Please wait a minute...
Frontiers of Chemical Science and Engineering

ISSN 2095-0179

ISSN 2095-0187(Online)

CN 11-5981/TQ

邮发代号 80-969

2019 Impact Factor: 3.552

Frontiers of Chemical Science and Engineering  2022, Vol. 16 Issue (4): 536-545   https://doi.org/10.1007/s11705-021-2067-5
  本期目录
Long acting carmustine loaded natural extracellular matrix hydrogel for inhibition of glioblastoma recurrence after tumor resection
Sunhui Chen1, Qiujun Qiu1, Dongdong Wang2, Dejun She2, Bo Yin2, Meihong Chai3, Huining He3(), Dong Nyoung Heo4, Jianxin Wang1()
1. Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
2. Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, China
3. Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
4. Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
 全文: PDF(1039 KB)   HTML
Abstract

Many scientific efforts have been made to penetrate the blood-brain barrier and target glioblastoma cells, but the outcomes have been limited. More attention should be given to local inhibition of recurrence after glioblastoma resection to meet real medical needs. A biodegradable wafer containing the chemotherapeutics carmustine (1,3-bis(2-chloroethyl)-1-nitrosourea, BCNU) was the only local drug delivery system approved for clinical glioblastoma treatment, but with a prolonged survival time of only two months and frequent side effects. In this study, to improve the sustained release and prolonged therapeutic effect of drugs for inhibiting tumor recurrence after tumor resection, both free BCNU and BCNU- poly (lactic-co-glycolic acid) (the ratio of lactic acid groups to glycolic acid groups is 75/25) nanoparticles were simultaneously loaded into natural extracellular matrix hydrogel from pigskin to prepare BCNU gels. The hydrogel was injected into the resection cavity of a glioblastoma tumor immediately after tumor removal in a fully characterized resection rat model. Free drugs were released instantly to kill the residual tumor cells, while drugs in nanoparticles were continuously released to achieve a continuous and effective inhibition of the residual tumor cells for 30 days. These combined actions effectively restricted tumor growth in rats. Thus, this strategy of local drug implantation and delivery may provide a reliable method to inhibit the recurrence of glioblastoma after tumor resection in vivo.

Key wordsBCNU    glioblastoma recurrence    tumor resection    nanoparticles    hydrogel
收稿日期: 2021-02-08      出版日期: 2022-03-21
Corresponding Author(s): Huining He,Jianxin Wang   
 引用本文:   
. [J]. Frontiers of Chemical Science and Engineering, 2022, 16(4): 536-545.
Sunhui Chen, Qiujun Qiu, Dongdong Wang, Dejun She, Bo Yin, Meihong Chai, Huining He, Dong Nyoung Heo, Jianxin Wang. Long acting carmustine loaded natural extracellular matrix hydrogel for inhibition of glioblastoma recurrence after tumor resection. Front. Chem. Sci. Eng., 2022, 16(4): 536-545.
 链接本文:  
https://academic.hep.com.cn/fcse/CN/10.1007/s11705-021-2067-5
https://academic.hep.com.cn/fcse/CN/Y2022/V16/I4/536
Characteristic Parameter
PLGA75/25/mg 30
PVA/% 5
BCNU/mg 20
Rotation speed/(r·min−1) 250
Rotation time/h 1
Size/nm 274.5 ± 23.5
PDI 0.065
Zeta potential/mV –19.3
EE/% 32.1
Tab.1  
Fig.1  
Fig.2  
Fig.3  
Fig.4  
1 R L Siegel, K D Miller, A Jemal. Cancer statistics 2019. CA: a Cancer Journal for Clinicians, 2019, 69(1): 7–34
https://doi.org/10.3322/caac.21551
2 C Bastiancich, J Bianco, K Vanvarenberg, B Ucakar, N Joudiou, B Gallez, G Bastiat, F Lagarce, V Préat, F Danhier. Injectable nanomedicine hydrogel for local chemotherapy of glioblastoma after surgical resection. Journal of Controlled Release, 2017, 264: 45–54
https://doi.org/10.1016/j.jconrel.2017.08.019
3 R Stupp, M Brada, M J van den Bent, J C Tonn, G Pentheroudakis. High-grade glioma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Annals of Oncology: Official Journal of the European Society for Medical Oncology, 2014, 25(Suppl 3): 93–101
https://doi.org/10.1093/annonc/mdu050
4 X L Wei, X S Chen, M Ying, W Y Lu. Brain tumor-targeted drug delivery strategies. Acta Pharmaceutica Sinica. B, 2014, 4(3): 193–201
https://doi.org/10.1016/j.apsb.2014.03.001
5 Z L Chai, X F Hu, W Y Lu. Cell membrane-coated nanoparticles for tumor-targeted drug delivery. Science China Materials, 2017, 60(6): 504–510
https://doi.org/10.1007/s40843-016-5163-4
6 J K Saucier-Sawyer, Y E Seo, A Gaudin, E Quijano, E Song, A J Sawyer, Y Deng, A Huttner, W M Saltzman. Distribution of polymer nanoparticles by convection-enhanced delivery to brain tumors. Journal of Controlled Release, 2016, 232: 103–112
https://doi.org/10.1016/j.jconrel.2016.04.006
7 G Strohbehn, D Coman, L Han, R R Ragheb, T M Fahmy, A J Huttner, F Hyder, J M Piepmeier, W M Saltzman, J Zhou. Imaging the delivery of brain-penetrating PLGA nanoparticles in the brain using magnetic resonance. Journal of Neuro-Oncology, 2015, 121(3): 441–449
https://doi.org/10.1007/s11060-014-1658-0
8 C Zhang, P Mastorakos, M Sobral, S Berry, E Song, E Nance, C G Eberhart, J Hanes, J S Suk. Strategies to enhance the distribution of nanotherapeutics in the brain. Journal of Controlled Release, 2017, 267: 232–239
https://doi.org/10.1016/j.jconrel.2017.07.028
9 T Shapira-Furman, R Serra, N Gorelick, M Doglioli, V Tagliaferri, A Cecia, M Peters, A Kumar, Y Rottenberg, R Langer, H Brem, B Tyler, A J Domb. Biodegradable wafers releasing Temozolomide and Carmustine for the treatment of brain cancer. Journal of Controlled Release, 2019, 295: 93–101
https://doi.org/10.1016/j.jconrel.2018.12.048
10 M Westphal, D C Hilt, E Bortey, P Delavault, R Olivares, P C Warnke, I R Whittle, J Jaaskelainen, Z Ram. A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma. Neuro-Oncology, 2003, 5(2): 79–88
https://doi.org/10.1093/neuonc/5.2.79
11 F J Attenello, D Mukherjee, G Datoo, M J McGirt, E Bohan, J D Weingart, A Olivi, A Quinones-Hinojosa, H Brem. Use of Gliadel (BCNU) wafer in the surgical treatment of malignant glioma: a 10-year institutional experience. Annals of Surgical Oncology, 2008, 15(10): 2887–2893
https://doi.org/10.1245/s10434-008-0048-2
12 B R Subach, T F Witham, D Kondziolka, D Lunsford, M Bozik, D Schiff. Morbidity and survival after 1,3-bis(2-chloroethyl)-1-nitrosourea wafer implantation for recurrent glioblastoma: a retrospective case-matched cohort series. Neurosurgery, 1999, 45(1): 17–22
https://doi.org/10.1227/00006123-199907000-00004
13 T C Holmes, S de Lacalle, X Su, G S Liu, A Rich, S G Zhang. Extensive neurite outgrowth and active synapse formation on self-assembling peptide scaffolds. Proceedings of the National Academy of Sciences of the United States of America, 2000, 97(12): 6728–6733
https://doi.org/10.1073/pnas.97.12.6728
14 Q Chen, C Wang, X D Zhang, G Chen, Q Hu, H Li, J Wang, D Wen, Y Zhang, Y Lu, G Yang, C Jiang, J Wang, G Dotti, Z Gu. In situ sprayed bioresponsive immunotherapeutic gel for post-surgical cancer treatment. Nature Nanotechnology, 2019, 14(1): 89–97
https://doi.org/10.1038/s41565-018-0319-4
15 J Park, P S Doyle. Multifunctional hierarchically-assembled hydrogel particles with pollen grains via pickering suspension polymerization. Langmuir, 2018, 34(48): 14643–14651
https://doi.org/10.1021/acs.langmuir.8b02957
16 L M De Leon-Rodriguez, Y Hemar, G Mo, A K Mitra, J Cornish, M A Brimble. Multifunctional thermoresponsive designer peptide hydrogels. Acta Biomaterialia, 2017, 47: 40–49
https://doi.org/10.1016/j.actbio.2016.10.014
17 C Bastiancich, P Danhier, V Preat, F Danhier. Anticancer drug-loaded hydrogels as drug delivery systems for the local treatment of glioblastoma. Journal of Controlled Release, 2016, 243: 29–42
https://doi.org/10.1016/j.jconrel.2016.09.034
18 H Ghuman, A R Massensini, J Donnelly, S M Kim, C J Medberry, S F Badylak, M Modo. ECM hydrogel for the treatment of stroke: characterization of the host cell infiltrate. Biomaterials, 2016, 91: 166–181
https://doi.org/10.1016/j.biomaterials.2016.03.014
19 C J Medberry, P M Crapo, B F Siu, C A Carruthers, M T Wolf, S P Nagarkar, V Agrawal, K E Jones, J Kelly, S A Johnson, S S Velankar, S C Watkins, M Modo, S F Badylak. Hydrogels derived from central nervous system extracellular matrix. Biomaterials, 2013, 34(4): 1033–1040
https://doi.org/10.1016/j.biomaterials.2012.10.062
20 B P Chan, K W Leong. Scaffolding in tissue engineering: general approaches and tissue-specific considerations. European Spine Journal, 2008, 17(S4 Suppl 4): 467–479
https://doi.org/10.1007/s00586-008-0745-3
21 J M Zhu, R E Marchant. Design properties of hydrogel tissue-engineering scaffolds. Expert Review of Medical Devices, 2011, 8(5): 607–626
https://doi.org/10.1586/erd.11.27
22 O Sayiner, S Arisoy, T Comoglu, F G Ozbay, G Esendagli. Development and in vitro evaluation of temozolomide-loaded PLGA nanoparticles in a thermoreversible hydrogel system for local administration in glioblastoma multiforme. Journal of Drug Delivery Science and Technology, 2020, 57: 1–10
https://doi.org/10.1016/j.jddst.2020.101627
23 M Singh, A Chakrapani, D O’Hagan. Nanoparticles and microparticles as vaccine-delivery systems. Expert Review of Vaccines, 2007, 6(5): 797–808
https://doi.org/10.1586/14760584.6.5.797
24 D J McClements. Encapsulation, protection, and delivery of bioactive proteins and peptides using nanoparticle and microparticle systems: a review. Advances in Colloid and Interface Science, 2018, 253: 1–22
https://doi.org/10.1016/j.cis.2018.02.002
25 J Y Sheng, L M Han, J Qin, G Ru, R X Li, L H Wu, D Q Cui, P Yang, Y W He, J X Wang. N-Trimethyl chitosan chloride-coated PLGA nanoparticles overcoming multiple barriers to oral insulin absorption. ACS Applied Materials & Interfaces, 2015, 7(28): 15430–15441
https://doi.org/10.1021/acsami.5b03555
26 M B Yang, R J Tamargo, H Brem. Controlled delivery of 1,3-bis(2-chloroethyl)-1-nitrosourea from ethylene-vinyl acetate copolymer. Cancer Research, 1989, 49(18): 5103–5107
27 S F Badylak. The extracellar matrix as a scaffold for tissue reconstruction. Cell & Developmental Biology, 2002, 13(5): 377–383
28 K T Sheets, J R Bago, I L Paulk, S D Hingtgen. Image-guided resection of glioblastoma and intracranial implantation of therapeutic stem cell-seeded scaffolds. Jove-Journal of Visualized Experiments, 2018, 137(137): e57452
https://doi.org/10.3791/57452
29 P W Lee, J K Pokorski. Poly(lactic-co-glycolic acid) devices: production and applications for sustained protein delivery. Wiley Interdisciplinary Reviews. Nanomedicine and Nanobiotechnology, 2018, 10(8): e1516
https://doi.org/10.1002/wnan.1516
30 K E Parrish, J N Sarkaria, W F Elmquist. Improving drug delivery to primary and metastatic brain tumors: strategies to overcome the blood-brain barrier. Clinical Pharmacology and Therapeutics, 2015, 97(4): 336–346
https://doi.org/10.1002/cpt.71
31 L Hamard, D Ratel, L Selek, F Berger, B van der Sanden, D Wion. The brain tissue response to surgical injury and its possible contribution to glioma recurrence. Journal of Neuro-Oncology, 2016, 128(1): 1–8
https://doi.org/10.1007/s11060-016-2096-y
32 L Ding, Q Wang, M Shen, Y Sun, X Zhang, C Huang, J Chen, R Li, Y Duan. Thermoresponsive nanocomposite gel for local drug delivery to suppress the growth of glioma by inducing autophagy. Autophagy, 2017, 13(7): 1176–1190
https://doi.org/10.1080/15548627.2017.1320634
33 J H Sherman, G T Redpath, J A Redick, B W Purow, E R Laws, J A Jr Jane, M E Shaffrey, I M Hussaini. A novel fixative for immunofluorescence staining of CD133-positive glioblastoma stem cells. Journal of Neuroscience Methods, 2011, 198(1): 99–102
https://doi.org/10.1016/j.jneumeth.2011.03.003
34 W Taal, J E C Bromberg, M J van den Bent. Chemotherapy in glioma. CNS Oncology, 2015, 4(3): 179–192
https://doi.org/10.2217/cns.15.2
35 J Jung, M R Gilbert, E M Park. Isolation and propagation of glioma stem cells from acutely resected tumors. Methods in Molecular Biology (Clifton, N.J.), 2016, 1516: 361–369
https://doi.org/10.1007/7651_2016_342
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed