Generic placeholder image

Pharmaceutical Nanotechnology

Editor-in-Chief

ISSN (Print): 2211-7385
ISSN (Online): 2211-7393

Review Article

Co-Delivery Nanosystems for Cancer Treatment: A Review

Author(s): Reza Baradaran Eftekhari, Niloufar Maghsoudnia, Shabnam Samimi, Ali Zamzami and Farid Abedin Dorkoosh*

Volume 7, Issue 2, 2019

Page: [90 - 112] Pages: 23

DOI: 10.2174/2211738507666190321112237

Abstract

Massive data available on cancer therapy more than ever lead our mind to the general concept that there is no perfect treatment for cancer. Indeed, the biological complexity of this disease is too excessive to be treated by a single therapeutic approach. Current delivery systems containing a specific drug or gene have their particular opportunities and restrictions. It is worth noting that a considerable number of studies suggest that single- drug delivery systems result in insufficient suppression of cancer growth. Therefore, one of the main ideas of co-delivery system designing is to enhance the intended response or to achieve the synergistic/combined effect compared to the single drug strategy. This review focuses on various strategies for co-delivery of therapeutic agents in the treatment of cancer. The primary approaches within the script are categorized into co-delivery of conventional chemotherapeutics, gene-based molecules, and plant-derived materials. Each one is explained in examples with the recent researches. In the end, a brief summary is provided to conclude the gist of the review.

Keywords: Anti-cancer materials, cancer therapy, co-delivery, drug-drug, drug-gene, gene-gene, therapeutic agents.

Graphical Abstract
[1]
Lin RK, Wu CY, Chang JW, et al. Dysregulation of p53/Sp1 control leads to DNA methyltransferase-1 overexpression in lung cancer. Cancer Res 2010; 70(14): 5807-17.
[2]
Rhodes DR, Barrette TR, Rubin MA, Ghosh D, Chinnaiyan AM. Meta-analysis of microarrays: interstudy validation of gene expression profiles reveals pathway dysregulation in prostate cancer. Cancer Res 2002; 62(15): 4427-33.
[3]
Chabner BA, Roberts TG Jr. Timeline: chemotherapy and the war on cancer. Nat Rev Cancer 2005; 5(1): 65-72.
[4]
Yap TA, Carden CP, Kaye SB. Beyond chemotherapy: targeted therapies in ovarian cancer. Nat Rev Cancer 2009; 9(3): 167-81.
[5]
McCormick F. Cancer gene therapy: fringe or cutting edge? Nat Rev Cancer 2001; 1(2): 130-41.
[6]
Wolf JK, Jenkins AD. Gene therapy for ovarian cancer. Int J Oncol 2002; 21(1): 461-8.
[7]
Hammond SM. MicroRNAs as tumor suppressors. Nat Genet 2007; 39(5): 582-3.
[8]
Stagos D, Amoutzias GD, Matakos A, Spyrou A, Tsatsakis AM, Kouretas D. Chemoprevention of liver cancer by plant polyphenols. Food Chem Toxicol 2012; 50(6): 2155-70.
[9]
Pujol M, Gavilondo J, Ayala M, Rodríguez M, González EM, Pérez L. Fighting cancer with plant-expressed pharmaceuticals. Trends Biotechnol 2007; 25(10): 455-9.
[10]
Cragg GM, Newman DJ. Plants as a source of anti-cancer agents. J Ethnopharmacol 2005; 100(1-2): 72-9.
[11]
Jamil A, Aamir MM, Anwer MK, et al. Co-delivery of gemcitabine and simvastatin through PLGA polymeric nanoparticles for the treatment of pancreatic cancer: in-vitro characterization, cellular uptake and pharmacokinetic studies. Drug Dev Ind Pharm 2019; 45(5): 1-34.
[12]
Madani F, Esnaashari SS, Mujokoro B, Dorkoosh F, Khosravani M, Adabi M. Investigation of effective parameters on size of paclitaxel loaded PLGA nanoparticles. Adv Pharm Bull 2018; 8(1): 77-84.
[13]
Varshosaz J, Emami J, Ahmadi F, et al. Preparation of budesonide-dextran conjugates using glutarate spacer as a colon-targeted drug delivery system: in vitro/in vivo evaluation in induced ulcerative colitis. J Drug Target 2011; 19(2): 140-53.
[14]
Jafary ON, Bahari JN, Dehpour AR, Partoazar A, Rafiee TM, Dorkoosh F. In-vitro and in-vivo cytotoxicity and efficacy evaluation of novel glycyl-glycine and alanyl-alanine conjugates of chitosan and trimethyl chitosan nano-particles as carriers for oral insulin delivery. Int J Pharm 2018; 535(1-2): 293-307.
[15]
Sadeghi AM, Dorkoosh FA, Avadi MR, Saadat P, Rafiee-Tehrani M, Junginger HE. Preparation, characterization and antibacterial activities of chitosan, N-trimethyl chitosan (TMC) and N-diethylmethyl chitosan (DEMC) nanoparticles loaded with insulin using both the ionotropic gelation and polyelectrolyte complexation methods. Int J Pharm 2008; 355(1-2): 299-306.
[16]
Frei E III, Karon M, Levin RH, et al. The effectiveness of combinations of antileukemic agents in inducing and maintaining remission in children with acute leukemia. Blood 1965; 26(5): 642-56.
[17]
Li C, Gao Y, Li Y, Ding D. TUG1 mediates methotrexate resistance in colorectal cancer via miR-186/CPEB2 axis. Biochem Biophys Res Commun 2017; 491(2): 552-7.
[18]
Meredith AM, Dass CR. Increasing role of the cancer chemotherapeutic doxorubicin in cellular metabolism. J Pharm Pharmacol 2016; 68(6): 729-41.
[19]
Moghimipour E, Rezaei M, Ramezani Z, et al. Transferrin targeted liposomal 5-fluorouracil induced apoptosis via mitochondria signaling pathway in cancer cells. Life Sci 2018; 194: 104-10.
[20]
Giampieri R, Restivo A, Pusceddu V, et al. The role of aspirin as antitumoral agent for heavily pretreated patients with metastatic colorectal cancer receiving capecitabine monotherapy. Clin Colorectal Cancer 2017; 16(1): 38-43.
[21]
El Sayed YM, Sadée W. Metabolic activation of R,S-1-(tetrahydro-2-furanyl)-5-fluorouracil (ftorafur) to 5-fluorouracil by soluble enzymes. Cancer Res 1983; 43(9): 4039-44.
[22]
Vicario A, Sergo V, Toffoli G, Bonifacio A. Surface-enhanced Raman spectroscopy of the anti-cancer drug irinotecan in presence of human serum albumin. Colloids Surf B Biointerfaces 2015; 127: 41-6.
[23]
Wang D, Lippard SJ. Cellular processing of platinum anticancer drugs. Nat Rev Drug Discov 2005; 4(4): 307-20.
[24]
McLeod HL, Cassidy J, Powrie RH, et al. Pharmacokinetic and pharmacodynamic evaluation of the glycinamide ribonucleotide formyltransferase inhibitor AG2034. Clin Cancer Res 2000; 6(7): 2677-84.
[25]
Vallet S, Palumbo A, Raje N, Boccadoro M, Anderson KC. Thalidomide and lenalidomide: mechanism-based potential drug combinations. Leuk Lymphoma 2008; 49(7): 1238-45.
[26]
Gelman JS, Sironi J, Berezniuk I, et al. Alterations of the intracellular peptidome in response to the proteasome inhibitor bortezomib. PLoS One 2013; 8(1): 53263.
[27]
Jordan MA. Mechanism of action of antitumor drugs that interact with microtubules and tubulin. Curr Med Chem Anticancer Agents 2002; 2(1): 1-17.
[28]
Franco MS, Roque MC, de Barros ALB, de Oliveira Silva J, Cassali GD, Oliveira MC. Investigation of the antitumor activity and toxicity of long-circulating and fusogenic liposomes co-encapsulating paclitaxel and doxorubicin in a murine breast cancer animal model. Biomed Pharmacother 2019; 109: 1728-39.
[29]
Duong HH, Yung LY. Synergistic co-delivery of doxorubicin and paclitaxel using multi-functional micelles for cancer treatment. Int J Pharm 2013; 454(1): 486-95.
[30]
Wan X, Beaudoin JJ, Vinod N, et al. Co-delivery of paclitaxel and cisplatin in poly(2-oxazoline) polymeric micelles: implications for drug loading, release, pharmacokinetics and outcome of ovarian and breast cancer treatments. Biomaterials 2019; 192: 1-14.
[31]
Mishra P, Dey RK. Co-delivery of docetaxel and doxorubicin using biodegradable PEG-PLA micelles for treatment of breast cancer with synergistic anti-tumour effects. J Macromol Sci A 2018; 55(3): 310-6.
[32]
Lv S, Tang Z, Li M, et al. Co-delivery of doxorubicin and paclitaxel by PEG-polypeptide nanovehicle for the treatment of non-small cell lung cancer. Biomaterials 2014; 35(23): 6118-29.
[33]
Guo X. Co-delivery of resveratrol and docetaxel via polymeric micelles to improve the treatment of drug-resistant tumors. Asian J Pharm 2018; 14(1): 78-85.
[34]
Dorkoosh FA, Setyaningsih D, Borchard G, Rafiee-Tehrani M, Verhoef JC, Junginger HE. Effects of superporous hydrogels on paracellular drug permeability and cytotoxicity studies in Caco-2 cell monolayers. Int J Pharm 2002; 241(1): 35-45.
[35]
Seo SH, Han HD, Noh KH, Kim TW, Son SW. Chitosan hydrogel containing GMCSF and a cancer drug exerts synergistic anti-tumor effects via the induction of CD8+ T cell-mediated anti-tumor immunity. Clin Exp Metastasis 2009; 26(3): 179-87.
[36]
Qin M, Lee YE, Ray A, Kopelman R. Overcoming cancer multidrug resistance by codelivery of doxorubicin and verapamil with hydrogel nanoparticles. Macromol Biosci 2014; 14(8): 1106-15.
[37]
Elzoghby AO, Mostafa SK, Helmy MW, ElDemellawy MA, Sheweita SA. Multi-reservoir phospholipid shell encapsulating protamine nanocapsules for co-delivery of letrozole and celecoxib in breast cancer therapy. Pharm Res 2017; 34(9): 1956-69.
[38]
Wu H, Jin H, Wang C, et al. Synergistic cisplatin/doxorubicin combination chemotherapy for multidrug-resistant cancer via polymeric nanogels targeting delivery. ACS Appl Mater Interfaces 2017; 9(11): 9426-36.
[39]
Tian J, Min Y, Rodgers Z, et al. Co-delivery of paclitaxel and cisplatin with biocompatible PLGA-PEG nanoparticles enhances chemoradiotherapy in non-small cell lung cancer models. J Mater Chem B Mater Biol Med 2017; 5(30): 6049-57.
[40]
Liu B, Han L, Liu J, Han S, Chen Z, Jiang L. Co-delivery of paclitaxel and TOS-cisplatin via TAT-targeted solid lipid nanoparticles with synergistic antitumor activity against cervical cancer. Int J Nanomedicine 2017; 12: 955-68.
[41]
Yang J, Ju Z, Dong S. Cisplatin and paclitaxel co-delivered by folate-decorated lipid carriers for the treatment of head and neck cancer. Drug Deliv 2016; 24(1): 792-9.
[42]
Eloy JO, Petrilli R, Chesca DL, Saggioro FP, Lee RJ, Marchetti JM. Anti-HER2 immunoliposomes for co-delivery of paclitaxel and rapamycin for breast cancer therapy. Eur J Pharm Biopharm 2017; 115: 159-67.
[43]
Chen Y, Cheng Y, Zhao P, et al. Co-delivery of doxorubicin and imatinib by pH sensitive cleavable PEGylated nanoliposomes with folate-mediated targeting to overcome multidrug resistance. Int J Pharm 2018; 542(1-2): 266-79.
[44]
Kushwah V, Katiyar SS, Dora CP, et al. Co-delivery of docetaxel and gemcitabine by anacardic acid modified self-assembled albumin nanoparticles for effective breast cancer management. Acta Biomater 2018; 73: 424-36.
[45]
Guo Y, He W, Yang S, Zhao D, Li Z, Luan Y. Co-delivery of docetaxel and verapamil by reduction-sensitive PEG-PLGA-SS-DTX conjugate micelles to reverse the multi-drug resistance of breast cancer. Colloids Surf B Biointerfaces 2017; 151: 119-27.
[46]
Dehghankelishadi P, Saadat E, Ravar F, et al. In vitro and in vivo evaluation of paclitaxel-lapatinib-loaded F127 pluronic micelles. Drug Dev Ind Pharm 2017; 43(3): 390-8.
[47]
Mo J, Wang L, Huang X, et al. Multifunctional nanoparticles for co-delivery of paclitaxel and carboplatin against ovarian cancer by inactivating the JMJD3-HER2 axis. Nanoscale 2017; 9(35): 13142-52.
[48]
Nie J, Cheng W, Peng Y, et al. Co-delivery of docetaxel and bortezomib based on a targeting nanoplatform for enhancing cancer chemotherapy effects. Drug Deliv 2017; 24(1): 1124-38.
[49]
Yin Y, Hu Q, Xu C, et al. Co-delivery of doxorubicin and interferon-γ by thermosensitive nanoparticles for cancer immunochemotherapy. Mol Pharm 2018; 15(9): 4161-72.
[50]
Jeannot V, Gauche C, Mazzaferro S, et al. Anti-tumor efficacy of hyaluronan-based nanoparticles for the co-delivery of drugs in lung cancer. J Control Release 2018; 275: 117-28.
[51]
Yang M, Ding H, Zhu Y, Ge Y, Li L. Co-delivery of paclitaxel and doxorubicin using mixed micelles based on the redox sensitive prodrugs. Colloids Surf B Biointerfaces 2019; 175: 126-35.
[52]
Lakkadwala S, Singh J. Co-delivery of doxorubicin and erlotinib through liposomal nanoparticles for glioblastoma tumor regression using an in vitro brain tumor model. Colloids Surf B Biointerfaces 2019; 173: 27-35.
[53]
Bhatnagar S, Bankar NG, Kulkarni MV, Venuganti VVK. Dissolvable microneedle patch containing doxorubicin and docetaxel is effective in 4T1 xenografted breast cancer mouse model. Int J Pharm 2019; 556: 263-75.
[54]
Khazaie Y, Dorkoosh FA, Novo L, et al. Poly[N-(2-aminoethyl)ethyleneimine] as a new non-viral gene delivery carrier: the effect of two protonatable nitrogens in the monomer unit on gene delivery efficiency. J Pharm Pharm Sci 2014; 17(4): 461-74.
[55]
Kang L, Gao Z, Huang W, Jin M, Wang Q. Nanocarrier-mediated co-delivery of chemotherapeutic drugs and gene agents for cancer treatment. Acta Pharm Sin B 2015; 5(3): 169-75.
[56]
Zamore PD, Tuschl T, Sharp PA, Bartel DP. RNAi: double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell 2000; 101(1): 25-33.
[57]
Hannon GJ. RNA interference. Nature 2002; 418(6894): 244.
[58]
Esquela-Kerscher A, Slack FJ. Oncomirs - micro-RNAs with a role in cancer. Nat Rev Cancer 2006; 6(4): 259-69.
[59]
McManus MT, Sharp PA. Gene silencing in mammals by small interfering RNAs. Nat Rev Genet 2002; 3(10): 737-47.
[60]
Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004; 116(2): 281-97.
[61]
Chitkara D, Mittal A, Mahato RI. MiRNAs in pancreatic cancer: therapeutic potential, delivery challenges and strategies. Adv Drug Deliv Rev 2015; 81: 34-52.
[62]
Aliabadi HM, Landry B, Sun C, Tang T, Uludağ H. Supramolecular assemblies in functional siRNA delivery: where do we stand? Biomaterials 2012; 33(8): 2546-69.
[63]
Van de Water FM, Boerman OC, Wouterse AC, Peters JG, Russel FG, Masereeuw R. Intravenously administered siRNA accumulates in the kidney and selectively suppresses gene function in renal proximal tubules. Drug Metab Dispos 2006; 34(8): 1393-7.
[64]
Takei Y, Kadomatsu K, Yuzawa Y, Matsuo S, Muramatsu T. A small interfering RNA targeting vascular endothelial growth factor as cancer therapeutics. Cancer Res 2004; 64(10): 3365-70.
[65]
Dai X, Tan C. Combination of microRNA therapeutics with small-molecule anticancer drugs: mechanism of action and co-delivery nanocarriers. Adv Drug Deliv Rev 2015; 81: 184-97.
[66]
Chitkara D, Singh S, Mittal A. Nanocarrier-based co-delivery of small molecules and siRNA/miRNA for treatment of cancer. Ther Deliv 2016; 7(4): 245-55.
[67]
Creixell M, Peppas NA. Co-delivery of siRNA and therapeutic agents using nanocarriers to overcome cancer resistance. Nano Today 2012; 7(4): 367-79.
[68]
Suh JS, Lee JY, Choi YS, Chung CP, Park YJ. Peptide-mediated intracellular delivery of miRNA-29b for osteogenic stem cell differentiation. Biomaterials 2013; 34(17): 4347-59.
[69]
Trang P, Wiggins JF, Daige CL, et al. Systemic delivery of tumor suppressor microRNA mimics using a neutral lipid emulsion inhibits lung tumors in mice. Mol Ther 2011; 19(6): 1116-22.
[70]
Cao M, Deng X, Su S, et al. Protamine sulfate-nanodiamond hybrid nanoparticles as a vector for MiR-203 restoration in esophageal carcinoma cells. Nanoscale 2013; 5(24): 12120-5.
[71]
Moghimi SM, Hunter AC, Murray JC. Nanomedicine: current status and future prospects. FASEB J 2005; 19(3): 311-30.
[72]
Fasanaro P, Greco S, Ivan M, Capogrossi MC, Martelli F. MicroRNA: emerging therapeutic targets in acute ischemic diseases. Pharmacol Ther 2010; 125(1): 92-104.
[73]
Bader AG, Brown D, Winkler M. The promise of microRNA replacement therapy. Cancer Res 2010; 70(18): 7027-30.
[74]
Tong AW, Nemunaitis J. Modulation of miRNA activity in human cancer: a new paradigm for cancer gene therapy? Cancer Gene Ther 2008; 15(6): 341-55.
[75]
Asangani IA, Rasheed SA, Nikolova DA, et al. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene 2008; 27(15): 2128-36.
[76]
Lawrie CH, Soneji S, Marafioti T, et al. MicroRNA expression distinguishes between germinal center B cell-like and activated B cell-like subtypes of diffuse large B cell lymphoma. Int J Cancer 2007; 121(5): 1156-61.
[77]
Jongen-Lavrencic M, Sun SM, Dijkstra MK, Valk PJ, Löwenberg B. MicroRNA expression profiling in relation to the genetic heterogeneity of acute myeloid leukemia. Blood 2008; 111(10): 5078-85.
[78]
Si ML, Zhu S, Wu H, Lu Z, Wu F, Mo YY. MiR-21-mediated tumor growth. Oncogene 2007; 26(19): 2799-803.
[79]
Volinia S, Calin GA, Liu CG, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA 2006; 103(7): 2257-61.
[80]
Zhang Z, Li Z, Gao C, et al. miR-21 plays a pivotal role in gastric cancer pathogenesis and progression. Lab Invest 2008; 88(12): 1358-66.
[81]
Chan JA, Krichevsky AM, Kosik KS. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res 2005; 65(14): 6029-33.
[82]
Yanaihara N, Caplen N, Bowman E, et al. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell 2006; 9(3): 189-98.
[83]
Iorio MV, Visone R, Di Leva G, et al. MicroRNA signatures in human ovarian cancer. Cancer Res 2007; 67(18): 8699-707.
[84]
Roldo C, Missiaglia E, Hagan JP, et al. MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. J Clin Oncol 2006; 24(29): 4677-84.
[85]
Devulapally R, Sekar TV, Paulmurugan R. Formulation of anti-miR-21 and 4-hydroxytamoxifen co-loaded biodegradable polymer nanoparticles and their antiproliferative effect on breast cancer cells. Mol Pharm 2015; 12(6): 2080-92.
[86]
Qian X, Long L, Shi Z, et al. Star-branched amphiphilic PLA-b-PDMAEMA copolymers for co-delivery of miR-21 inhibitor and doxorubicin to treat glioma. Biomaterials 2014; 35(7): 2322-35.
[87]
Ren Y, Wang R, Gao L, et al. Sequential co-delivery of miR-21 inhibitor followed by burst release doxorubicin using NIR-responsive hollow gold nanoparticle to enhance anticancer efficacy. J Control Release 2016; 228: 74-86.
[88]
Hu N, Yin JF, Ji Z, et al. Strengthening gastric cancer therapy by trastuzumab-conjugated nanoparticles with simultaneous encapsulation of anti-MiR-21 and 5-fluorouridine. Cell Physiol Biochem 2017; 44(6): 2158-73.
[89]
Chang T-C, Wentzel EA, Kent OA, et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol Cell 2007; 26(5): 745-52.
[90]
Liu C, Kelnar K, Vlassov AV, Brown D, Wang J, Tang DG. Distinct microRNA expression profiles in prostate cancer stem/progenitor cells and tumor-suppressive functions of let-7. Cancer Res 2012; 72(13): 3393-404.
[91]
Hermeking H. The miR-34 family in cancer and apoptosis. Cell Death Differ 2010; 17(2): 193-9.
[92]
Emi M, Kim R, Tanabe K, Uchida Y, Toge T. Targeted therapy against Bcl-2-related proteins in breast cancer cells. Breast Cancer Res 2005; 7(6): 940-52.
[93]
Al-Qadi S, Alatorre-Meda M, Zaghloul EM, Taboada P, Remunán-López C. Chitosan-hyaluronic acid nanoparticles for gene silencing: the role of hyaluronic acid on the nanoparticles’ formation and activity. Colloids Surf B Biointerfaces 2013; 103: 615-23.
[94]
de la Fuente M, Seijo B, Alonso MJ. Novel hyaluronic acid-chitosan nanoparticles for ocular gene therapy. Invest Ophthalmol Vis Sci 2008; 49(5): 2016-24.
[95]
Deng X, Cao M, Zhang J, et al. Hyaluronic acid-chitosan nanoparticles for co-delivery of MiR-34a and doxorubicin in therapy against triple negative breast cancer. Biomaterials 2014; 35(14): 4333-44.
[96]
Yao C, Liu J, Wu X, et al. Reducible self-assembling cationic polypeptide-based micelles mediate co-delivery of doxorubicin and microRNA-34a for androgen-independent prostate cancer therapy. J Control Release 2016; 232: 203-14.
[97]
Shi S, Han L, Deng L, et al. Dual drugs (microRNA-34a and paclitaxel)-loaded functional solid lipid nanoparticles for synergistic cancer cell suppression. J Control Release 2014; 194: 228-37.
[98]
Cui X, Sun Y, Shen M, et al. Enhanced chemotherapeutic efficacy of paclitaxel nanoparticles co-delivered with microRNA-7 by inhibiting paclitaxel-induced EGFR/ERK pathway activation for ovarian cancer therapy. ACS Appl Mater Interfaces 2018; 10(9): 7821-31.
[99]
Gandhi NS, Tekade RK, Chougule MB. Nanocarrier mediated delivery of siRNA/miRNA in combination with chemotherapeutic agents for cancer therapy: current progress and advances. J Control Release 2014; 194: 238-56.
[100]
Dehghan Kelishady P, Saadat E, Ravar F, Akbari H, Dorkoosh F. Pluronic F127 polymeric micelles for co-delivery of paclitaxel and lapatinib against metastatic breast cancer: preparation, optimization and in vitro evaluation. Pharm Dev Technol 2015; 20(8): 1009-17.
[101]
Xiong X-B, Lavasanifar A. Traceable multifunctional micellar nanocarriers for cancer-targeted co-delivery of MDR-1 siRNA and doxorubicin. ACS Nano 2011; 5(6): 5202-13.
[102]
Patil YB, Swaminathan SK, Sadhukha T, Ma L, Panyam J. The use of nanoparticle-mediated targeted gene silencing and drug delivery to overcome tumor drug resistance. Biomaterials 2010; 31(2): 358-65.
[103]
Wang M, Wang J, Li B, Meng L, Tian Z. Recent advances in mechanism-based chemotherapy drug-siRNA pairs in co-delivery systems for cancer: a review. Colloids Surf B Biointerfaces 2017; 157: 297-308.
[104]
Saad M, Garbuzenko OB, Minko T. Co-delivery of siRNA and an anticancer drug for treatment of multidrug-resistant cancer. Nanomedicine 2008; 3(6): 761-76.
[105]
Yu YH, Kim E, Park DE, et al. Cationic solid lipid nanoparticles for co-delivery of paclitaxel and siRNA. Eur J Pharm Biopharm 2012; 80(2): 268-73.
[106]
Zhang Y, Peng L, Mumper RJ, Huang L. Combinational delivery of c-myc siRNA and nucleoside analogs in a single, synthetic nanocarrier for targeted cancer therapy. Biomaterials 2013; 34(33): 8459-68.
[107]
Zhu L, Perche F, Wang T, Torchilin VP. Matrix metalloproteinase 2-sensitive multifunctional polymeric micelles for tumor-specific co-delivery of siRNA and hydrophobic drugs. Biomaterials 2014; 35(13): 4213-22.
[108]
Gavai AV, Quesnelle C, Norris D, et al. Discovery of clinical candidate BMS-906024: a potent pan-notch inhibitor for the treatment of leukemia and solid tumors. ACS Med Chem Lett 2015; 6(5): 523-7.
[109]
Zhu C, Jung S, Luo S, et al. Co-delivery of siRNA and paclitaxel into cancer cells by biodegradable cationic micelles based on PDMAEMA-PCL-PDMAEMA triblock copolymers. Biomaterials 2010; 31(8): 2408-16.
[110]
Jia H-Z, Zhang W, Zhu JY, et al. Hyperbranched-hyperbranched polymeric nanoassembly to mediate controllable co-delivery of siRNA and drug for synergistic tumor therapy. J Control Release 2015; 216: 9-17.
[111]
Zhan C, Wei X, Qian J, Feng L, Zhu J, Lu W. Co-delivery of TRAIL gene enhances the anti-glioblastoma effect of paclitaxel in vitro and in vivo. J Control Release 2012; 160(3): 630-6.
[112]
Su B, Cengizeroglu A, Farkasova K, et al. Systemic TNFα gene therapy synergizes with liposomal doxorubicine in the treatment of metastatic cancer. Mol Ther 2013; 21(2): 300-8.
[113]
Sundaram S, Trivedi R, Durairaj C, Ramesh R, Ambati BK, Kompella UB. Targeted drug and gene delivery systems for lung cancer therapy. Clin Cancer Res 2009; 15(23): 7299-308.
[114]
Kotmakçı M, Çetintaş VB, Kantarcı AG. Preparation and characterization of lipid nanoparticle/pDNA complexes for STAT3 downregulation and overcoming chemotherapy resistance in lung cancer cells. Int J Pharm 2017; 525(1): 101-11.
[115]
Teo PY, Cheng W, Hedrick JL, Yang YY. Co-delivery of drugs and plasmid DNA for cancer therapy. Adv Drug Deliv Rev 2016; 98: 41-63.
[116]
Yang Z, Gao D, Cao Z, et al. Drug and gene co-delivery systems for cancer treatment. Biomater Sci 2015; 3(7): 1035-49.
[117]
Yu D, Li W, Zhang Y, Zhang B. Anti-tumor efficiency of paclitaxel and DNA when co-delivered by pH responsive ligand modified nanocarriers for breast cancer treatment. Biomed Pharmacother 2016; 83: 1428-35.
[118]
Chowdhury N, Vhora I, Patel K, Doddapaneni R, Mondal A, Singh M. Liposomes co-loaded with 6-phosphofructo-2-kinase/fructose-2, 6-biphosphatase 3 (PFKFB3) shRNA plasmid and docetaxel for the treatment of non-small cell lung cancer. Pharm Res 2017; 34(11): 2371-84.
[119]
Davoodi P, Srinivasan MP, Wang C-H. Synthesis of intracellular reduction-sensitive amphiphilic polyethyleneimine and poly(ε-caprolactone) graft copolymer for on-demand release of doxorubicin and p53 plasmid DNA. Acta Biomater 2016; 39: 79-93.
[120]
Xu Z, Zhang Z, Chen Y, Chen L, Lin L, Li Y. The characteristics and performance of a multifunctional nanoassembly system for the co-delivery of docetaxel and iSur-pDNA in a mouse hepatocellular carcinoma model. Biomaterials 2010; 31(5): 916-22.
[121]
Mai Q, Shen S, Liu Y, Tang C, Yin C. PEG modified trimethyl chitosan based nanoparticles for the codelivery of doxorubicin and iSur-pDNA. Mater Lett 2019; 238: 143-6.
[122]
Dong S, Zhou X, Yang J. TAT modified and lipid - PEI hybrid nanoparticles for co-delivery of docetaxel and pDNA. Biomed Pharmacother 2016; 84: 954-61.
[123]
Minaei A, Sabzichi M, Ramezani F, Hamishehkar H, Samadi N. Co-delivery with nano-quercetin enhances doxorubicin-mediated cytotoxicity against MCF-7 cells. Mol Biol Rep 2016; 43(2): 99-105.
[124]
Qureshi WA, Zhao R, Wang H, et al. Co-delivery of doxorubicin and quercetin via mPEG-PLGA copolymer assembly for synergistic anti-tumor efficacy and reducing cardio-toxicity. Sci Bull 2016; 61(21): 1689-98.
[125]
Sarisozen C, Abouzeid AH, Torchilin VP. The effect of co-delivery of paclitaxel and curcumin by transferrin-targeted PEG-PE-based mixed micelles on resistant ovarian cancer in 3-D spheroids and in vivo tumors. Eur J Pharm Biopharm 2014; 88(2): 539-50.
[126]
Yan J, Wang Y, Jia Y, et al. Co-delivery of docetaxel and curcumin prodrug via dual-targeted nanoparticles with synergistic antitumor activity against prostate cancer. Biomed Pharmacother 2017; 88: 374-83.
[127]
Fatma S, Talegaonkar S, Iqbal Z, et al. Novel flavonoid-based biodegradable nanoparticles for effective oral delivery of etoposide by P-glycoprotein modulation: an in vitro, ex vivo and in vivo investigations. Drug Deliv 2016; 23(2): 500-11.
[128]
Katiyar SS, Muntimadugu E, Rafeeqi TA, Domb AJ, Khan W. Co-delivery of rapamycin- and piperine-loaded polymeric nanoparticles for breast cancer treatment. Drug Deliv 2016; 23(7): 2608-16.
[129]
Zhang J, Wang L, Fai Chan H, et al. Co-delivery of paclitaxel and tetrandrine via iRGD peptide conjugated lipid-polymer hybrid nanoparticles overcome multidrug resistance in cancer cells. Sci Rep 2017; 7: 46057.
[130]
Zhang Y, Yang C, Wang W, et al. Co-delivery of doxorubicin and curcumin by pH-sensitive prodrug nanoparticle for combination therapy of cancer. Sci Rep 2016; 6: 21225.
[131]
Zhang J, Li J, Shi Z, et al. pH-sensitive polymeric nanoparticles for co-delivery of doxorubicin and curcumin to treat cancer via enhanced pro-apoptotic and anti-angiogenic activities. Acta Biomater 2017; 58: 349-64.
[132]
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.
[133]
Sabra SA, Elzoghby AO, Sheweita SA, et al. Self-assembled amphiphilic zein-lactoferrin micelles for tumor targeted co-delivery of rapamycin and wogonin to breast cancer. Eur J Pharm Biopharm 2018; 128: 156-69.
[134]
Yan J, Wang Y, Jia Y, et al. Co-delivery of docetaxel and curcumin prodrug via dual-targeted nanoparticles with synergistic antitumor activity against prostate cancer. Biomed Pharmacother 2017; 88: 374-83.
[135]
Baek J-S, Cho C-W. A multifunctional lipid nanoparticle for co-delivery of paclitaxel and curcumin for targeted delivery and enhanced cytotoxicity in multidrug resistant breast cancer cells. Oncotarget 2017; 8(18): 30369-82.
[136]
Wang Z, Li X, Wang D, et al. Concurrently suppressing multidrug resistance and metastasis of breast cancer by co-delivery of paclitaxel and honokiol with pH-sensitive polymeric micelles. Acta Biomater 2017; 62: 144-56.
[137]
Dong XY, Lang TQ, Yin Q, Zhang PC, Li YP. Co-delivery of docetaxel and silibinin using pH-sensitive micelles improves therapy of metastatic breast cancer. Acta Pharmacol Sin 2017; 38(12): 1655-62.
[138]
Li C, Ge X, Wang L. Construction and comparison of different nanocarriers for co-delivery of cisplatin and curcumin: a synergistic combination nanotherapy for cervical cancer. Biomed Pharmacother 2017; 86: 628-36.
[139]
Sabra SA, Elzoghby AO, Sheweita SA, et al. Self-assembled amphiphilic zein-lactoferrin micelles for tumor targeted co-delivery of rapamycin and wogonin to breast cancer. Eur J Pharm Biopharm 2018; 128: 156-69.
[140]
Yu J, Chen H, Jiang L, Wang J, Dai J, Wang J. Co-delivery of Adriamycin and P-gp inhibitor Quercetin using PEGylated liposomes to overcome cancer resistance. J Pharm Sci 2019; 108(5): 1788-99.
[141]
Yang T, Lan Y, Cao M, et al. Glycyrrhetinic acid-conjugated polymeric prodrug micelles co-delivered with doxorubicin as combination therapy treatment for liver cancer. Colloids Surf B Biointerfaces 2019; 175: 106-15.
[142]
Saneja A, Kumar R, Mintoo MJ, et al. Gemcitabine and betulinic acid co-encapsulated PLGA-PEG polymer nanoparticles for improved efficacy of cancer chemotherapy. Mater Sci Eng C 2019; 98: 764-71.
[143]
Wang R, Yang M, Li G, et al. Paclitaxel-betulinic acid hybrid nanosuspensions for enhanced anti-breast cancer activity. Colloids Surf B Biointerfaces 2019; 174: 270-9.
[144]
Zhang X, Li L, Liu Q, et al. Co-delivery of rose bengal and doxorubicin nanoparticles for combination photodynamic and chemo-therapy. J Biomed Nanotechnol 2019; 15(1): 184-95.
[145]
Sesarman A, Tefas L, Sylvester B, et al. Co-delivery of curcumin and doxorubicin in PEGylated liposomes favored the antineoplastic C26 murine colon carcinoma microenvironment. Drug Deliv Transl Res 2019; 9(1): 260-72.
[146]
Li Y, Yang D, Wang Y, Li Z, Zhu C. Co-delivery doxorubicin and silybin for anti-hepatoma via enhanced oral hepatic-targeted efficiency. Int J Nanomedicine 2018; 14: 301-15.
[147]
Jeong EH, Ryu JH, Jeong H, et al. Efficient delivery of siRNAs by a photothermal approach using plant flavonoid-inspired gold nanoshells. Chem Commun 2014; 50(87): 13388-90.
[148]
Desai PR, Marepally S, Patel AR, Voshavar C, Chaudhuri A, Singh M. Topical delivery of anti-TNFα siRNA and capsaicin via novel lipid-polymer hybrid nanoparticles efficiently inhibits skin inflammation in vivo. J Control Release 2013; 170(1): 51-63.
[149]
Tang L, Wang K. Chronic inflammation in skin malignancies. J Mol Signal 2016; 11: 2.
[150]
Kantara C, O’Connell M, Sarkar S, Moya S, Ullrich R, Singh P. Curcumin promotes autophagic survival of a subset of colon cancer stem cells, which are ablated by DCLK1-siRNA. Cancer Res 2014; 74(9): 2487-98.
[151]
Arranz-Romera A, Davis BM, Bravo-Osuna I, et al. Simultaneous co-delivery of neuroprotective drugs from multi-loaded PLGA microspheres for the treatment of glaucoma. J Control Release 2019; 297: 26-38.
[152]
Uz M, Kalaga M, Pothuraju R, et al. Dual delivery nanoscale device for miR-345 and gemcitabine co-delivery to treat pancreatic cancer. J Control Release 2019; 294: 237-46.
[153]
Hu Q, Sun W, Wang C, Gu Z. Recent advances of cocktail chemotherapy by combination drug delivery systems. Adv Drug Deliv Rev 2016; 98: 19-34.
[154]
Akbarzadeh A, Rezaei-Sadabady R, Davaran S, et al. Liposome: classification, preparation, and applications. Nanoscale Res Lett 2013; 8(1): 102.
[155]
Assunção-Silva RC, Gomes ED, Silva NA, Salgado AJ. Nanoengineered biomaterials for spinal cord regeneration. Nanoengin Biomater Regen Med 2019; pp. 167-85.
[156]
Wu J, Xu F, Li S, et al. Porous polymers as multifunctional material platforms toward task-specific applications. Adv Mater 2019; 31(4): 1802922.
[157]
Chaudhary Z, Ahmed N, Ur-Rehman A, Khan GM. Lipid polymer hybrid carrier systems for cancer targeting: a review. Int J Polym Mater Po 2018; 67(2): 86-100.
[158]
Zhang Y, Zhang P, Zhu T. Ovarian carcinoma biological nanotherapy: comparison of the advantages and drawbacks of lipid, polymeric, and hybrid nanoparticles for cisplatin delivery. Biomed Pharmacother 2019; 109: 475-83.
[159]
Khor SY, Hu J, McLeod VM, et al. Molecular weight (hydrodynamic volume) dictates the systemic pharmacokinetics and tumour disposition of PolyPEG star polymers. Nanomedicine 2015; 11(8): 2099-108.
[160]
Xu CL, Ruan MZC, Mahajan VB, Tsang SH. Viral delivery systems for CRISPR. Viruses 2019; 11(1): 28.
[161]
Yang N. An overview of viral and nonviral delivery systems for microRNA. Int J Pharm Investig 2015; 5(4): 179-81.

© 2024 Bentham Science Publishers | Privacy Policy