Skip to main content
Log in

Calorimetric and spectrophotometric investigation of PLGA nanoparticles and their complex with DNA

  • Published:
Journal of Thermal Analysis and Calorimetry Aims and scope Submit manuscript

Abstract

The calorimetric investigation of non-coated and chitosan-coated PLGA nanoparticles (NP) shows that at initial temperatures of heating particle swelling takes place what results in an internal architectural change at lower than physiological temperature. It has shown that the temperature of NP tightness perturbing depends on solvent polarity: as more polar is the solvent more stable are particles. The break of existing bonds in NP shell is accompanied with heat absorption peak which undergoes significant changes depending on heating rate. In the wide pH 2–8 interval in transition temperature no changes occurred. The obtained results show that such NP could be used in acidic area for drug transfer, which gives possibility to take medicine orally. It was shown that DNA attaches only to chitosan-coated NP. The optimal ratio for DNA loading onto the NP was found to be 7:1 (WNP/WDNA).

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.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10
Fig. 11
Fig. 12
Fig. 13

Similar content being viewed by others

References

  1. Panyam J, Labhasetwar V. Biodegradable nanoparticles for drug and gene delivery to cells and tissue. Adv Drug Del Rev. 2003;55:329–47.

    Article  CAS  Google Scholar 

  2. Patil Y, Panyam J. Polymeric nanoparticles for siRNA delivery and gene silencing. Int J Pharm. 2009;367:195–203.

    Article  CAS  Google Scholar 

  3. Patil Y, Toti U, Khdair A, Ma L, Panyam J. Facile single-step multifunctionalization of nanoparticles for targeted drug delivery. Biomaterials. 2009;30:859–66.

    Article  CAS  Google Scholar 

  4. Bejjani RA, BenEzra D, Cohen H, Rieger J, Andrieu C, Jeanny J, et al. Nanoparticles for gene delivery to retinal pigment epithelial cells. Mol Vis. 2005;11:124–32.

    CAS  Google Scholar 

  5. Nishikawa M, Huang L. Nonviral vectors in the new millennium: delivery barriers in gene transfer. Hum Gene Ther. 2001;12:861–70.

    Article  CAS  Google Scholar 

  6. Li S, Huang L. Nonviral gene therapy: promises and challenges. Gene Ther. 2000;7:31–4.

    Article  CAS  Google Scholar 

  7. Ferber D. Gene therapy: safer and virus-free? Science. 2001;294:1638–42.

    Article  CAS  Google Scholar 

  8. Moghimi SM, Hunter AC, Murray JC. Long circulating and target-specific nanoparticles: theory to practice. Pharmacol Rev. 2001;53:283–318.

    CAS  Google Scholar 

  9. Douglas SJ, Davis SS, Illum L. Nanoparticles in drug delivery. Crit Rev Ther Drug Carrier Syst. 1987;3:233–61.

    CAS  Google Scholar 

  10. Kreuter J, Tauber U, Illi V. Distribution and elimination of poly(methyl methacrylate) nanoparticles after subcutaneous administration to rats. J Pharm Sci. 1979;68:1443–9.

    Article  CAS  Google Scholar 

  11. Hasadsri L, Kreuter J, Hattori H, Iwasaki T, George JM. Functional protein delivery into neurons using polymeric nanoparticles. J Biol Chem. 2009;284:6972–81.

    Article  CAS  Google Scholar 

  12. Brown MD, Schatzlein AG, Uchegbu IF. Gene delivery with synthetic (non viral) carriers. Int J Pharm. 2001;229:1–21.

    Article  CAS  Google Scholar 

  13. Fievez V, Plapied L, des Rieux A, Pourcelle V, Freichels H, Wascotte V, Vanderhaeghen ML, Jerôme C, Vanderplasschen A, Marchand-Brynaert J, Schneider YJ, Préat V. Targeting nanoparticles to M cells with non-peptidic ligands for oral vaccination. Eur J Pharm Biopharm 2009; May 3 [Epub ahead of print].

  14. De S, Robinson D. Polymer relationships during preparation of chitosan-alginate and poly-l-lysine-alginate nanospheres. J Control Rel. 2003;89:101–12.

    Article  CAS  Google Scholar 

  15. Ricci M, Blasi P, Giovagnoli S, Perioli L, Vescovi C, Rossi C. Leucinostatin-A loaded nanospheres: characterization and in vivo toxicity and efficacy evaluation. Int J Pharm. 2004;275:61–72.

    Article  CAS  Google Scholar 

  16. Muller M, Voros J, Csucs G, Walter E, Danuser G, Textor M, et al. Surface modification of PLGA microspheres. J Biomed Mater Res. 2003;66A:55–61.

    Article  Google Scholar 

  17. Feng SS, Huang GF, Mu L. Nanospheres of biodegradable polymers: a system for clinical administration of an anticancer drug paclitaxel. Ann Acad Med Singapore. 2000;29:633–9.

    CAS  Google Scholar 

  18. Feng SS, Huang GF. Effects of emulsifiers on the controlled release of paclitaxel (Taxol) from nanospheres of biodegradable polymers. J Control Rel. 2001;71:53–69.

    Article  CAS  Google Scholar 

  19. Pan J, Feng SS. Targeting and imaging cancer cells by folate decorated, “quantum dots loaded nanoparticles of biodegradable polymers”. Biomaterials. 2009;30:1176–83.

    Article  CAS  Google Scholar 

  20. Mu L, Feng SS. Fabrication, characterization and in vivo release of paclitaxel (Taxol) loaded poly(lactic-co-glycolic acid) microspheres prepared by spray drying technique with lipid/cholesterol emulsifiers. J Control Rel. 2001;76:239–54.

    Article  CAS  Google Scholar 

  21. Mu L, Feng SS. Vitamin E TPGS used as emulsifier in the solvent evaporation/extraction technique for fabrication of polymeric nanospheres for controlled release of paclitaxel (Taxol). J Control Rel. 2002;80:129–44.

    Article  CAS  Google Scholar 

  22. Labhasetwar V. Nanoparticles for drug delivery. Pharm News. 1997;4:28–31.

    CAS  Google Scholar 

  23. Allemann E, Leroux JC, Gurny R. Polymeric nano- and microparticles for the oral delivery of peptides and peptidomimetics. Adv Drug Del Rev. 1998;34:171–89.

    Article  CAS  Google Scholar 

  24. Nishioka Y, Yoshino H. Lymphatic targeting with nanoparticulate system. Adv Drug Del Rev. 2001;47:55–64.

    Article  CAS  Google Scholar 

  25. Soppimath KS, Aminabhavi TM, Kulkarni AR, Rudzinski WE. Biodegradable polymeric nanoparticles as drug delivery devices. J Contr Rel. 2001;70:1–20.

    Article  CAS  Google Scholar 

  26. Hariharan S, Bhardwaj V, Bala I, Sitterberg J, Bakowsky U, Kumar R. Design of estradiol loaded PLGA nanoparticulate formulations: a potential oral delivery system for hormone therapy. Pharm Res. 2006;23:184–95.

    Article  CAS  Google Scholar 

  27. Mittal G, Sahana D, Bhardwaj V, Kumar R. Estradiol loaded PLGA nanoparticles for oral administration: effect of polymer molecular weight and copolymer composition on release behavior in vitro and in vivo. J Control Rel. 2007;119:77–85.

    Article  CAS  Google Scholar 

  28. Sonaje K, Italia J, Sharma G, Bhardwaj V, Tikoo K, Kumar R. Development of biodegradable nanoparticles for oral delivery of ellagic acid and evaluation of their antioxidant efficacy against cyclosporine A-induced nephrotoxicity in rats. Pharm Res. 2007;24:899–908.

    Article  CAS  Google Scholar 

  29. Jeon HJ, Jeong YL, Jang MK, Park YH, Nah JW. Effect of solvent on the preparation of surfactant-free poly (d, l-lactide-co-glycolide) nanoparticles and norfloxacin release characteristics. Int J Pharm. 2000;207:99–108.

    Article  CAS  Google Scholar 

  30. Day M, Nawaby AV, Liao X. A DSC study of the crystallization behaviour of polylactic acid and its nanocomposites. J Therm Anal Cal. 2006;86:623–9.

    Article  CAS  Google Scholar 

  31. Cilurzo F, Selmin F, Liberti V, Montanari L. Thermal characterization of poly(lactide-co-glycolide) microspheres containing bupivacaine base polymorphs. J Therm Anal Cal. 2005;79:9–12.

    Article  CAS  Google Scholar 

  32. Anderson JM, Shive MS. Biodegradation and biocompatibility of PLA and PLGA microspheres. Adv Drug Del Rev. 1997;28:5–24.

    Article  CAS  Google Scholar 

  33. Jain RA. The manufacturing techniques of various drug loaded biodegradable poly (lactide-co-glycolide) (PLGA) devices. Biomaterials. 2000;21:2475–90.

    Article  CAS  Google Scholar 

  34. Langer R. Tissue engineering: a new field and its challenges. Pharm Res. 1997;14:840–1.

    Article  CAS  Google Scholar 

  35. Uhrich KE, Cannizzaro SM, Langer RS, Shakeshelf KM. Polymeric systems for controlled drug release. Chem Rev. 1999;99:3181–98.

    Article  CAS  Google Scholar 

  36. Vert M, Schwach G, Engel R, Coudane J. Something new in the field of PLA/GA bioresorbable polymers? J Control Rel. 1998;53:85–92.

    Article  CAS  Google Scholar 

  37. Hans MK, Lowman AM. Biodegradable nanoparticles for drug delivery and targeting. Curr Opin Solid State Mater Sci. 2002;6:319–27.

    Article  CAS  Google Scholar 

  38. Sahoo SK, Panyam J, Prabha S, Labhasetwar V. Residual polyvinyl alcohol associated with poly(d, l-lactide-co-glycolide) nanoparticles affects their physical properties and cellular uptake. J Contr Rel. 2002;82:105–14.

    Article  CAS  Google Scholar 

  39. Jawahar N, Eagappanath T, Venkatesh N, Jubie S, Samanta MK. Preparation and characterisation of PLGA-nanoparticles containing an anti-hypertensive agent. Int J Pharm Tech Res. 2009;1:390–3.

    CAS  Google Scholar 

  40. Bala I, Haribaran S, Kumar R. PLGA nanoparticles in drug delivery: the state of the art. Crit Rev Ther Drug Carrier Syst. 2004;21:387–422.

    Article  CAS  Google Scholar 

  41. Hyon SH. Biodegtadable poly(lactic acid) microspheres for drug delivery systems. Yonsei Med J. 2000;41:720–34.

    CAS  Google Scholar 

  42. Hanafusa S, Matsusue Y, Yasunaga T, Yamamuro T, Oka M, Shikinami Y, et al. Biodegradable plate fixation of rabbit femoral shaft osteotomies. A comparative study. Clin Orthop Relat Res. 1995;315:262–71.

    Google Scholar 

  43. Matsusue Y, Hanafusa S, Yamamuro T, Shikinami Y, Lkada Y. Tissue reaction of bioabsorbable ultra high strength poly (l-lactide) rod. A long-term study in rabbits. Clin Orthop Relat Res. 1995;317:246–53.

    Google Scholar 

  44. Mooney DJ, Sano K, Kaufmann PM, Majahod K, Schloo B, Vacanti JP, et al. Long-term engraftment of hepatocytes transplanted on biodegradable polymer sponges. J Biomed Mater Res. 1997;37:413–20.

    Article  CAS  Google Scholar 

  45. Eiselt P, Kim BS, Chacko B, Isenberg B, Peters MC, Greene KG, et al. Development of technologies aiding large-tissue engineering. Biotechnol Prog. 1998;14:134–40.

    Article  CAS  Google Scholar 

  46. Cartiera MS, Johnson KM, Rajendran V, Caplan MJ, Saltzman WM. The uptake and intracellular fate of PLGA nanoparticles in epithelial cells. Biomaterials. 2009;30:2790–8.

    Article  CAS  Google Scholar 

  47. Leong KW, Langer R. Polymeric controlled drug delivery. Adv Drug Del Rev. 1987;1:33–199.

    Google Scholar 

  48. Park TG. Degradation of poly(lactide-co-glycolide acid) microspheres: effect of copolymer composition. Biomaterials. 1995;16:1123–30.

    Article  CAS  Google Scholar 

  49. Hedley ML, Curley J, Urban R. Microspheres containing plasmid-encoded antigens elicit cytotoxic T-cell responses. Nat Med. 1998;4:365–8.

    Article  CAS  Google Scholar 

  50. Lin SY, Chen KS, Teng HH, Li MJ. In vitro degradation and dissolution behaviours of microspheres prepared by three low molecular weight polyesters. J Microencapsul. 2000;17:577–86.

    Article  CAS  Google Scholar 

  51. Brannon-Peppas L. Recent advances on the use of biodegradable microparticles and nanoparticles in controlled drug delivery. Int J Pharm. 1995;116:1–9.

    Article  CAS  Google Scholar 

  52. Görner T, Gref R, Michenot D, Sommer F, Tran MN, Dellacherie E. Lidocaine-loaded biodegradable nanosperes. I. Optimization of the drug incorporation into the polymer matrix. J Contr Rel. 1999;57:259–69.

    Article  Google Scholar 

  53. Burkersroda FV, Schedl L, Göpferich A. Why degradable polymers undergo surface erosion or bulk erosion. Biomaterials. 2002;23:4221–31.

    Article  Google Scholar 

  54. Kneuer C, Sameti M, Haltner EG, Schiestel T, Schirra H, Schmidt H, et al. Silica nanoparticles modified with aminosilanes as carriers for plasmid DNA. Int J Pharm. 2000;196:257–61.

    Article  CAS  Google Scholar 

  55. Kneuer C, Sameti M, Bakowsky U, Schiestel T, Schirra H, Schmidt H, et al. A nonviral DNA delivery system based on surface modified silica-nanoparticles can efficiently transfect cells in vitro. Bioconjug Chem. 2000;11:926–32.

    Article  CAS  Google Scholar 

  56. Florence AT, Sakthivel T, Toth I. Oral uptake and translocation of a polylysine dendrimer with a lipid surface. J Control Rel. 2000;65:253–9.

    Article  CAS  Google Scholar 

  57. Ramaswamy C, Sakthivel T, Wilderspin AF, Florence AT. Dendriplexes and their characterization. Int J Pharm. 2003;254:17–21.

    Article  CAS  Google Scholar 

  58. Pouton CW, Lucas P, Thomas BJ, Uduehi AN, Mikroy DA, Moss SH. Polycation-DNA complexes for gene delivery: a comparison of the biopharmaceutical properties of cationic polypeptides and cationic lipids. J Control Rel. 1998;53:289–99.

    Article  CAS  Google Scholar 

  59. Ramsay E, Gumbleton M. Polylysine and polyornithine gene transfer complexes: a study of complex stability and cellular uptake as a basis for their differential in vitro transfection efficiency. J Drug Target. 2002;10:1–9.

    Article  CAS  Google Scholar 

  60. Oupicky D, Konak C, Ulbrich K, Wolfert MA, Seymour LW. DNA delivery systems based on complexes of DNA with synthetic polycations and their copolymers. J Control Rel. 2000;65:149–71.

    Article  CAS  Google Scholar 

  61. Blacklock J, You YZ, Zhou QH, Mao G, Oupicky D. Gene delivery in vitro and in vivo from bioreducible multilayered polyelectrolyte films of plasmid DNA. Biomaterials. 2009;30:939–50.

    Article  CAS  Google Scholar 

  62. Wagner E, Plank C, Zatloukal K, Cotton M, Birnstiel ML. Influenza virus hemagglutinin HA-2 N-terminal fusogenic peptides augment gene transfer by transferrin-polylysine-DNA complexes: toward a synthetic virus-like gene-transfer vehicle. Proc Natl Acad Sci USA. 1992;89:7934–8.

    Article  CAS  Google Scholar 

  63. Fischer D, Bieber T, Li Y, Elsasser HP, Kissel T. A novel non-viral vector for DNA delivery based on low molecular weight, branched polyethylenimine: effect of molecular weight on transfection efficiency and cytotoxicity. Pharm Res. 1999;16:1273–9.

    Article  CAS  Google Scholar 

  64. Kunath K, Harpe A, Fischer D, Petersen H, Bickel U, Voigt K, et al. Low-molecular-weight polyethylenimine as a non-viral vector for DNA delivery: comparison of physicochemical properties, transfection efficiency and in vivo distribution with high-molecular-weight polyethylenimine. J Control Rel. 2003;89:113–25.

    Article  CAS  Google Scholar 

  65. Singh M, Briones M, Ott G, O’Hagan D. Cationic microparticles: a potent delivery system for DNA vaccines. Proc Natl Acad Sci USA. 2000;97:811–6.

    Article  CAS  Google Scholar 

  66. Singh M, Ott G, Kazzaz J, Ugozzoli M, Briones M, Donnelly J, et al. Cationic microparticles are an effective delivery system for immune stimulatory CpG DNA. Pharm Res. 2001;18:1476–9.

    Article  CAS  Google Scholar 

  67. Singh M, Vajdy M, Gardner J, Briones M, O’Hagan D. Mucosal immunization with HIV-1 gag DNA on cationic microparticles prolongs gene expression and enhances local and systemic immunity. Vaccine. 2001;20:2–594.

    Article  Google Scholar 

  68. Smith JG, Wedeking T, Vernachio JH, Way H, Niven RW. Characterization and in vivo testing of a heterogeneous cationic lipid-DNA formulation. Pharm Res. 1998;15:1356–63.

    Article  CAS  Google Scholar 

  69. Olbrich C, Bakowsky U, Lehr C-M, Muller RH, Kneuer C. Cationic solid-lipid nanoparticles can efficiently bind and transfect plasmid DNA. J Control Rel. 2001;77:345–55.

    Article  CAS  Google Scholar 

  70. Oberle V, Bakowsky U, Zuhorn IS, Hoekstra D. Lipoplex formation under equilibrium conditions reveals a three step mechanism. Biophys J. 2000;79:1447–54.

    Article  CAS  Google Scholar 

  71. Mahato RI, Kawabata K, Nomura T, Takakura Y, Hashida M. Physicochemical and pharmacokinetic characteristics of plasmid DNA/cationic liposome complexes. J Pharm Sci. 1995;84:1267–71.

    Article  CAS  Google Scholar 

  72. Cui Z, Mumper RJ. Plasmid dna-entrapped nanoparticles engineered from microemulsion precursors: in vitro and in vivo evaluation. Bioconjug Chem. 2002;13:1319–27.

    Article  CAS  Google Scholar 

  73. Farhood H, Serbina N, Huang L. The role of dioleyl phosphatidylethanolamine in cationic liposome mediated gene transfer. Biochim Biophys Acta. 1995;1235:289–95.

    Article  Google Scholar 

  74. Sternberg B, Hong K, Zheng W, Papahadjopoulos D. Ultrastructural characterization of cationic liposome-DNA complexes showing enhanced stability in serum and high transfection activity in vivo. Biochim Biophys Acta. 1998;1375:23–35.

    Article  CAS  Google Scholar 

  75. Meyer O, Kirpotin D, Hong K, Sternberg B, Park J, Woodle M, et al. Cationic liposomes coated with polyethylene glycol as carriers for oligonucleotides. J Biol Chem. 1998;273:15621–7.

    Article  CAS  Google Scholar 

  76. Behr JP, Demeneix B, Loeffler JP, Perez-Mutul J. Efficient gene transfer into mammalian primary endocrine cells with lipopolyamine-coated DNA. Proc Natl Acad USA. 1989;86:6982–6.

    Article  CAS  Google Scholar 

  77. Torchilin VP, Levchenko TS, Rammohan R, Volodina N, Papahadjopoulos-Sternberg B, D’Souza GG. Cell transfection in vitro and in vivo with nontoxic TAT peptide–liposome–DNA complexes. Proc Natl Acad Sci USA. 2003;100:1972–7.

    Article  CAS  Google Scholar 

  78. Rädler J, Koltover I, Salditt T, Safinya C. Structure of DNA–cationic liposome complexes: DNA intercalation in multilamellar membranes in distinct interhelical packing regimes. Science. 1997;7:810–4.

    Article  Google Scholar 

  79. Mrevlishvili G, Kankia B, Mdzinarashvili T, Brelidze T, Khvedelidze M, Metreveli N, et al. Liposome–DNA interaction: microcalorimetric study. Chem Phys Lipids. 1998;94:139–43.

    Article  CAS  Google Scholar 

  80. Zuzzi S, Onori G, Cametti C. Thermal stability of DNA in DNA-induced dotap liposome aggregates. J Therm Anal Cal. 2008;93:527–33.

    Article  CAS  Google Scholar 

  81. Cui Z, Mumper RJ. Plasmid DNA-entrapped nanoparticles engineered from microemulsion precursors: in vitro and in vivo evaluation. Bioconjug Chem. 2002;13:1319–27.

    Article  CAS  Google Scholar 

  82. Kumar R, Bakowsky U, Lehr C-M. Preparation and characterization of cationic PLGA nanospheres as DNA carriers. Biomaterials. 2004;25:1771–7.

    Article  CAS  Google Scholar 

  83. Kumar R, Mohapatra SS, Kong X, Jena P, Bakowsky U, Lehr C-M. Cationic poly(lactide-co-glycolide) nanoparticles as efficient in vivo gene transfection agents. J Nanosci Nanotechnol. 2004;4:990–4.

    Article  CAS  Google Scholar 

  84. Zoua W, Liua C, Chena Z, Zhang N. Studies on bioadhesive PLGA nanoparticles: a promising gene delivery system for efficient gene therapy to lung cancer. Int J Pharm. 2009;370:187–95.

    Article  Google Scholar 

  85. Lira AAM, Nanclares DMA, Federman Neto A, Marchetti JM. Drug-polymer interaction in the all-trans retinoic acid release from chitosan microparticles. J Therm Anal Cal. 2007;87:899–903.

    Article  CAS  Google Scholar 

  86. Haas J, Ravi K, Borchard G, Bakowsky U, Lehr C-M. Preparation and characterization of chitosan and trimethyl-chitosan-modified poly-(ε-caprolactone) nanoparticles as DNA carriers. AAPS Pharm Sci Tech. 2005;6:22–30.

    Article  Google Scholar 

  87. Luengo J, Weiss B, Schneider M, Ehlers A, Stracke F, König K, et al. Influence of nanoencapsulation on human skin transport of flufenamic acid. Skin Pharmacol Physiol. 2006;19:190–7.

    Article  CAS  Google Scholar 

  88. Xu P, Gullotti E, Tong L, Highley CB, Errabelli DR, Hasan T, et al. Intracellular drug delivery by poly(lactic-co-glycolic acid) nanoparticles, revisited. Mol Pharm. 2009;6:190–201.

    Article  CAS  Google Scholar 

  89. Nafee N, Taetz S, Schneider M, Schaefer U, Lehr C-M. Chitosan-coated PLGA nanoparticles for DNA/RNA delivery: effect of the formulation parameters on complexation and transfection of antisense oligonucleotides. Nanomedicine: nanotechnology. Biol Med. 2007;3:173–83.

    CAS  Google Scholar 

  90. Privalov PL, Potekhin SA. Scanning microcalorimetry in studying temperature-induced changes in proteins. Meth Enzim. 1986;131:4–51.

    Article  CAS  Google Scholar 

  91. De S, Robinson DH. Particle size and temperature effect on the physical stability of PLGA nanospheres and microspheres containing Bodipy. AAPS PharmSciTech. 2004;5:1–7.

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Tamaz Mdzinarashvili.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Khvedelidze, M., Mdzinarashvili, T., Partskhaladze, T. et al. Calorimetric and spectrophotometric investigation of PLGA nanoparticles and their complex with DNA. J Therm Anal Calorim 99, 337–348 (2010). https://doi.org/10.1007/s10973-009-0137-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10973-009-0137-x

Keywords

Navigation