Skip to main content
Log in

Proton Oriented-“Smart Depot” for Responsive Release of Ca2+ to Inhibit Peptide Acylation in PLGA Microspheres

  • Research Paper
  • Published:
Pharmaceutical Research Aims and scope Submit manuscript

Abstract

Purpose

The purpose of this study was to characterize and detail the mechanism of a smart Ca2+ release depot (Ca3(PO4)2) about its ability for sustainable inhibition on peptide acylation within PLGA microspheres.

Methods

The octreotide acetate release and acylation kinetics were analyzed by RP-HPLC. Changes of Ca2+ concentration and adsorption behavior were determined by a Calcium Colorimetric Assay Kit. The inner pH changes were delineated by a classic pH sensitive probe, Lysosensor yellow/ blue® dextran. Morphological changes of microspheres, adsorption between polymer and additive, transformation of Ca3(PO4)2 were characterized using SEM, FTIR and SSNMR separately.

Results

Before and after microspheres formulation, the property and effectiveness of Ca3(PO4)2 were investigated. Compared with a commonly used calcium salt (CaCl2), high encapsulation efficiency (96.56%) of Ca3(PO4)2 guarantees lasting effectiveness. In an increasingly acidic environment that simulated polymer degradation, the poorly water-soluble Ca3(PO4)2 could absorb protons and transform into the more and more soluble CaHPO4 and Ca(H2PO4)2 to produce sufficient Ca2+ according to severity of acylation. The corresponding Ca2+ produce capacity fully met the optimum inhibition requirement since the real-time adsorption sites (water-soluble carboxylic acids) inside the degrading microspheres were rare. A sustained retention of three switchable calcium salts and slow release of Ca2+ were observed during the microsphere incubation. FTIR results confirmed the long-term inhibition effect induced by Ca3(PO4)2 on the adsorption between drug and polymer.

Conclusions

With the presence of the smart Ca2+ depot (Ca3(PO4)2) in the microspheres, a sustainable and long-term inhibition of peptide acylation was achieved.

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

Similar content being viewed by others

Abbreviations

PLGA:

Poly(D,L-lactic-co-glycolic acid)

RP-HPLC:

Reversed phase high performance liquid chromatography

SEM:

Scanning Electron Microscopy

FTIR:

Fourier Transform Infrared Spectroscopy

SSNMR:

Solid State Nuclear Magnetic Resonance

PEG:

Polyethylene glycol

CMCS:

O-Carboxymethyl chitosan

Ca3(PO4)2 :

Calcium phosphate

CaHPO4 :

Calcium hydrophosphate

Ca(H2PO4)2 :

Calcium dihydrophosphate

CaCl2 :

Calcium chloride

Mw:

Molecular weight

PVA:

Poly (vinyl alcohol)

NaOH:

Sodium hydroxide

HCl:

Hydrochloric acid

TFA:

Trifluoroacetic acid

PBST:

Phosphate buffer solution (0.02 wt.% Tween 80)

HEPES:

2-[4-(2-Hydroxyethyl)-1-piperazinyl]ethanesulfonic acid

PLA:

Polylactic acid

PGA:

Polyglycolic acid

References

  1. Y.W. Yi, M.H. Seo, B.O. Kim, I.J. Choi, H.J. Yoon, S.Y. Kim, S.J. Lee, and J.W. Cho. Macromolecule for delivering protein, polypeptide or peptide drugs and a production method for the same, and a slow release composition for protein, polypeptide or peptide drugs and a production method for the same, Google Patents (2017).

  2. van Dijk F, Teekamp N, Beljaars L, Post E, Zuidema J, Steendam R, et al. Pharmacokinetics of a sustained release formulation of PDGFβ-receptor directed carrier proteins to target the fibrotic liver. J Control Release. 2018;269:258–65.

    Article  Google Scholar 

  3. Chen L, Mei L, Feng D, Huang D, Tong X, Pan X, et al. Anhydrous reverse micelle lecithin nanoparticles/PLGA composite microspheres for long-term protein delivery with reduced initial burst. Colloids Surf B: Biointerfaces. 2018;163:146–54.

    Article  CAS  Google Scholar 

  4. Vaishya RD, Mandal A, Gokulgandhi M, Patel S, Mitra AK. Reversible hydrophobic ion-paring complex strategy to minimize acylation of octreotide during long-term delivery from PLGA microparticles. Int J Pharm. 2015;489:237–45.

    Article  CAS  Google Scholar 

  5. Sophocleous AM, Desai KG, Mazzara JM, Tong L, Cheng JX, Olsen KF, et al. The nature of peptide interactions with acid end-group PLGAs and facile aqueous-based microencapsulation of therapeutic peptides. Journal of controlled release : official journal of the Controlled Release Society. 2013;172:662–70.

    Article  CAS  Google Scholar 

  6. Niu Z, Tedesco E, Benetti F, Mabondzo A, Montagner IM, Marigo I, et al. Rational design of polyarginine nanocapsules intended to help peptides overcoming intestinal barriers. J Control Release. 2017;263:4–17.

    Article  CAS  Google Scholar 

  7. Wong CY, Al-Salami H, Dass CR. Microparticles, microcapsules and microspheres: a review of recent developments and prospects for oral delivery of insulin. Int J Pharm. 2017.

  8. Dai C, Wang B, Zhao H. Microencapsulation peptide and protein drugs delivery system. Colloids Surf B: Biointerfaces. 2005;41:117–20.

    Article  CAS  Google Scholar 

  9. Wei G, Jin L, Xu L, Liu Y, Lu W. Preparation, characterization and in vivo pharmacodynamic evaluation of thymopentin loaded poly (lactide acid)/poly (lactide-co-glycolide acid) implants. Int J Pharm. 2010;398:123–9.

    Article  CAS  Google Scholar 

  10. Widmer J, Thauvin C, Mottas I, Nguyen VN, Delie F, Allémann E, et al. Polymer-based nanoparticles loaded with a TLR7 ligand to target the lymph node for immunostimulation. Int J Pharm. 2018;535:444–51.

    Article  CAS  Google Scholar 

  11. Le Devedec F, Boucher H, Dubins D, Allen C. Factors controlling drug release in cross-linked poly (valerolactone) based matrices. Mol Pharm. 2018;15:1565–77.

    Article  Google Scholar 

  12. Qi F, Wu J, Fan Q, He F, Tian G, Yang T, et al. Preparation of uniform-sized exenatide-loaded PLGA microspheres as long-effective release system with high encapsulation efficiency and bio-stability. Colloids Surf B: Biointerfaces. 2013;112:492–8.

    Article  CAS  Google Scholar 

  13. Pai SS, Tilton RD, Przybycien TM. Poly (ethylene glycol)-modified proteins: implications for poly (lactide-co-glycolide)-based microsphere delivery. AAPS J. 2009;11:88–98.

    Article  CAS  Google Scholar 

  14. Shpigel T, Uziel A, Lewitus DY. SPHRINT – printing drug delivery microspheres from polymeric melts. Eur J Pharm Biopharm. 2018;127:398–406.

    Article  CAS  Google Scholar 

  15. Wischkeand C, Schwendeman SP. Principles of encapsulating hydrophobic drugs in PLA/PLGA microparticles. Int J Pharm. 2008;364:298–327.

    Article  Google Scholar 

  16. Wang J, Wang BM, Schwendeman SP. Characterization of the initial burst release of a model peptide from poly (D, L-lactide-co-glycolide) microspheres. J Control Release. 2002;82:289–307.

    Article  CAS  Google Scholar 

  17. Schwendeman SP. Recent advances in the stabilization of proteins encapsulated in injectable PLGA delivery systems, vol. 19; 2002. p. 26.

    Google Scholar 

  18. Giteau A, Venier-Julienne M-C, Marchal S, Courthaudon J-L, Sergent M, Montero-Menei C, et al. Reversible protein precipitation to ensure stability during encapsulation within PLGA microspheres. Eur J Pharm Biopharm. 2008;70:127–36.

    Article  CAS  Google Scholar 

  19. He J-T, Su H-B, Li G-P, Tao X-M, Mo W, Song H-Y. Stabilization and encapsulation of a staphylokinase variant (K35R) into poly (lactic-co-glycolic acid) microspheres. Int J Pharm. 2006;309:101–8.

    Article  CAS  Google Scholar 

  20. Murty SB, Goodman J, Thanoo B, DeLuca PP. Identification of chemically modified peptide from poly (D, L-lactide-co-glycolide) microspheres under in vitro release conditions. AAPS PharmSciTech. 2003;4:392–405.

    Article  CAS  Google Scholar 

  21. Na DH, Youn YS, Lee SD, Son M-W, Kim W-B, DeLuca PP, et al. Monitoring of peptide acylation inside degrading PLGA microspheres by capillary electrophoresis and MALDI-TOF mass spectrometry. J Control Release. 2003;92:291–9.

    Article  CAS  Google Scholar 

  22. Ghassemi AH, van Steenbergen MJ, Barendregt A, Talsma H, Kok RJ, van Nostrum CF, et al. Controlled release of octreotide and assessment of peptide acylation from poly (D, L-lactide-co-hydroxymethyl glycolide) compared to PLGA microspheres. Pharm Res. 2012;29:110–20.

    Article  CAS  Google Scholar 

  23. Shirangi M, Hennink WE, Somsen GW, van Nostrum CF. Acylation of arginine in goserelin-loaded PLGA microspheres. European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik eV, vol. 99; 2016. p. 18–23.

    Google Scholar 

  24. Na DH, De Luca PP. PEGylation of octreotide: I. separation of positional isomers and stability against acylation by poly (D, L-lactide-co-glycolide). Pharm Res. 2005;22:736–42.

    Article  CAS  Google Scholar 

  25. Shirangi M, Hennink WE, Somsen GW, Van Nostrum CF. Identification and assessment of octreotide acylation in polyester microspheres by LC–MS/MS. Pharm Res. 2015;32:3044–54.

    Article  CAS  Google Scholar 

  26. Sophocleous AM, Zhang Y, Schwendeman SP. A new class of inhibitors of peptide sorption and acylation in PLGA. J Control Release. 2009;137:179–84.

    Article  CAS  Google Scholar 

  27. Shirangi M, Najafi M, Rijkers DT, Kok RJ, Hennink WE, van Nostrum CF. Inhibition of octreotide acylation inside plga microspheres by derivatization of the amines of the peptide with a self-immolative protecting group. Bioconjug Chem. 2016;27:576–85.

    Article  CAS  Google Scholar 

  28. Lucke A, Kiermaier J, Göpferich A. Peptide acylation by poly (α-hydroxy esters). Pharm Res. 2002;19:175–81.

    Article  CAS  Google Scholar 

  29. Göpferich ALA. Acylation of peptides by lactic acid solutions. Eur J Pharm Biopharm. 2003;55:27–33.

    Article  Google Scholar 

  30. Na DH, Lee KC, DeLuca PP. PEGylation of octreotide: II. Effect of N-terminal mono-PEGylation on biological activity and pharmacokinetics. Pharm Res. 2005;22:743–9.

    Article  CAS  Google Scholar 

  31. Ahn JH, Park EJ, Lee HS, Lee KC, Na DH. Reversible blocking of amino groups of octreotide for the inhibition of formation of acylated peptide impurities in poly (lactide-co-glycolide) delivery systems. AAPS PharmSciTech. 2011;12:1220–6.

    Article  CAS  Google Scholar 

  32. Zhang Y, Sophocleous AM, Schwendeman SP. Inhibition of peptide acylation in PLGA microspheres with water-soluble divalent cationic salts. Pharm Res. 2009;26:1986–94.

    Article  CAS  Google Scholar 

  33. Zhangand Y, Schwendeman SP. Minimizing acylation of peptides in PLGA microspheres. Journal of controlled release : official journal of the Controlled Release Society. 2012;162:119–26.

    Article  Google Scholar 

  34. Wright ME. Side-chain and end-group modified poly-p-phenylene oligomers. In: Google Patents2015.

  35. Wang J, Wang BM, Schwendeman SP. Mechanistic evaluation of the glucose-induced reduction in initial burst release of octreotide acetate from poly (D, L-lactide-co-glycolide) microspheres. Biomaterials. 2004;25:1919–27.

    Article  CAS  Google Scholar 

  36. Liu Y, Ghassemi AH, Hennink WE, Schwendeman SP. The microclimate pH in poly(d,l-lactide-co-hydroxymethyl glycolide) microspheres during biodegradation. Biomaterials. 2012;33:7584–93.

    Article  CAS  Google Scholar 

  37. Dingand AG, Schwendeman SP. Acidic microclimate pH distribution in PLGA microspheres monitored by confocal laser scanning microscopy. Pharm Res. 2008;25:2041–52.

    Article  Google Scholar 

  38. Liuand Y, Schwendeman SP. Mapping microclimate pH distribution inside protein-encapsulated PLGA microspheres using confocal laser scanning microscopy. Mol Pharm. 2012;9:1342–50.

    Article  Google Scholar 

  39. Loste E, Díaz-Martí E, Zarbakhsh A, Meldrum FC. Study of calcium carbonate precipitation under a series of fatty acid Langmuir monolayers using Brewster angle microscopy. Langmuir. 2003;19:2830–7.

    Article  CAS  Google Scholar 

  40. Govers MJ, Termont DS, Lapré JA, Kleibeuker JH, Vonk RJ, Van der Meer R. Calcium in milk products precipitates intestinal fatty acids and secondary bile acids and thus inhibits colonic cytotoxicity in humans. Cancer Res. 1996;56:3270–5.

    CAS  PubMed  Google Scholar 

  41. Zolnikand BS, Burgess DJ. Effect of acidic pH on PLGA microsphere degradation and release. J Control Release. 2007;122:338–44.

    Article  Google Scholar 

  42. Rawat A, Stippler E, Shah VP, Burgess DJ. Validation of USP apparatus 4 method for microsphere in vitro release testing using Risperdal® Consta®. Int J Pharm. 2011;420:198–205.

    Article  CAS  Google Scholar 

  43. Guand B, Burgess DJ. Prediction of dexamethasone release from PLGA microspheres prepared with polymer blends using a design of experiment approach. Int J Pharm. 2015;495:393–403.

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to He Huang or Xueming Li.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic Supplementary Material

ESM 1

(DOCX 1923 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, J., Xu, Y., Wang, Y. et al. Proton Oriented-“Smart Depot” for Responsive Release of Ca2+ to Inhibit Peptide Acylation in PLGA Microspheres. Pharm Res 36, 119 (2019). https://doi.org/10.1007/s11095-019-2640-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s11095-019-2640-5

KEY WORDS

Navigation