous leg ulcers. clinicaltrials.gov. https://clinicaltrials.gov/ct2/show/NCT00000431
    • 109. Kessler JA, Smith AG, Cha B-S et al. Double-blind, placebo-controlled study of HGF gene therapy in diabetic neuropathy. Ann. Clin. Transl. Neurol. 2(5), 465–478 (2015).
    • 110. Losordo DW. VEGF gene transfer for critical limb ischemia. clinicaltrials.gov. https://clinicaltrials.gov/ct2/show/NCT00304837
    • 111. AnGes USA, Inc. A phase II double-blind, randomized, placebo-controlled study to assess the safety and efficacy of AMG0001 to improve perfusion in critical limb ischemia in subjects who have peripheral ischemic ulcers. clinicaltrials.gov. https://clinicaltrials.gov/ct2/show/NCT00060892
    • 112. Kessler JA, Shaibani A, Sang CN et al. Gene therapy for diabetic peripheral neuropathy: a randomized, placebo-controlled phase III study of VM202, a plasmid DNA encoding human hepatocyte growth factor. Clin. Transl. Sci. 14(3), 1176–1184 (2021).
    • 113. Gurevich I, Agarwal P, Zhang P et al. In vivo topical gene therapy for recessive dystrophic epidermolysis bullosa: a phase 1 and 2 trial. Nat. Med. 28(4), 780–788 (2022).