348
Views
65
CrossRef citations to date
0
Altmetric
Original Research

Co-Delivery of Curcumin and Paclitaxel by “Core-Shell” Targeting Amphiphilic Copolymer to Reverse Resistance in the Treatment of Ovarian Cancer

, , , , , , ORCID Icon, , ORCID Icon & show all
Pages 9453-9467 | Published online: 02 Dec 2019

References

  • Perets R, Drapkin R. It’s totally tubular ….riding the new wave of ovarian cancer research. Cancer Res. 2016;76(1):10–17. doi:10.1158/0008-5472.CAN-15-138226669862
  • Vallius T, Hynninen J, Kemppainen J, et al. (18)F-FDG-PET/CT based total metabolic tumor volume change during neoadjuvant chemotherapy predicts outcome in advanced epithelial ovarian cancer. Eur J Nucl Med Mol Imaging. 2018;45(7):1224–1232. doi:10.1007/s00259-018-3961-z29476227
  • Ju X, Yu H, Liang D, et al. LDR reverses DDP resistance in ovarian cancer cells by affecting ERCC-1, Bcl-2, Survivin and Caspase-3 expressions. Biomed Pharmacother. 2018;102:549–554. doi:10.1016/j.biopha.2018.03.09229597088
  • Peng S, Li Z, Zou L, Liu W, Liu C, McClements DJ. Improving curcumin solubility and bioavailability by encapsulation in saponin-coated curcumin nanoparticles prepared using a simple pH-driven loading method. Food Funct. 2018;9(3):1829–1839. doi:10.1039/C7FO01814B29517797
  • Li W, Jiang Z, Xiao X, et al. Curcumin inhibits superoxide dismutase-induced epithelial-to-mesenchymal transition via the PI3K/Akt/NF-kappaB pathway in pancreatic cancer cells. Int J Oncol. 2018;52(5):1593–1602.
  • Sathe G, Pinto SM, Syed N, et al. Phosphotyrosine profiling of curcumin-induced signaling. Clin Proteomics. 2016;13:13. doi:10.1186/s12014-016-9114-027307780
  • Cheng Y, Zhao P, Wu S, et al. Cisplatin and curcumin co-loaded nano-liposomes for the treatment of hepatocellular carcinoma. Int J Pharm. 2018;545(1–2):261–273. doi:10.1016/j.ijpharm.2018.05.00729730175
  • Ma L, Zhang X, Wang Z, et al. Anti-cancer effects of curcumin on myelodysplastic syndrome through the inhibition of enhancer of zeste homolog-2 (EZH2). Curr Cancer Drug Targets. 2019. doi:10.2174/1568009619666190212121735
  • Momtazi-Borojeni AA, Mosafer J, Nikfar B, et al. Curcumin in advancing treatment for gynecological cancers with developed drug- and radiotherapy-associated resistance. Rev Physiol Biochem Pharmacol. 2018;176:107–129.
  • Tomeh MA, Hadianamrei R, Zhao X. A review of curcumin and its derivatives as anticancer agents. Int J Mol Sci. 2019;20(5). doi:10.3390/ijms20051033
  • Kesharwani SS, Ahmad R, Bakkari MA, et al. Site-directed non-covalent polymer-drug complexes for inflammatory bowel disease (IBD): formulation development, characterization and pharmacological evaluation. J Control Release. 2018;290:165–179. doi:10.1016/j.jconrel.2018.08.00430142410
  • Kumar S, Kesharwani SS, Mathur H, Tyagi M, Bhat GJ, Tummala H. Molecular complexation of curcumin with pH sensitive cationic copolymer enhances the aqueous solubility, stability and bioavailability of curcumin. Eur J Pharm Sci. 2016;82:86–96. doi:10.1016/j.ejps.2015.11.01026588875
  • Kesharwani SS, Kaur S, Tummala H, Sangamwar AT. Overcoming multiple drug resistance in cancer using polymeric micelles. Expert Opin Drug Deliv. 2018;15(11):1127–1142. doi:10.1080/17425247.2018.153726130324813
  • Zhang M, Hagan CTT, Min Y, et al. Nanoparticle co-delivery of wortmannin and cisplatin synergistically enhances chemoradiotherapy and reverses platinum resistance in ovarian cancer models. Biomaterials. 2018;169:1–10. doi:10.1016/j.biomaterials.2018.03.05529631163
  • Kesharwani SS, Kaur S, Tummala H, Sangamwar AT. Multifunctional approaches utilizing polymeric micelles to circumvent multidrug resistant tumors. Colloids Surf B Biointerfaces. 2019;173:581–590. doi:10.1016/j.colsurfb.2018.10.02230352379
  • Zhao MD, Sun YM, Fu GF, et al. Gene therapy of endometriosis introduced by polymeric micelles with glycolipid-like structure. Biomaterials. 2012;33(2):634–643. doi:10.1016/j.biomaterials.2011.09.07721996531
  • Tjhay F, Motohara T, Tayama S, et al. CD44 variant 6 is correlated with peritoneal dissemination and poor prognosis in patients with advanced epithelial ovarian cancer. Cancer Sci. 2015;106(10):1421–1428. doi:10.1111/cas.1276526250934
  • Wang J, He H, Xu X, Wang X, Chen Y, Yin L. Far-red light-mediated programmable anti-cancer gene delivery in cooperation with photodynamic therapy. Biomaterials. 2018;171:72–82. doi:10.1016/j.biomaterials.2018.04.02029680675
  • Zhao MD, Hu FQ, Du YZ, et al. Coadministration of glycolipid-like micelles loading cytotoxic drug with different action site for efficient cancer chemotherapy. Nanotechnology. 2009;20(5):055102. doi:10.1088/0957-4484/20/5/05510219417333
  • Wang XJ, Peng CH, Zhang S, et al. Polysialic-acid-based micelles promote neural regeneration in spinal cord injury therapy. Nano Lett. 2019;19(2):829–838. doi:10.1021/acs.nanolett.8b0402030605619
  • Palma DA, Nguyen TK, Louie AV, et al. Measuring the integration of stereotactic ablative radiotherapy plus surgery for early-stage non-small cell lung cancer: a phase 2 clinical trial. JAMA ONCOL. 2019;5:681. doi:10.1001/jamaoncol.2018.699330789648
  • Zehra B, Ahmed A, Sarwar R, et al. Apoptotic and antimetastatic activities of betulin isolated from quercus incana against non-small cell lung cancer cells. Cancer Manag Res. 2019;11:1667–1683. doi:10.2147/CMAR.S18695630863176
  • Shi SJ, Wang LJ, Han DH, et al. Therapeutic effects of human monoclonal PSMA antibody-mediated TRIM24 siRNA delivery in PSMA-positive castration-resistant prostate cancer. Theranostics. 2019;9(5):1247–1263. doi:10.7150/thno.2988430867828
  • Wu D, Zhang Y, Xu X, et al. RGD/TAT-functionalized chitosan-graft-PEI-PEG gene nanovector for sustained delivery of NT-3 for potential application in neural regeneration. Acta Biomater. 2018;72:266–277. doi:10.1016/j.actbio.2018.03.03029578088
  • Andrade S, Ramalho MJ, Pereira MDC, Loureiro JA. Resveratrol brain delivery for neurological disorders prevention and treatment. Front Pharmacol. 2018;9:1261. doi:10.3389/fphar.2018.0126130524273
  • Que X, Su J, Guo P, et al. Study on preparation, characterization and multidrug resistance reversal of red blood cell membrane-camouflaged tetrandrine-loaded PLGA nanoparticles. Drug Deliv. 2019;26(1):199–207. doi:10.1080/10717544.2019.157386130835586
  • Alsaab HO, Sau S, Alzhrani RM, et al. Tumor hypoxia directed multimodal nanotherapy for overcoming drug resistance in renal cell carcinoma and reprogramming macrophages. Biomaterials. 2018;183:280–294. doi:10.1016/j.biomaterials.2018.08.05330179778
  • Li N, Liu YY, Li Y, et al. Fine tuning of emission behavior, self-assembly, anion sensing, and mitochondria targeting of pyridinium-functionalized tetraphenylethene by alkyl chain engineering. ACS Appl Mater Interfaces. 2018;10(28):24249–24257. doi:10.1021/acsami.8b0411329939714
  • Wang W, Zhang K, Chen D. From tunable DNA/polymer self-assembly to tailorable and morphologically pure core-shell nanofibers. Langmuir. 2018;34(50):15350–15359. doi:10.1021/acs.langmuir.8b0299230427695
  • Dong Q, Zhang H, Han Y, et al. Tumor environment differentiated “nanodepot” programmed for site-specific drug shuttling and combinative therapy on metastatic cancer. J Control Release. 2018;283:59–75. doi:10.1016/j.jconrel.2018.05.02729803613
  • Kalyane D, Raval N, Maheshwari R, Tambe V, Kalia K, Tekade RK. Employment of enhanced permeability and retention effect (EPR): nanoparticle-based precision tools for targeting of therapeutic and diagnostic agent in cancer. Mater Sci Eng C Mater Biol Appl. 2019;98:1252–1276. doi:10.1016/j.msec.2019.01.06630813007
  • Liang C, Chao Y, Yi X, et al. Nanoparticle-mediated internal radioisotope therapy to locally increase the tumor vasculature permeability for synergistically improved cancer therapies. Biomaterials. 2019;197:368–379. doi:10.1016/j.biomaterials.2019.01.03330703742
  • Wang Q, Zhong Y, Liu W, et al. Enhanced chemotherapeutic efficacy of the low-dose doxorubicin in breast cancer via nanoparticle delivery system crosslinked hyaluronic acid. Drug Deliv. 2019;26(1):12–22. doi:10.1080/10717544.2018.150705730691317
  • Carvalho AM, Teixeira R, Novoa-Carballal R, Pires RA, Reis RL, Pashkuleva I. Redox-responsive micellar nanoparticles from glycosaminoglycans for CD44 targeted drug delivery. Biomacromolecules. 2018;19(7):2991–2999. doi:10.1021/acs.biomac.8b0056129758159
  • Hossaini Nasr S, Tonson A, El-Dakdouki MH, et al. Effects of nanoprobe morphology on cellular binding and inflammatory responses: hyaluronan-conjugated magnetic nanoworms for magnetic resonance imaging of atherosclerotic plaques. ACS Appl Mater Interfaces. 2018;10(14):11495–11507. doi:10.1021/acsami.7b1970829558108
  • Hu L, Li M, Zhang Z, Shen Y, Guo S. Self-assembly of biotinylated poly(ethylene glycol)-poly(curcumin) for paclitaxel delivery. Int J Pharm. 2018;553(1–2):510–521. doi:10.1016/j.ijpharm.2018.10.01930308274
  • Fratantonio D, Molonia MS, Bashllari R, et al. Curcumin potentiates the antitumor activity of Paclitaxel in rat glioma C6 cells, Phytomedicine: international. Phytomedicine. 2018;55:23–30. doi:10.1016/j.phymed.2018.08.00930668434
  • Alemi A, Zavar Reza J, Haghiralsadat F, et al. Paclitaxel and curcumin coadministration in novel cationic PEGylated niosomal formulations exhibit enhanced synergistic antitumor efficacy. J Nanobiotechnology. 2018;16(1):28. doi:10.1186/s12951-018-0351-429571289
  • Yang Z, Sun N, Cheng R, et al. pH multistage responsive micellar system with charge-switch and PEG layer detachment for co-delivery of paclitaxel and curcumin to synergistically eliminate breast cancer stem cells. Biomaterials. 2017;147:53–67. doi:10.1016/j.biomaterials.2017.09.01328930649
  • Yao Q, Gutierrez DC, Hoang NH, et al. Efficient codelivery of paclitaxel and curcumin by novel bottlebrush copolymer-based micelles. Mol Pharm. 2017;14(7):2378–2389. doi:10.1021/acs.molpharmaceut.7b0027828605595
  • Calaf GM, Ponce-Cusi R, Carrion F. Curcumin and paclitaxel induce cell death in breast cancer cell lines. Oncol Rep. 2018;40(4):2381–2388. doi:10.3892/or.2018.660330066930
  • Gawde KA, Sau S, Tatiparti K, et al. Paclitaxel and di-fluorinated curcumin loaded in albumin nanoparticles for targeted synergistic combination therapy of ovarian and cervical cancers. Colloids Surf B Biointerfaces. 2018;167:8–19. doi:10.1016/j.colsurfb.2018.03.04629625422
  • Zhao MD, Cheng JL, Yan JJ, et al. Hyaluronic acid reagent functional chitosan-PEI conjugate with AQP2-siRNA suppressed endometriotic lesion formation. Int J Nanomedicine. 2016;11:1323–1336. doi:10.2147/IJN.S9969227099493
  • Biswas P, Mukhopadhyay A, Kabir SN, Mukhopadhyay PK. High-protein diet ameliorates arsenic-induced oxidative stress and antagonizes uterine apoptosis in rats. Biol Trace Elem Res. 2019;192:222–233. doi:10.1007/s12011-019-1657-230723882
  • Camozzato GC, Martinez MN, Bastos HBA, et al. Ultrastructural and histological characteristics of the endometrium during early embryo development in mares. Theriogenology. 2019;123:1–10. doi:10.1016/j.theriogenology.2018.09.01830253251
  • Zhou J, Li M, Lim WQ, et al. A transferrin-conjugated hollow nanoplatform for redox-controlled and targeted chemotherapy of tumor with reduced inflammatory reactions. Theranostics. 2018;8(2):518–532. doi:10.7150/thno.2119429290824
  • Zou H, Zhu J, Huang DS. Cell membrane capsule: a novel natural tool for antitumour drug delivery. Expert Opin Drug Deliv. 2019;16(3):1–19.30558447
  • Tang X, Wang G, Shi R, et al. Enhanced tolerance and antitumor efficacy by docetaxel-loaded albumin nanoparticles. Drug Deliv. 2016;23(8):2686–2696. doi:10.3109/10717544.2015.104972026004129
  • Hattinger CM, Patrizio MP, Luppi S, Magagnoli F, Picci P, Serra M. Current understanding of pharmacogenetic implications of DNA damaging drugs used in osteosarcoma treatment. Expert Opin Drug Metab Toxicol. 2019;15:299–311. doi:10.1080/17425255.2019.158888530822170