Abstract
One of the major problems being faced by researchers and clinicians in leukemic treatment is the development of multidrug resistance (MDR) which restrict the action of several tyrosine kinase inhibitors (TKIs). MDR is a major obstacle to the success of cancer chemotherapy. The mechanism of MDR involves active drug efflux transport of ABC superfamily of proteins such as Pglycoprotein (P-gp/ABCB1), multidrug resistance-associated protein 2 (MRP2/ABCC2), and breast cancer resistance protein (BCRP/ABCG2) that weaken the effectiveness of chemotherapeutics and negative impact on the future of anticancer therapy. In this review, the authors aim to provide an overview of various multidrug resistance (MDR) mechanisms observed in cancer cells as well as the various strategies developed to overcome these MDR. Extensive studies have been carried out since last several years to enhance the efficacy of chemotherapy by defeating these MDR mechanisms with the use of novel anticancer drugs that could escape from the efflux reaction, MDR modulators or chemosensitizers, multifunctional nanotechnology, and RNA interference (RNAi) therapy.
Key words: ABC transporter, breast cancer resistance protein (BCRP), chronic myeloid leukemia (CML), multidrug resistance (MDR), P-glycoprotein
Introduction
Chronic myeloid leukemia (CML) is characterized by the increased proliferation of pluripotent hematopoietic stem cells.1-3 It is genetically distinguished by the presence of the t (9;22), (q34; q11) reciprocal chromosomal translocation. The Abelson protooncogene (ABL) on chromosome 9 is translocated to the Breakpoint Cluster Region (BCR) on chromosome 22, forming a fusion gene in which ABL-related tyrosine kinase activity is constitutively activated.2,4-6 Imatinib, also called imatinib mesylate or signal transduction inhibitor 571 (STI571) or commercially known as Gleevec7,8 is a BCR-ABL tyrosine kinase inhibitor (TKI), which has revolutionized the treatment of chronic myeloid leukemia. Imatinib is the first approved medicine that targets the ATP binding domain of BCR- ABL tyrosine kinase by reversing its effects.9,10 While 5-years overall survival rate of CML patients without imatinib was recorded as 24%, it increased gradually to 39% (1966-1998) and 56.8% (1999-2006) and the overall survival was estimated to 85% with the application of imatinib.2,11,12 Simultaneously during treatment, the development of resistance in many leukemia patients in the chronic phase8,13,14 and accelerated/ blast crisis phase8 limits the use of tyrosine kinase inhibitors. The phenomenon of resistance against the chemotherapeutic agents called chemoresistance or multidrug resistance (MDR).15 It was described by Keld Dano as the active outward transport of Vinca alkaloids and anthracyclines from murine Ehrlich ascites tumor cells.16
MDR is a phenomenon in which the cancer cell’s resistance to a drug is accompanied by resistance to a pharmacologically and structurally different class of drugs.17 Although, the mechanisms of anticancer drug resistances are a complex process and commonly categorized into drug dependent, target-dependent, and drug/target-independent. Drug-dependent MDR is caused by overexpression of efflux drug transporters and detoxifying enzymes which lowers the uptake or enhance the efflux of drugs in cancerous cells. Target-dependent MDR is primarily attributed to the factors influencing drug targets such as translocation, deletion, mutation, and amplification. Drug/target-independent MDR occur either genetically or epigenetically due to the inactivation of drug targeting by changing cell signaling cascades.18-21 Interestingly, one of the most significant mechanism causing MDR is the overexpression of adenosine triphosphate (ATP)-binding cassette (ABC) superfamily of transporters, which efflux both cytotoxic agents and targeted anticancer drugs using ATP driven energy.22-24 Therefore, this review aims to discuss the role of those three mammalian ABC transporters that mediate MDR in cancerous cells and subsequent development of strategies to overcome MDR in cancer chemotherapy.
Properties of ABC transporters in human physiology
The human ABC transporters, one of the largest groups of membrane protein complexes consist of 49 members that has been divided into seven subfamilies (ABC-A through to ABC-G) (Table 1) based on the sequence similarities of nucleotide-binding domain (NBD) as well as structural organization,25-27 though there are more in bacteria and parasites.28
Transporters are membrane-bound proteins that facilitate the translocation of substrate molecules across the biological membranes. Those mammalian ABC transporters which are localized to the plasma membrane throughout the body such as the liver, intestine, kidney, and organs with barrier functions, such as the brain, testes and placenta, as well as to membranes that compose various subcellular organelles, significantly decrease the intracellular concentration of various drugs, drug conjugates and metabolites by export.27,29 Structurally, all ABC transporters contain two transmembrane domains (TMDs) and two nucleotides (ATP)-binding domains (NBDs).30 Generally, they exhibit a common structural fold which is made up of a core of six TM helices per TMD. The hydrophobic TMDs are structurally different, which alternately identify and translocate various substrates upon conformational changes. So, the TMDs which lengthen the membrane and form channels may determine the transport features of substrates.31,32 The energy required for translocation or efflux of physiological and xenobiotic substances from the cytosol to the extracellular space is provided by ATP hydrolysis via ATPase.30 Till date, at least 15 human ABC superfamily transporter proteins such as Pglycoprotein (P-gp/ABCB1), multidrug resistance-associated protein 2 (MRP2/ABCC2) and breast cancer resistance protein (BCRP/ABCG2) deal with MDR as drug efflux pumps. However, various recent studies have confirmed that ABC transporter proteins efficacy enhance their role through some other mechanisms in addition to drug efflux (Figure 1).33-36
P-glycoprotein (ABCB1)
P-glycoprotein is the first member of ABC transporter family that linked with the overexpression of the ATP binding cassette transporter which is encoded by the ABCB1 gene.37 It is the highly studied ABC drug efflux transporter to date38,39 and its role in resistance against anticancer drugs has been known for more than thirty years.40,41 The most outstanding property of P-gp is the difference in the structure of substrates transported, including a large number of drugs useful in therapeutic applications. These substrates include anticancer drugs, analgesics, antibiotics, antiarrhythmics, antihistamines, calcium-channel blockers, chemotherapeutic drugs, fluorophores HIV-protease inhibitors, immunosuppressive agents, natural products, neuroleptics, pesticides and many others (Table 2).42-53 P-glycoprotein is highly expressed on the liver, placenta, lower gastrointestinal tract (jejunum, ileum, and colon), proximal tubules epithelia of kidney and luminal bloodbrain barrier.41 P-gp seems to be an essential factor of pharmacokinetics due to its wide range of substrate localization, and a good negotiator of transporter-mediated drug-drug interactions. Some of the current research findings also suggest that transport of cytotoxic substrates for P-glycoprotein can be suppressed by a large variety of chemical compounds known as reversal agents or P-gp blockers.38 Not all but some of the identified inhibitors act as competitive inhibitors, for example, calcium channel inhibitor verapamil or an immunosuppressor cyclosporin A. Also, the other inhibitors probably work through some different mechanisms.
Figure 1.
Model of the secondary structure of efflux membrane transporters of the ABC family. A) P-gp/ABCB1; B) MRP2/ABCC2; C) BCRP/ABCG2. TMD, transmembrane domain; NBD, nucleotide-binding domain; L0, loop 0.
Table 1.
Human ABC transporter gene family.
Subfamily names | Assumed name | Number of genes | Number of pseudogenes |
---|---|---|---|
ABCA | ABC1 | 12 | 5 |
ABCB | MDR | 11 | 4 |
ABCC | MRP | 13 | 2 |
ABCD | ALD | 4 | 4 |
ABCE | OABP | 1 | 2 |
ABCF | GGN20 | 3 | 2 |
ABCG | White | 5 | 2 |
Multidrug resistance protein 2 (MRP2/ABCC2)
The ABCC gene family has been divided into 13 subfamily members (ABCC1 through to ABCC13). MRP2 is the second member of the MRP subfamily of ABC transporter encoded by ABCC2 gene.54,55 MRP2 was initially cloned from rat liver using the same approaches that took benefit of its structural similarity to human MRP1.56,57 MRP2 has entirely different expression pattern in the apical plasma membrane of hepatocytes, the brush-border membrane of renal proximal tubules and small intestine, where it is situated to play an essential role in the elimination and oral bioavailability of a wide variety of drugs including endogenous glucuronides, sulphates and GSH conjugates from the cells. Furthermore, MRP2 involves in biliary elimination of LTC4 and bilirubin conjugates.58,59 Also, MRP2 mRNA has been detected in the gallbladder, peripheral nerves, placental trophoblasts, and CD4+ lymphocytes.60-63 A new report on cancer chemotherapy suggest that effectiveness of cisplatin based treatment in patients with hepatocellular carcinoma depends upon MRP2 mRNA expression level.64 Similar to MRP1, MRP2 can transport many different chemotherapeutic substrates which are listed in Table 2.
The transport of various substrates by MRP2 is based on different approaches, such as the analysis of distinct compounds transported into the bile of normal rats, but none of Mrp2 mutant rats; irregular uptake of substrates into vesicles of bile canalicular membrane obtained from normal and Mrp2 mutant rats; transduction or transfection of human MRP2 or rat/rabbit Mrp2 cDNA into cell lines followed by drug resistance analysis, accumulation of compounds into cells, and transepithelial transfer of compounds. 42,65
Same as P-glycoprotein inhibitors some MRP2 inhibitors have been verified to work to a greater or lesser extent in entire cells which comprise of cyclosporin A, benzbromarone, probenecid, sulfinpyrazone, PSC 833, PAK-104P and MK571.66-68 The compounds recognized so far have significant activity against MRP1 and P-gp but, hard to obtain against MRP2.
Table 2.
Some clinically selected cytotoxic substrates associated with multidrug resistance transporters.
MDR1 |
---|
Anticancer drugs: Actinomycin D, Anthracenes (bisantrene, mitoxantrone), Anthracyclines (daunorubicin, doxorubicin, epirubicin), chlorambucil, cisplatin, colchicine, cytarabine, Epipodophyllotoxins [etoposide (VP-16), Teniposide (VM-26)], 5-fluorouracil, gefitinib, hydroxyurea, irinotecan (CPT-11), methotrexate, mitomycin C, Taxanes (docetaxel, paclitaxel), tamoxifen, topotecan, Vinca alkaloids (vinblastine, vincristine, Vinorelbine Vindesine) |
Analgesics: asimadoline, fentanyl, morphine, pentazocine |
Antibiotics: cefoperazone, ceftriaxone, clarithromycin, doxycycline, erythromycin, gramicidin A, gramicidin D, grepafloxacin, itraconazole, ketoconazole, levofloxacin, rifampicin, sparfloxacin, tetracycline, valinomycin |
Antiarrhythmics: amiodarone, digoxin, lidocaine, propafenone, quinidine, verapamil |
Antihistamines: cimetidine, fexofenadine, ranitidine, terfenadine |
Antilipidemic: lovastatin, simvastatin |
Calcium channel blockers: azidopine, bepridil, diltiazem, felodipine, nifedipine, nisoldipine, nitrendipine, tiapamil, verapamil |
Fluorophores: Hoechst 33342/33258, rhodamine 123, calcein AM (calcein acetoxymethyl ester), Fluo-3 AM, Fura-2 AM |
HIV protease inhibitors: saquinavir, ritonavir, nelfinavir, lopinavir, indinavir, amprenavir |
Immunosuppressive agents: cyclosporin A, cyclosporin H, FK506, sirolimus, tacrolimus, valspodar (PSC-833) |
Natural products: curcuminoids, flavonoids |
Neuroleptics: chlorpromazine, phenothiazine |
Others: Amitriptyline (antidepressant), Dipyridamole (anticoagulant), BCECF-AM, bepridil, diltiazem, endosulfan, leupeptin, methyl parathion, paraquat, pepstatin A, trifluoperazine, trans-flupentixol |
MRP2 |
Anticancer drugs: cisplatin, doxorubicin, epirubicin, etoposide, irinotecan, mitoxantrone, methotrexate, SN-38, Vinca alkaloids (vinblastine, vincristine) |
Antibiotics: ampicillin, azithromycin, cefodizime, ceftriaxone, grepafloxacine, irinotecan |
Antihypertensives: olmesartan, temocaprilate |
HIV drugs: adevovir, cidofovir, indinavir, lopinavir, nelfinavir, ritonavir, saquinavir |
Others: ethinylestradiol-3-O-glucuronide, genistein-7-glucoside, p-Aminohippurate, phloridzin, quercetin 4 -β-glucoside |
BCRP |
Anticancer drugs: Anthracenes (mitoxantrone), Anthracyclines (daunorubicin, doxorubicin, epirubicin), Epipodophyllotoxins (etoposide, teniposide), gefitinib, imatinib (TKI), irinotecan, methotrexate, SN-38, topotecan, |
Antibiotics: ciprofloxacin, norfloxacin, ofloxacin |
Antihypertensives: reserpine |
Antivirals: delavirdine, lamivudine, lopinavir, nelfinavir, zidovudine |
Calcium channel blockers: nicardipine |
Lipid lowering drugs: cerivastatin, pravastatin, rosuvastatin |
Others: azidothymidine, cyclosporin A, chrysin, lamivudine, ortataxel, quercetin |
Data were collected from References.42-53
Breast cancer resistance protein (BCRP/ABCG2)
Although we have discussed earlier two transporters, MDR1 P-gp and MRP2 which are involved in MDR development similar to that, the third subfamily of transporter known as breast cancer resistance protein have an enormous impact in the physiology, pathophysiology, pharmacokinetics, and toxicokinetics.69 Based on analogy with other ABC transporters BCRP is the second member of the G subfamily (ABCG2) which is thought to function as homo or heterodimers.24,27,70 BCRP is the first clone based on its higher expression in doxorubicin-resistant breast cancer cell line (MCF-7).69-73 This gene was isolated from the breast cancer cell line, that’s why called breast cancer resistance protein (BCRP). This transporter is extensively distributed in the endothelial cells of brain capillaries, liver, intestine, proximal tubule cells of the kidney, and contributing to the absorption, distribution, and elimination of the drugs and endogenous compounds in addition to protection of tissues against deadly xenobiotic exposures.22,74 Just like MDR1/P-gp and MRP2, BCRP transports structurally and functionally diverse kind of substrates such as analgesics, antibiotics, anticancer and antivirals drugs, etc. (Table 2). Furthermore, the transport through above transporters can be inhibited by small molecules listed in Table 3.32,49,68,75-82
Strategic development to overcome MDR in cancer chemotherapy
Several strategies have been made to overcome MDR in cancer cells through the synthesis of new molecules applying biochemical and physical approaches as well as nanotechnologies.83-85 One such strategy is the development of novel anticancer drugs that are not P-gp substrates, but many different chemosensitizers identified were substrates for P-gp, therefore, worked by contesting with cytotoxic compounds for efflux by the P-gp pump. Verapamil and cyclosporine A are the first-generation P-gp modulators which block the transport function of the transporter.86,87 Other examples of modulators are trifluoperazine, quinidine, and progesterone.82 The second-generation P-gp modulators such as dexverapamil, dexniguldipine, valspodar (PSC 833), and biricodar (VX-710) are more effective and less toxic than their predecessors.88 Among them, the best-studied agent is valspodar, a non-immunosuppressive derivative of cyclosporin D that inhibits P-gp by 10-20 folds higher than cyclosporin A.89,90 Many cytotoxic agents that are substrates for P-gp are also substrates for the cytochrome P450 isoenzyme 3A4, and they create toxic pharmacokinetic interactions due to competition between them.91 Therefore, different anticancer drugs are being developed that cannot be identified by P-gp and/or ABC transporters. For example, BMS-184476,92 Ortataxel93 and Taxane analogues DJ-927.94 Similarly, Chinese traditional medicine is known as indirubin usually do not carried out by P-gp, but prevent the efflux of doxorubicin and vincristine by P-gp.95
Still, the problem with MDR continues which motivated the scientists to develop third-generation P-gp modulators such as Tariquidar (XR9576),96 Zosuquidar (LY335979),97 Laniquidar (R101933),98 Elacridar (F12091).99 While Tariquidar inhibits ATPase activity of P-gp even at a very low concentration (25-80 nM),100 Zosuquidar (an oral P-gp inhibitor) stimulate the intake of daunorubicin, idarubicin, mitoxantrone, and mylotarg in acute myeloid leukemia.101
The other strategies involve the use of microRNAs (miRNAs). They are small, highly conserved non-coding RNA molecules that bind to the 3’ UTR of mRNA and suppress the protein expression throughout the translation.102,103 Generally, miRNAs get modified within cancer cells that may lead to the development of MDR.104 These are some miRNAs (miR-27a, miR-296, miR-298, miR-451, miR-1253) which have been recognized as an inhibitor of P-glycoprotein, and their therapeutic index was evaluated in breast cancer cells lines (MCF-7) and esophageal squamous carcinoma cells.105-107 The complete understanding of the mechanism of miRNAs regulation may contribute to the development of a drug against MDR.108 siRNAs can also reverse the MDR through inhibition of MDR genes, for example, ABCB1 (MDR1), ABCB4 (MDR3), ABCG2 (BCRP).109-112
The monoclonal antibodies also play a crucial role in reversing drug resistance mediated by P-gp, such as MRK- 16 and MRK-17 were developed to reverse the drug resistance effect both in vivo and in vitro during the 1980s.113-116 While MRK-16 act as an effective blocker against actinomycin D and vincristine efflux, MRK-17 has an active role in the inhibition of MDR cell proliferation. UIC2 is a newly designed mouse monoclonal antibody which binds to a cell surface epitope of P-gp in a specific manner and suppresses the drug efflux and boost cell cytotoxicity.117 The conjugates of monoclonal antibodies with P-gp-reversing agents may increase anticancerous properties.
Currently, nanotechnology-based approaches are being used as a more efficient strategy to overcome MDR. Different types of nanoparticles such as metals, polymers, dendrimers, liposomes, solid lipids, quantum dots, and micelles are widely used to transport anti-cancer, anti-infection, or anti-inflammatory drugs to exact target cells/tissues of patients. The size of nanoparticles greatly varies up to several hundred nm.83,84,118-121 The assembly of nanoparticles takes place in several layers, but the surface coating is a major beneficiary step for the solubility, specificity, and stability of these nanoparticles.122,123 The most frequently used nanovehicles for drug delivery to the target cells/tissues are bio-degradable polymeric nanoparticles. The polymers may be either natural such as gelatin, chitosan, and albumin or synthetic for example, poly (d, l-lactic acid) (PLA), poly (d, l-lactic-co-glycolic acid) (PLGA), and poly (ε-caprolactone) (PLC).124,125 Liposome nanoparticles are also used in drug delivery systems. Liposomes may encapsulate soluble drugs and retain their natural activity by forming phospholipid bilayers and micelle spheres. It is primarily used for the delivery of those drugs which are unable to diffuse through membranes. Doxil and Daunoxome are the two nanodrugs in which doxorubicin or daunorubicin have been merged into 80-90 nm single layer liposome nanoparticles.126 Liposomes nanoparticles show potential activity in the battle against MDR. Gold nanoparticles (AuNPs) also considered as the right choice for drug delivery and can be synthesized easily.127 Gold nanoparticles provide their surface for targeting tumors and drug release in a controlled way.127,128 For example, Dox-PLGA-Au has enough potential to reduce tumor growth and win the battle against MDR.129
Table 3.
Inhibitors of P-gp, MRP2 and BCRP transporters.
P-gp/ABCB1 | Atorvastatin, amlodipine, cyclosporin A, dexniguldipine, disulfiram, verapamil, quinidine, nifedipine, MS-209, GF120918, LY475776, V-104, LY335979, OC144-093, pluronic L61, PSC-833, R101933, S9788, VX-710, XR-9576 |
MRP2/ABCC2 | Azithromycin, cyclosporin A, furosemide, glibenclamide, probenecid, MK-571 |
BRCP/ABCG2 | Cyclosporin A, dipyridamole, elacridar, fumitremorgin C, novobiocin, ortataxel, reserpine, ritonavir, tariquidar, GF120918, VX-710, XR-9576 |
Conclusions
The current review article summarizes the most recent studies of ABC superfamily transporters and their contribution to MDR in cancer chemotherapy. Almost 90% of cancer patients are facing treatment failure due to drug resistance. From the previous experimental studies, it has been confirmed that ABC drug transporters play an important role in the efflux of endogenous toxicants and xenobiotics. Therefore, some new strategies for effective chemotherapeutic treatment against MDR triggered by ABC drug transporters are required. The structural and functional understanding of ABC drug transporters has also been useful in explaining the mechanisms of MDR. The recent findings suggested that numerous signaling pathways such as protein tyrosine kinase, EGFR (epidermal growth factor receptor), MAPK (mitogen-activated protein kinase) are involved in drug resistance. Therefore, a multidisciplinary approach with a combination of technologies (genomics, epigenetics, transcriptomics, proteomics) would be a classical model in changing our therapeutic thinking and diagnosis against cancer. The ongoing research in this area is at a very early stage about the integration of these studies into everyday practice. So, the development of suitable markers for host responses will be a key factor in this research.
Acknowledgments
The authors acknowledge the support received from the Central University of Punjab, Bhatinda, India, in writing this manuscript. PS and VA are grateful to CSIR, New Delhi, India for the award of the fellowship.
References
- 1.Deininger MW. Optimizing therapy of chronic myeloid leukemia. Exper Hematol 2007;35:144-54. [DOI] [PubMed] [Google Scholar]
- 2.Karimiani EG, Marriage F, Merritt AJ, et al. Single-cell analysis of K562 cells: an imatinib-resistant subpopulation is adherent and has upregulated expression of BCR-ABL mRNA and protein. Exper Hematol 2014;42:183-91. [DOI] [PubMed] [Google Scholar]
- 3.Verma M, Karimiani EG, Byers RJ, et al. Mathematical modelling of miRNA mediated BCR. ABL protein regulation in chronic myeloid leukaemia vis-a-vis therapeutic strategies. Integrat Biol 2013;5:543-54. [DOI] [PubMed] [Google Scholar]
- 4.Chereda B, Melo JV. The biology and pathogenesis of chronic myeloid leukemia. Chron Myeloid Leuk 2016:17-39. [Google Scholar]
- 5.Markose P, Chendamarai E, Balasubramanian P, et al. Spectrum of BCR-ABL kinase domain mutations in patients with chronic myeloid leukemia from India with suspected resistance to imatinib-mutations are rare and have different distributions. Leuk Lymph 2009;50:2092-5. [DOI] [PubMed] [Google Scholar]
- 6.Wieczorek A, Uharek L. Management of chronic myeloid leukemia patients resistant to tyrosine kinase inhibitors treatment. Biomark Insights 2015;10:BMI-S22431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Roskoski JrR. STI-571: an anticancer protein-tyrosine kinase inhibitor. Biochem Biophys Res Commun 2003;309:709-17. [DOI] [PubMed] [Google Scholar]
- 8.Walz C, Sattler M. Novel targeted therapies to overcome imatinib mesylate resistance in chronic myeloid leukemia (CML). Crit Rev Oncol/Hematol 2006;57:145-64. [DOI] [PubMed] [Google Scholar]
- 9.Sawyers CL. Disabling Abl-Perspectives on Abl kinase regulation and cancer therapeutics. Cancer Cell 2002;1:13-5. [DOI] [PubMed] [Google Scholar]
- 10.Shah NP, Sawyers CL. Mechanisms of resistance to STI571 in Philadelphia chromosome-associated leukemias. Oncogene 2003;22:7389-95. [DOI] [PubMed] [Google Scholar]
- 11.Dusetzina SB, Winn AN, Abel GA, et al. Cost sharing and adherence to tyrosine kinase inhibitors for patients with chronic myeloid leukemia. J Clin Oncol 2014;32:306-11. [DOI] [PubMed] [Google Scholar]
- 12.Gupta SK, Singh P, Ali V, Verma M. Therapeutic approaches to enhance the BCR-ABL tyrosine kinase inhibitors efficacy in chronic myeloid leukemia. Asian Pac J Health Sci 2019;6:58-69. [Google Scholar]
- 13.Kantarjian H, Sawyers C, Hochhaus A, et al. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. N Engl J Med 2002;346:645-52. [DOI] [PubMed] [Google Scholar]
- 14.O'brien SG, Guilhot F, Larson RA, et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med 2003;348:994-1004. [DOI] [PubMed] [Google Scholar]
- 15.Szakacs G, Paterson JK, Ludwig JA, et al. Targeting multidrug resistance in cancer. Nature Rev Drug Discov 2006;5:219. [DOI] [PubMed] [Google Scholar]
- 16.Dano K. Cross resistance between vinca alkaloids and anthracyclines in Ehrlich ascites tumor in vivo. Cancer Chem Rep 1972;56:701-8. [PubMed] [Google Scholar]
- 17.Baguley BC. Multiple drug resistance mechanisms in cancer. Mol Biotechnol 2010;46:308-16. [DOI] [PubMed] [Google Scholar]
- 18.Fojo T, Menefee M. Mechanisms of multidrug resistance: the potential role of microtubule-stabilizing agents. Ann Oncol 2007;18:3-8. [DOI] [PubMed] [Google Scholar]
- 19.Lage H. An overview of cancer multidrug resistance: a still unsolved problem. Cell Mol Life Sci 2008;65:3145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gillet JP, Gottesman MM. Mechanisms of multidrug resistance in cancer. Methods Mol Biol 2010;596:47-76. [DOI] [PubMed] [Google Scholar]
- 21.Rochat B. Importance of influx and efflux systems and xenobiotic metabolizing enzymes in intratumoral disposition of anticancer agents. Curr Cancer Drug Targets 2009;9:652-74. [DOI] [PubMed] [Google Scholar]
- 22.Konig J, Muller F, Fromm MF. Transporters and drug-drug interactions: important determinants of drug disposition and effects. Pharmacol Rev 2013;65:944-66. [DOI] [PubMed] [Google Scholar]
- 23.Levatic J, Curak J, Kralj M, et al. Accurate models for P-gp drug recognition induced from a cancer cell line cytotoxicity screen. J Med Chem 2013;56:5691-708. [DOI] [PubMed] [Google Scholar]
- 24.Li S, Zhang W, Yin X, et al. Mouse ATP-binding cassette (ABC) transporters conferring multi-drug resistance. Anti- Cancer Agents Med Chem 2015;15:423-32. [PubMed] [Google Scholar]
- 25.Ter Beek J, Guskov A, Slotboom DJ. Structural diversity of ABC transporters. J General Physiol 2014;143:419-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kumar P, Zhang DM, Degenhardt K, Chen ZS. Autophagy and transporter-based multi-drug resistance. Cells 2012;1:558-75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ween MP, Armstrong MA, Oehler MK, Ricciardelli C. The role of ABC transporters in ovarian cancer progression and chemoresistance. Crit Rev Oncol/Hematol 2015;96:220-56. [DOI] [PubMed] [Google Scholar]
- 28.Vasiliou V, Vasiliou K, Nebert DW. Human ATP-binding cassette (ABC) transporter family. Human Genom 2009;3:281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Wu CP, Hsieh CH, Wu YS. The emergence of drug transporter- mediated multidrug resistance to cancer chemotherapy. Mol Pharm 2011;8:1996-2011. [DOI] [PubMed] [Google Scholar]
- 30.Rice AJ, Park A, Pinkett HW. Diversity in ABC transporters: type I, II and III importers. Crit Rev Biochem Mol Biol 2014;49:426-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kunjachan S, Rychlik B, Storm G, et al. Multidrug resistance: Physiological principles and nanomedical solutions. Adv Drug Deliv Rev 2013;65:1852-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hee Choi Y, Yu AM. ABC transporters in multidrug resistance and pharmacokinetics, and strategies for drug development. Curr Pharm Design 2014;20:793-807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Shapira A, Livney YD, Broxterman HJ, Assaraf YG. Nanomedicine for targeted cancer therapy: towards the overcoming of drug resistance. Drug Resist Updates 2011;14:150-63. [DOI] [PubMed] [Google Scholar]
- 34.Chen KG, Sikic BI. Molecular pathways: regulation and therapeutic implications of multidrug resistance. Clin Cancer Res 2012;18:1863-69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Breier A, Gibalova L, Seres M, et al. New insight into p-glycoprotein as a drug target. Anti-Cancer Agents Med Chem 2013;13:159-70. [PubMed] [Google Scholar]
- 36.Videira M, Reis RL, Brito MA. Deconstructing breast cancer cell biology and the mechanisms of multidrug resistance. Biochim Biophys Acta Rev Cancer 2014;1846:312-25. [DOI] [PubMed] [Google Scholar]
- 37.Glavinas H, Krajcsi P, Cserepes J, Sarkadi B. The role of ABC transporters in drug resistance, metabolism and toxicity. Curr Drug Deliv 2004;1:27-42. [DOI] [PubMed] [Google Scholar]
- 38.Sharom FJ. ABC multidrug transporters: structure, function and role in chemoresistance. Pharmacogenomics 2008;9:105-27. [DOI] [PubMed] [Google Scholar]
- 39.Huisman MT, Smit JW, Schinkel AH. Significance of P-glycoprotein for the pharmacology and clinical use of HIV protease inhibitors. Aids 2000;14:237-42. [DOI] [PubMed] [Google Scholar]
- 40.Gottesman MM, Ling V. The molecular basis of multidrug resistance in cancer: The early years of P‐glycoprotein research. FEBS Lett 2006;580:998-1009. [DOI] [PubMed] [Google Scholar]
- 41.Loo TW, Bartlett MC, Clarke DM. Human P-glycoprotein contains a greasy ball-and-socket joint at the second transmission interface. J Biol Chem 2013;288:20326-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Borst P, Evers R, Kool M, Wijnholds J. A family of drug transporters: the multidrug resistance-associated proteins. J Natl Cancer Inst 2000;92:1295-302. [DOI] [PubMed] [Google Scholar]
- 43.Matheny CJ, Lamb MW, Brouwer KL, Pollack GM. Pharmacokinetic and pharmacodynamic implications of P‐glycoprotein modulation. Pharmacother J Hum Pharmacol Drug Ther 2001;21:778-96. [DOI] [PubMed] [Google Scholar]
- 44.Kim RB. Drugs as P-glycoprotein substrates, inhibitors, and inducers. Drug Metab Rev 2002;34:47-54. [DOI] [PubMed] [Google Scholar]
- 45.Schinkel AH, Jonker JW. Mammalian drug efflux transporters of the ATP binding cassette (ABC) family: an overview. Adv Drug Deliv Revi 2003;55:3-29. [DOI] [PubMed] [Google Scholar]
- 46.Choudhuri S, Klaassen CD. Structure, function, expression, genomic organization, and single nucleotide polymorphisms of human ABCB1 (MDR1), ABCC (MRP), and ABCG2 (BCRP) efflux transporters. Int J Toxicol 2006;25:231-59. [DOI] [PubMed] [Google Scholar]
- 47.Jedlitschky G, Hoffmann U, Kroemer HK. Structure and function of the MRP2 (ABCC2) protein and its role in drug disposition. Exp Opin Drug Metab Toxicol 2006;2:351-66. [DOI] [PubMed] [Google Scholar]
- 48.Nies AT, Schwab M, Keppler D. Interplay of conjugating enzymes with OATP uptake transporters and ABCC/MRP efflux pumps in the elimination of drugs. Exp Opin Drug Metab Toxicol 2008;4:545-68. [DOI] [PubMed] [Google Scholar]
- 49.Zhou SF, Wang LL, Di YM, et al. Substrates and inhibitors of human multidrug resistance associated proteins and the implications in drug development. Curr Med Chem 2008;15:1981-39. [DOI] [PubMed] [Google Scholar]
- 50.Sharom FJ. The P-glycoprotein multidrug transporter. Essays Biochem 2011;50:161-78. [DOI] [PubMed] [Google Scholar]
- 51.Slot AJ, Molinski SV, Cole SP. Mammalian multidrug-resistance proteins (MRPs). Essays in Biochem 2011;50:179-207. [DOI] [PubMed] [Google Scholar]
- 52.Brand W, Oosterhuis B, Krajcsi P, et al. Interaction of hesperetin glucuronide conjugates with human BCRP, MRP2 and MRP3 as detected in membrane vesicles of overexpressing baculovirus‐infected Sf9 cells. Biopharm Drug Dispos 2011;32:530-5. [DOI] [PubMed] [Google Scholar]
- 53.Morrissey KM, Wen CC, Johns SJ, et al. The UCSF‐FDA TransPortal: a public drug transporter database. Clin Pharmacol Therap 2012;92:545-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Drozdzik M, Gröer C, Penski J, et al. Protein abundance of clinically relevant multidrug transporters along the entire length of the human intestine. Mol Pharm 2014;11:3547-55. [DOI] [PubMed] [Google Scholar]
- 55.Gröer C, Brück S, Lai Y., et al. Oswald S. LC-MS/MS-based quantification of clinically relevant intestinal uptake and efflux transporter proteins. J Pharm Biomed Analysis 2013;85:253-61. [DOI] [PubMed] [Google Scholar]
- 56.Buchler M, Konig J, Brom M, et al. cDNA cloning of the hepatocyte canalicular isoform of the multidrug resistance protein, cMrp, reveals a novel conjugate export pump deficient in hyperbilirubinemic mutant rats. J Biol Chem 1996;271:15091-8. [DOI] [PubMed] [Google Scholar]
- 57.Paulusma CC, Bosma PJ, Zaman GJ, et al. Congenital jaundice in rats with a mutation in a multidrug resistance-associated protein gene. Science 1996;271:1126-28. [DOI] [PubMed] [Google Scholar]
- 58.Nies AT, Keppler D. The apical conjugate efflux pump ABCC2 (MRP2). Pflugers Archiv-Eur J Physiol 2007;453:643-59. [DOI] [PubMed] [Google Scholar]
- 59.Jemnitz K, Heredi-Szabo K, Janossy J, et al. ABCC2/Abcc2: a multispecific transporter with dominant excretory functions. Drug Metab Rev 2010;42:402-36. [DOI] [PubMed] [Google Scholar]
- 60.Rost D, Konig J, Weiss G, et al. Expression and localization of the multidrug resistance proteins MRP2 and MRP3 in human gallbladder epithelia. Gastroenterology 2001;121: 1203-8. [DOI] [PubMed] [Google Scholar]
- 61.St-Pierre MV, Serrano MA, Macias RIR, et al. Expression of members of the multidrug resistance protein family in human term placenta. Am J Physiol-Regul Integrat Comparat Physiol 2000;279:1495-03. [DOI] [PubMed] [Google Scholar]
- 62.Prouillac C, Videmann B, Mazallon M, Lecoeur S. Induction of cells differentiation and ABC transporters expression by a myco-estrogen, zearalenone, in human choriocarcinoma cell line (BeWo). Toxicology 2009;263:100-7. [DOI] [PubMed] [Google Scholar]
- 63.Janneh O, Jones E, Chandler B, et al. Inhibition of P-glycoprotein and multidrug resistance-associated proteins modulates the intracellular concentration of lopinavir in cultured CD4 T cells and primary human lymphocytes. J Antimicrob Chemother 2007;60:987-93. [DOI] [PubMed] [Google Scholar]
- 64.Korita PV, Wakai T, Shirai Y, et al. Multidrug resistance-associated protein 2 determines the efficacy of cisplatin in patients with hepatocellular carcinoma. Oncol Rep 2010;23:965-72. [DOI] [PubMed] [Google Scholar]
- 65.Konig J, Nies AT, Cui Y, et al. Conjugate export pumps of the multidrug resistance protein (MRP) family: localization, substrate specificity, and MRP2-mediated drug resistance. Biochim Biophys Acta (BBA)-Biomembr 1999;1461:377-94. [DOI] [PubMed] [Google Scholar]
- 66.Cantz T, Nies AT, Brom M, et al. MRP2, a human conjugate export pump, is present and transports fluo 3 into apical vacuoles of Hep G2 cells. Am J Physiol-Gastrointest Liver Physiol 2000;278:522-G531. [DOI] [PubMed] [Google Scholar]
- 67.Evers R, De Haas M, Sparidans R, et al. Vinblastine and sulfinpyrazone export by the multidrug resistance protein MRP2 is associated with glutathione export. Br J Cancer 2000;83:375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Bark H, Choi CH. PSC833, cyclosporine analogue, downregulates MDR1 expression by activating JNK/c-Jun/AP-1 and suppressing NF-κB. Cancer Chemother Pharmacol 2010;65:1131-6. [DOI] [PubMed] [Google Scholar]
- 69.Jani M, Ambrus C, Magnan R, et al. Structure and function of BCRP, a broad specificity transporter of xenobiotics and endobiotics. Archiv Toxicol 2014;88:1205-48. [DOI] [PubMed] [Google Scholar]
- 70.Mo W, Zhang JT. Human ABCG2: structure, function, and its role in multidrug resistance. Int J Biochem Mol Biol 2012;3:1-27. [PMC free article] [PubMed] [Google Scholar]
- 71.Jiao X, Zhao L, Ma M, et al. MiR-181a enhances drug sensitivity in mitoxantone-resistant breast cancer cells by targeting breast cancer resistance protein (BCRP/ABCG2). Breast Cancer Res Treat 2013;139:717-30. [DOI] [PubMed] [Google Scholar]
- 72.Ma MT, He M, Wang Y, et al. MiR-487a resensitizes mitoxantrone (MX)-resistant breast cancer cells (MCF-7/MX) to MX by targeting breast cancer resistance protein (BCRP/ABCG2). Cancer Lett 2013;339:107-15. [DOI] [PubMed] [Google Scholar]
- 73.Wang H, Wang X, Hu R, et al. Methylation of SFRP5 is related to multidrug resistance in leukemia cells. Cancer Gene Ther 2014;21:83-9. [DOI] [PubMed] [Google Scholar]
- 74.Mao Q, Unadkat JD. Role of the breast cancer resistance protein (BCRP/ABCG2) in drug transport-an update. AAPS J 2015;17:65-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Silva R, Vilas-Boas V, Carmo H, et al. Modulation of P-glycoprotein efflux pump: induction and activation as a therapeutic strategy. Pharmacol Ther 2015;149:1-123. [DOI] [PubMed] [Google Scholar]
- 76.Abdallah HM, Al-Abd AM, El-Dine RS, El-Halawany AM. Pglycoprotein inhibitors of natural origin as potential tumor chemo-sensitizers: A review. J Adv Res 2015;6:45-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Cherigo L, Lopez D, Martinez-Luis S. Marine natural products as breast cancer resistance protein inhibitors. Marine Drugs 2015;13:2010-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Binkhathlan Z, Shayeganpour A, Brocks DR, Lavasanifar A. Encapsulation of P-glycoprotein inhibitors by polymeric micelles can reduce their pharmacokinetic interactions with doxorubicin. Eur J Pharm Biopharm 2012;81:142-8. [DOI] [PubMed] [Google Scholar]
- 79.O'brien MM, Lacayo NJ, Lum BL, et al. Phase I study of valspodar (PSC‐833) with mitoxantrone and etoposide in refractory and relapsed pediatric acute leukemia: A report from the Children's Oncology Group. Pediatr Blood Cancer 2010;54:694-702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kolitz JE, George SL, Marcucci G, et al. P-glycoprotein inhibition using valspodar (PSC-833) does not improve outcomes for patients younger than age 60 years with newly diagnosed acute myeloid leukemia: Cancer and Leukemia Group B study 19808. Blood 2010;116:1413-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Cripe LD, Uno H, Paietta EM, et al. Zosuquidar, a novel modulator of P-glycoprotein, does not improve the outcome of older patients with newly diagnosed acute myeloid leukemia: a randomized, placebo-controlled trial of the Eastern Cooperative Oncology Group 3999. Blood 2010;116:4077-85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Kelly RJ, Draper D, Chen CC, et al. A pharmacodynamic study of docetaxel in combination with the P-glycoprotein antagonist tariquidar (XR9576) in patients with lung, ovarian, and cervical cancer. Clin Cancer Res 2011;17:569-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Livney YD, Assaraf YG. Rationally designed nanovehicles to overcome cancer chemoresistance. Adv Drug Deliv Rev 2013;65:1716-30. [DOI] [PubMed] [Google Scholar]
- 84.Shapira A, Davidson I, Avni N, et al. β-Casein nanoparticlebased oral drug delivery system for potential treatment of gastric carcinoma: Stability, target-activated release and cytotoxicity. Eur J Pharm Biopharm 2012;80:298-305. [DOI] [PubMed] [Google Scholar]
- 85.Tiram G, Segal E, Krivitsky A, et al. Identification of dormancy- associated microRNAs for the design of osteosarcoma-targeted dendritic polyglycerol nanopolyplexes. ACS Nano 2016;10:2028-45. [DOI] [PubMed] [Google Scholar]
- 86.Shukla S, Chen ZS, Ambudkar SV. Tyrosine kinase inhibitors as modulators of ABC transporter-mediated drug resistance. Drug Resist Updates 2012;15:70-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Tinsley HN, Gary BD, Thaiparambil J, et al. Colon tumor cell growth inhibitory activity of sulindac sulfide and other NSAIDs is associated with PDE5 inhibition. Cancer Prevent Res 2010;3:1303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Krishna R, Mayer LD. Multidrug resistance (MDR) in cancer: mechanisms, reversal using modulators of MDR and the role of MDR modulators in influencing the pharmacokinetics of anticancer drugs. Eur J Pharm Sci 2000;11:265-83. [DOI] [PubMed] [Google Scholar]
- 89.te Boekhorst PA, van Kapel J, Schoester M, Sonneveld P. Reversal of typical multidrug resistance by cyclosporin and its non-immunosuppressive analogue SDZ PSC 833 in Chinese hamster ovary cells expressing themdr1 phenotype. Cancer Chemother Pharmacol 1992;30:238-42. [DOI] [PubMed] [Google Scholar]
- 90.Twentyman PR, Bleehen NM. Resistance modification by PSC-833, a novel non-immunosuppressive cyclosporin A. Eur J Cancer Clin Oncol 1991;27:1639-42. [DOI] [PubMed] [Google Scholar]
- 91.Thomas H, Coley HM. Overcoming multidrug resistance in cancer: an update on the clinical strategy of inhibiting p-glycoprotein. Cancer Control 2003;10:159-65. [DOI] [PubMed] [Google Scholar]
- 92.Altstadt TJ, Fairchild CR, Golik J, et al. Synthesis and antitumor activity of novel C-7 paclitaxel ethers: discovery of BMS-184476. J Med Chem 2001;44:4577-83. [DOI] [PubMed] [Google Scholar]
- 93.Cassinelli G, Lanzi C, Supino R, et al. Cellular bases of the antitumor activity of the novel taxane IDN 5109 (BAY59-8862) on hormone-refractory prostate cancer. Clin Cancer Res 2002;8:2647-54. [PubMed] [Google Scholar]
- 94.Shionoya M, Jimbo T, Kitagawa M, et al. DJ‐927, a novel oral taxane, overcomes P‐glycoprotein‐mediated multidrug resistance in vitro and in vivo. Cancer Sci 2003;94:459-66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Su Y, Cheng X, Tan Y, et al. Synthesis of a dual functional anti-MDR tumor agent PH II-7 with elucidations of antitumor effects and mechanisms. PLoS One 2012;7:32782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Fox E, Bates SE. Tariquidar (XR9576): a P-glycoprotein drug efflux pump inhibitor. Exp Rev Anticancer Ther 2007;7:447-59. [DOI] [PubMed] [Google Scholar]
- 97.Kemper EM, Cleypool C, Boogerd W, et al. The influence of the P-glycoprotein inhibitor zosuquidar trihydrochloride (LY335979) on the brain penetration of paclitaxel in mice. Cancer Chemother Pharmacol 2004;53:173-8. [DOI] [PubMed] [Google Scholar]
- 98.Luurtsema G, Schuit RC, Klok RP, et al. Evaluation of [11C] laniquidar as a tracer of P-glycoprotein: radiosynthesis and biodistribution in rats. Nucl Med Biol 2009;36:643-9. [DOI] [PubMed] [Google Scholar]
- 99.Dörner B, Kuntner C, Bankstahl JP, et al. Synthesis and smallanimal positron emission tomography evaluation of [11C]- elacridar as a radiotracer to assess the distribution of P-glycoprotein at the blood-brain barrier. J Med Chem 2009;52:6073-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Lubelski J, van Merkerk R, Konings WN, Driessen AJ. Nucleotide-binding sites of the heterodimeric LmrCD ABCmultidrug transporter of Lactococcus lactis are asymmetric. Biochem 2006;45:648-56. [DOI] [PubMed] [Google Scholar]
- 101.Tang R, Faussat AM, Perrot JY, et al. Zosuquidar restores drug sensitivity in P-glycoprotein expressing acute myeloid leukemia (AML). BMC Cancer 2008;8:51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Li X, Gao L, Cui Q, et al. Sulindac inhibits tumor cell invasion by suppressing NF-κB-mediated transcription of microRNAs. Oncogene 2012;31:4979-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Shen H, Liu T, Fu L, et al. Identification of microRNAs involved in dexamethasone-induced muscle atrophy. Mol Cell Biochem 2013;381:105-13. [DOI] [PubMed] [Google Scholar]
- 104.To KK. MicroRNA: a prognostic biomarker and a possible druggable target for circumventing multidrug resistance in cancer chemotherapy. J Biomed Sci 2013;20:99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Bao L, Hazari S, Mehra S, et al. Increased expression of Pglycoprotein and doxorubicin chemoresistance of metastatic breast cancer is regulated by miR-298. Am J Pathol 2012;180:2490-2503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Hong L, Han Y, Zhang H, et al. The prognostic and chemotherapeutic value of miR-296 in esophageal squamous cell carcinoma. Ann Surg 2010;251:1056-63. [DOI] [PubMed] [Google Scholar]
- 107.Zhu H, Wu H, Liu X, et al. Role of MicroRNA miR-27a and miR-451 in the regulation of MDR1/P-glycoprotein expression in human cancer cells. Biochem Pharmacol 2008;76:582-88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Dong WH, Li Q, Zhang XY, et al. Deep sequencing identifies deregulation of microRNAs involved with vincristine drugresistance of colon cancer cells. Int J Clin Exper Pathol 2015;8:11524-30. [PMC free article] [PubMed] [Google Scholar]
- 109.Fisher M, Abramov M, Van Aerschot A, et al. Inhibition of MDR1 expression with altritol-modified siRNAs. Nucl Acids Res 2007;35:1064-74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Li H, Zhou S, Li T, et al. Suppression of BCRP expression and restoration of sensitivity to chemotherapy in multidrug-resistant HCC cell line HEPG2/ADM by RNA interference. Hepato-gastroenterol 2012;59:2238-42. [DOI] [PubMed] [Google Scholar]
- 111.Perez J, Bardin C, Rigal C, et al. Anti-MDR1 siRNA restores chemosensitivity in chemoresistant breast carcinoma and osteosarcoma cell lines. Anticancer Res 2011;31:2813-20. [PubMed] [Google Scholar]
- 112.Wu H, Hait WN, Yang JM. Small interfering RNA-induced suppression of MDR1 (P-glycoprotein) restores sensitivity to multidrug-resistant cancer cells. Cancer Res 2003;63:1515-19. [PubMed] [Google Scholar]
- 113.Broxterman HJ, Kuiper CM, Schuurhuis GJ, et al. Increase of daunorubicin and vincristine accumulation in multidrug resistant human ovarian carcinoma cells by a monoclonal antibody reacting with P-glycoprotein. Biochem Pharmacol 1988;37:2389-93. [DOI] [PubMed] [Google Scholar]
- 114.Hamada H, Tsuruo T. Functional role for the 170-to 180-kDa glycoprotein specific to drug-resistant tumor cells as revealed by monoclonal antibodies. Proc Natl Acad Sci 1986;83:7785-89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Pearson JW, Fogler WE, Volker K, et al. Reversal of drug resistance in a human colon cancer xenograft expressing MDR1 complementary DNA by in vivo administration of MRK-16 monoclonal antibody. JNCI J Natl Cancer Inst 1991;83:1386-91. [DOI] [PubMed] [Google Scholar]
- 116.Tsuruo T, Hamada H, Sato S, Heike Y. Inhibition of multidrug‐ resistant human tumor growth in athymic mice by anti‐P‐glycoprotein monoclonal antibodies. Japan J Cancer Res 1989;80:627-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Mechetner EB, Roninson IB. Efficient inhibition of P-glycoprotein- mediated multidrug resistance with a monoclonal antibody. Proc Natl Acad Sci 1992;89:5824-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Bar-Zeev M, Assaraf YG, Livney YD. β-casein nanovehicles for oral delivery of chemotherapeutic drug combinations overcoming P-glycoprotein-mediated multidrug resistance in human gastric cancer cells. Oncotarget 2016;7:23322-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Zhang M, Akbulut M. Adsorption, desorption, and removal of polymeric nanomedicine on and from cellulose surfaces: effect of size. Langmuir 2011;27:12550-9. [DOI] [PubMed] [Google Scholar]
- 120.Sun T, Zhang YS, Pang B, et al. Engineered nanoparticles for drug delivery in cancer therapy. Angewandte Chemie Int 2014;53:12320-64. [DOI] [PubMed] [Google Scholar]
- 121.Yang L, Li W, Kirberger M, et al. Design of nanomaterialbased systems for novel vaccine development. Biomat Sci 2016;4:785-802. [DOI] [PubMed] [Google Scholar]
- 122.Cho K, Wang XU, Nie S, Shin DM. Therapeutic nanoparticles for drug delivery in cancer. Clin Cancer Res 2008;14:1310-6. [DOI] [PubMed] [Google Scholar]
- 123.Zhang TT, Li W, Meng G, et al. Strategies for transporting nanoparticles across the blood–brain barrier. Biomat Sci 2016;4:219-29. [DOI] [PubMed] [Google Scholar]
- 124.Devulapally R, Paulmurugan R. Polymer nanoparticles for drug and small silencing RNA delivery to treat cancers of different phenotypes. Wiley Interdiscipl Rev Nanomed Nanobiotechnol 2014;6:40-60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Li Z, Qiu L, Chen Q, et al. pH-sensitive nanoparticles of poly (L-histidine)–poly (lactide-co-glycolide)–tocopheryl polyethylene glycol succinate for anti-tumor drug delivery. Acta Biomater 2015;11:137-50. [DOI] [PubMed] [Google Scholar]
- 126.Park JW. Liposome-based drug delivery in breast cancer treatment. Breast Cancer Res 2002;4:95-99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Wu X, He X, Wang K, et al. Ultrasmall near-infrared gold nanoclusters for tumor fluorescence imaging in vivo. Nanoscale 2010;2:2244-9. [DOI] [PubMed] [Google Scholar]
- 128.Yang L, Zhang X, Ye M, et al. Aptamer-conjugated nanomaterials and their applications. Adv Drug Deliv Rev 2011;63: 1361-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Lee SM, Kim HJ, Kim SY, et al. Drug-loaded gold plasmonic nanoparticles for treatment of multidrug resistance in cancer. Biomaterials 2014;35:2272-82. [DOI] [PubMed] [Google Scholar]