1,123
Views
1
CrossRef citations to date
0
Altmetric
Research Article

Triptorelin nanoparticle-loaded microneedles for use in assisted reproductive technology

, , , , , & show all
Article: 2226367 | Received 18 Jan 2023, Accepted 12 Jun 2023, Published online: 30 Jun 2023

References

  • Adamson G, Dyer S, Chambers G, et al. (2019). ICMART preliminary world report 2015. Hum Reprod 37:1.
  • Asmus LR, Tille JC, Kaufmann B, et al. (2013). In vivo biocompatibility, sustained-release and stability of triptorelin formulations based on a liquid, degradable polymer. J Control Release 165:199–11. doi: 10.1016/j.jconrel.2012.11.014.
  • Bethesda M. (2012). Clinical and research information on drug-induced liver injury-triptorelin. In: LiverTox. Available at: https://www.ncbi.nlm.nih.gov/books/NBK548756/.
  • Chen S, Liu M, Huang H, et al. (2019). Mechanical properties of Bombyx mori silkworm silk fibre and its corresponding silk fibroin filament: a comparative study. Mater Des 181:108077. doi: 10.1016/j.matdes.2019.108077.
  • Collado-González M, Ferreri MC, Freitas AR, et al. (2020). Complex polysaccharide-based nanocomposites for oral insulin delivery. Mar Drugs 18:55. doi: 10.3390/md18010055.
  • DeMuth PC, Moon JJ, Suh H, et al. (2012). Releasable layer-by-layer assembly of stabilized lipid nanocapsules on microneedles for enhanced transcutaneous vaccine delivery. ACS Nano 6:8041–51. doi: 10.1021/nn302639r.
  • Detloff T, Sobisch D, Lerche JDLT. (2013). Instability index. Dispers Lett Techn 4:1–4.
  • Eftekhar M, Rahmani E, Mohammadian F. (2013). Comparison of pregnancy outcome in half-dose triptorelin and short-acting decapeptyl in long protocol in ART cycles: a randomized clinical trial. Iran J Reprod Med 11:133–8.
  • Fields E,Chard J,James D,Treasure T. (2013). Fertility (update): summary of NICE guidance. BMJ 346:f650. doi: 10.1136/bmj.f650.
  • Han J, Zhang S, Liu W, et al. (2014). An analytical strategy to characterize the pharmacokinetics and pharmacodynamics of triptorelin in rats based on simultaneous LC–MS/MS analysis of triptorelin and endogenous testosterone in rat plasma. Anal Bioanal Chem 406:2457–65. doi: 10.1007/s00216-014-7616-z.
  • He M, Yang G, Zhang S, et al. (2018). Dissolving microneedles loaded with etonogestrel microcrystal particles for intradermal sustained delivery. J Pharm Sci 107:1037–45. doi: 10.1016/j.xphs.2017.11.013.
  • Hines DJ, Kaplan DL. (2011). Mechanisms of controlled release from silk fibroin films. Biomacromolecules 12:804–12. doi: 10.1021/bm101421r.
  • Holland C, Numata K, Rnjak-Kovacina J, Seib FP. (2019). The biomedical use of silk: past, present, future. Adv Healthc Mater 8:e1800465. doi: 10.1002/adhm.201800465.
  • Hu X, Kaplan D, Cebe P. (2006). Determining beta-sheet crystallinity in fibrous proteins by thermal analysis and infrared spectroscopy. Macromolecules 39:6161–70. doi: 10.1021/ma0610109.
  • Huang W, Ling S, Li C, et al. (2018). Silkworm silk-based materials and devices generated using bio-nanotechnology. Chem Soc Rev 47:6486–504. doi: 10.1039/c8cs00187a.
  • Ingrole RSJ, Azizoglu E, Dul M, et al. (2021). Trends of microneedle technology in the scientific literature, patents, clinical trials and internet activity. Biomaterials 267:120491. doi: 10.1016/j.biomaterials.2020.120491.
  • Ito Y, Murano H, Hamasaki N, et al. (2011). Incidence of low bioavailability of leuprolide acetate after percutaneous administration to rats by dissolving microneedles. Int J Pharm 407:126–31. doi: 10.1016/j.ijpharm.2011.01.039.
  • Kundu B, Rajkhowa R, Kundu SC, Wang X. (2013). Silk fibroin biomaterials for tissue regenerations. Adv Drug Deliv Rev 65:457–70. doi: 10.1016/j.addr.2012.09.043.
  • Lammel AS, Hu X, Park SH, et al. (2010). Controlling silk fibroin particle features for drug delivery. Biomaterials 31:4583–91. doi: 10.1016/j.biomaterials.2010.02.024.
  • Lan X, She J, Lin DA, et al. (2018). Microneedle-mediated delivery of lipid-coated cisplatin nanoparticles for efficient and safe cancer therapy. ACS Appl Mater Interfaces 10:33060–9. doi: 10.1021/acsami.8b12926.
  • Larrañeta E, Lutton REM, Woolfson AD, Donnelly RF. (2016). Microneedle arrays as transdermal and intradermal drug delivery systems: materials science, manufacture and commercial development. Mater Sci Eng R Rep 104:1–32. doi: 10.1016/j.mser.2016.03.001.
  • Lin S, Quan G, Hou A, et al. (2019). Strategy for hypertrophic scar therapy: improved delivery of triamcinolone acetonide using mechanically robust tip-concentrated dissolving microneedle array. J Control Release 306:69–82. doi: 10.1016/j.jconrel.2019.05.038.
  • Lu X, Sun Y, Han M, et al. (2022). Silk fibroin double-layer microneedles for the encapsulation and controlled release of triptorelin. Int J Pharm 613:121433. doi: 10.1016/j.ijpharm.2021.121433.
  • Nguyen TP, Nguyen QV, Nguyen VH, et al. (2019). Silk fibroin-based biomaterials for biomedical applications: a review. Polymers 11:1933. doi: 10.3390/polym11121933.
  • Numata K, Kaplan DL. (2010). Silk-based delivery systems of bioactive molecules. Adv Drug Deliv Rev 62:1497–508. doi: 10.1016/j.addr.2010.03.009.
  • Permana AD, Tekko IA, McCrudden MTC, et al. (2019). Solid lipid nanoparticle-based dissolving microneedles: a promising intradermal lymph targeting drug delivery system with potential for enhanced treatment of lymphatic filariasis. J Control Release 316:34–52. doi: 10.1016/j.jconrel.2019.10.004.
  • Pham DT, Tiyaboonchai W. (2020). Fibroin nanoparticles: a promising drug delivery system. Drug Deliv 27:431–48. doi: 10.1080/10717544.2020.1736208.
  • Qi Z, Cao J, Tao X, et al. (2021). Silk fibroin microneedle patches for the treatment of insomnia. Pharmaceutics 13:2198. doi: 10.3390/pharmaceutics13122198.
  • Qiu Y, Li C, Zhang S, et al. (2016). Systemic delivery of artemether by dissolving microneedles. Int J Pharm 508:1–9. doi: 10.1016/j.ijpharm.2016.05.006.
  • Raja WK, MacCorkle S, Diwan IM, et al. (2013). Transdermal delivery devices: fabrication, mechanics and drug release from silk. Small 9:3704–13. doi: 10.1002/smll.201202075.
  • Sabri AH, Kim Y, Marlow M, et al. (2020). Intradermal and transdermal drug delivery using microneedles – fabrication, performance evaluation and application to lymphatic delivery. Adv Drug Deliv Rev 153:195–215. doi: 10.1016/j.addr.2019.10.004.
  • Singh P, Carrier A, Chen Y, et al. (2019). Polymeric microneedles for controlled transdermal drug delivery. J Control Release 315:97–113. doi: 10.1016/j.jconrel.2019.10.022.
  • Siristatidis CS, Gibreel A, Basios G, et al. (2015). Gonadotrophin-releasing hormone agonist protocols for pituitary suppression in assisted reproduction. Cochrane Database Syst Rev 2015:CD006919.
  • Suchaoin W, Bernkop-Schnürch A. (2017). Nanocarriers protecting toward an intestinal pre-uptake metabolism. Nanomedicine 12:255–69. doi: 10.2217/nnm-2016-0331.
  • Ulloa Rojas JE, Oliveira VL, de Araujo DR, et al. (2022). Silk fibroin/poly(vinyl alcohol) microneedles as carriers for the delivery of singlet oxygen photosensitizers. ACS Biomater Sci Eng 8:128–39. doi: 10.1021/acsbiomaterials.1c00913.
  • Varamini P, Rafiee A, Giddam AK, et al. (2017). Development of new gonadotropin-releasing hormone-modified dendrimer platforms with direct antiproliferative and gonadotropin releasing activity. J Med Chem 60:8309–20. doi: 10.1021/acs.jmedchem.6b01771.
  • Wang C, Ye Y, Hochu GM, et al. (2016). Enhanced cancer immunotherapy by microneedle patch-assisted delivery of anti-PD1 antibody. Nano Lett 16:2334–40. doi: 10.1021/acs.nanolett.5b05030.
  • Wang Z, Yang Z, Jiang J, et al. (2022). Silk microneedle patch capable of on-demand multidrug delivery to the brain for glioblastoma treatment. Adv Mater 34:e2106606. doi: 10.1002/adma.202106606.
  • WHO. (2020). Infertility. Available at: https://www.who.int/news-room/fact-sheets/detail/infertility.
  • Yavuz B, Chambre L, Harrington K, et al. (2020). Silk fibroin microneedle patches for the sustained release of levonorgestrel. ACS Appl Bio Mater 3:5375–82. doi: 10.1021/acsabm.0c00671.
  • Yin Z, Kuang D, Wang S, et al. (2018). Swellable silk fibroin microneedles for transdermal drug delivery. Int J Biol Macromol 106:48–56. doi: 10.1016/j.ijbiomac.2017.07.178.
  • Yu J, Zhang Y, Ye Y, et al. (2015). Microneedle-array patches loaded with hypoxia-sensitive vesicles provide fast glucose-responsive insulin delivery. Proc Natl Acad Sci U S A 112:8260–5. doi: 10.1073/pnas.1505405112.
  • Zhang H, Lai L, Wang Y, et al. (2019). Silk fibroin for CpG oligodeoxynucleotide delivery. ACS Biomater Sci Eng 5:6082–8. doi: 10.1021/acsbiomaterials.9b01413.
  • Zielińska A, Martins-Gomes C, Ferreira NR, et al. (2018). Anti-inflammatory and anti-cancer activity of citral: optimization of citral-loaded solid lipid nanoparticles (SLN) using experimental factorial design and LUMiSizer®. Int J Pharm 553:428–40. doi: 10.1016/j.ijpharm.2018.10.065.