Abstract
Multidrug resistance (MDR), which significantly decreases the efficacy of anticancer drugs and causes tumor recurrence, has been a major challenge in clinical cancer treatment with chemotherapeutic drugs for decades. Several mechanisms of overcoming drug resistance have been postulated. Well known P-glycoprotein (P-gp) and other drug efflux transporters are considered to be critical in pumping anticancer drugs out of cells and causing chemotherapy failure. Innovative theranostic (therapeutic and diagnostic) strategies with nanoparticles are rapidly evolving and are anticipated to offer opportunities to overcome these limits. In this review, we discuss the mechanisms of drug efflux-mediated resistance and the application of multiple nanoparticle-based platforms to overcome chemoresistance and improve therapeutic outcome.
Keywords: multidrug resistance, drug efflux transporter, cancer nanotechnology
According to the Global Cancer Report issued by the World Health Organization (WHO), there are over 10 million new cases of cancer each year and over 7.9 million annual deaths from the disease[1]. Serious toxicity is a critical problem for effective chemotherapy because most anticancer agents lack selective efficacy in tumors. Another vital issue is the development of tumor resistance to conventional chemotherapy. Drug resistance, which allows tumors to evade chemotherapeutic agents, has emerged as a major obstacle that limits the efficacy of chemotherapy. The formidable side effects of individual or combined anticancer agents as well as the treatment history and clinical status of the patient must be considered to circumvent the tumor resistance to chemotherapy in clinic. Tumors generally develop significant resistance to repeated treatment with one kind of anticancer agent and then often become resistant to similar or completely different drugs. This mechanism for tumor survival under chemotherapeutic treatment is known as multidrug resistance (MDR). MDR can be intrinsic or acquired through chemotherapeutic drug exposure, and multiple mechanisms are likely to contribute to clinical MDR. Historically, the most significant discovery about MDR was the identification of P-glycoprotein (P-gp)[2], which is overexpressed on the plasma membrane of cancer cells with MDR. Following P-gp, other transporters, such as multidrug resistance-associated protein 1 (MRP1)[3] and multixenobiotic resistance (MXR)[4], are also recognized to relate with drug efflux. In recent years, applications of nanotechnology have shown great promise, with several kinds of nanomedicine entering clinical studies. Although the feasibility and efficacy of reversing drug resistance has been confirmed in vitro and in vivo, the mechanisms by which to use nanotechnology to circumvent this phenotype have not been clarified or fully explored.
This article introduces nanotechnology-based formulations and possible nanomedical approaches to address MDR in tumors, with a specific focus on the use of nanotechnology in cancer to overcome drug efflux-mediated resistance.
Possible Mechanisms of Drug Efflux-mediated Resistance in Cancer
The changes that drive antitumor drug resistance include the following: increased activity of drug efflux pump, such as the ATP-binding cassette (ABC) superfamily; decreased drug influx; activation of DNA repair; metabolic modification or detoxification; and altered expression of apoptosis-associated protein Bcl-2[5] and tumor suppressor protein p53[6],[7]. Of these mechanisms, overexpression of ABC transporters is the most frequent. ABC transport molecules are generally expressed on the plasma membrane and on the membranes of cellular vesicles, and they play vital physiologic functions and also affect the pharmacokinetic properties of chemotherapeutics in humans. ABC transporters are transmembrane proteins that use the energy of ATP hydrolysis to shuttle various substrates across the cell membrane. To date, there are 48 known transporters in the ABC family, which are classified into seven different subfamilies (ABC A through ABC G)[8]−[11] (Table 1). Thirteen ABC transporters contribute to tumor MDR, including P−gp (MDR1/ABCBI), multidrug resistance proteins (MRPs/ABCCs), and breast cancer resistance protein (BCRP/ABCG2), all of which are the most characterized ABC transporters[12]. The normal function of ABC transporters as pumps is to extrude toxins and foreign substances out of the cell. P−gp is the best−known membrane pump molecule of the ABC transporters involved in MDR. Human P−gp, a 170 kDa membrane−associated protein containing 1280 amino acids, is able to carry out an ATP−dependent conformational change that moves the intracellular substrates to the exterior of the cell[2]. It can transport a broad range of structurally related or unrelated compounds including anticancer drugs out of cells and thereby decrease intracellular accumulation of these compounds. Therefore, P−gp expression can physiologically prevent cytotoxic compounds by pump them out of cells to reduce their intracellular concentration. In patients with tumors, P−gp can efflux various anticancer drugs such as doxorubicin [13]–[15] and paclitaxel[16],[17] out of cancer cells. Overexpression of P−gp is a common feature of most acquired MDR in solid tumors[18],[19]. Other transporters such as MRP1, MRP2, and BCRP also contribute to drug distribution in the human body in cases where P−gp expression is not significantly altered after treatment.
Table 1. Demographic and clinical characteristics of the studied population.
| Gene | Alias | Subfamily | Location | Gene size (bp) | dN/dS |
Transcript | Protein | Main tissue expression | Function | Disease | ||
| Dog | Mouse | Rat | ||||||||||
| ABCA1 | ABC1 | ABC1 | 9q31.1 | 147154 | 0.078 | 0.048 | 0.072 | 1 | 1 | Ubiquitous | Cholesterol efflux onto HDL drug resistance | Tangier's disease, familial hypoapo-proteinemia |
| ABCA2 | ABC2 | ABC1 | 9q34.3 | 21689 | 0.043 | 0.030 | 2 | 2 | Brain | Drug resistance | ||
| ABCA3 | ABC3, ABCC | ABC1 | 16p13.3 | 53974 | 0.072 | 0.069 | 0.072 | 1 | 1 | Lung | Phospholipid metabolism | Surfactant deficiency in newborns |
| ABCA4 | ABCR | ABC1 | 1p21.3 | 128287 | 0.151 | 0.128 | 0.126 | 1 | 1 | Photoreceptors | N-retinylidene-PE efflux | Stargardt's disease/fundusflavimaculatis, retinitis pigmentosa, cone-rod dystrpohy, age-related macular degeneration |
| ABCA5 | ABC1 | 17q24.3 | 80499 | 0.132 | 0.091 | 0.087 | 2 | 2 | Muscle, heart, testes | |||
| ABCA6 | ABC1 | 17q24.3 | 63169 | 0.356 | 0.282 | 1 | 1 | Liver | ||||
| ABCA7 | ABC1 | 19p13.3 | 10347 | 0.118 | 0.130 | 2 | 2 | Spleen, thymus | ||||
| ABCA8 | ABC1 | 17q24.3 | 88101 | 0.256 | 0.256 | 0.298 | 1 | 1 | Ovary, heart, skeletal muscle, liver | |||
| ABCA9 | ABC1 | 17q24.3 | 86155 | 0.239 | 0.245 | 0.271 | 2 | 2 | Heart | |||
| ABCA10 | ABC1 | 17q24.3 | 96808 | NA | NA | NA | 1 | 1 | Muscle, heart | |||
| ABCA12 | ABC1 | 2q34 | 206885 | 0.113 | 0.075 | 0.101 | 2 | 2 | Stomach | Glucosylceramide, other epidermal lipids | Lamellar ichthyosis type 2 (mild); Harlequin ichthyosis (severe) | |
| ABCA13 | ABC1 | 7p12.3 | 449249 | 0.397 | 0.221 | 0.218 | 1 | 1 | Low in all tissues | |||
| ABCBI | PGY1, MDR, P-gp | MDR | 7p21.12 | 209617 | 0.180 | 0.16 | 0.145 | 1 | 1 | Adrenal, kidney, brain, liver, intestine, testis, gland, uterus, ovary | Multidrug resistance | Ivermectin sensitivity, digoxin uptake |
| ABCB2 | TAP1 | MDR | 6p21 | 8765 | 0.016 | 0.031 | 0.026 | 1 | 1 | All cell | Peptide transport | Immune deficiency |
| ABCB3 | TAP2 | MDR | 6p21 | 16912 | 0.225 | 0.204 | 0.207 | 2 | 2 | All cell | Peptide transport | Immune deficiency |
| ABCB4 | PGY3 or MDR3 | MDR | 7q21.12 | 75506 | 0.152 | 0.097 | 0.091 | 3 | 3 | Liver | PC transport | Progressive familial intrahepatic cholestasis-3, intrahepatic cholestasis of pregnancy |
| ABCB5 | MDR | 7p21.1 | 108203 | 0.283 | 0.212 | 0.197 | 1 | 1 | Ubiquitous | |||
| ABCB6 | MTABC3 | MDR | 2q35 | 9179 | 0.201 | 0.134 | 0.156 | 1 | 1 | Mitochondria | Iron transport | Unknown |
| ABCB7 | ABC7 | MDR | Xq21-22 | 103026 | 0.215 | 0.11 | 0.059 | 1 | 1 | Mitochondria | Fe/S cluster transport | X-linked sideroblastosis and anemia |
| ABCB8 | MABC1 | MDR | 7q36.1 | 17116 | 0.135 | 0.137 | 0.150 | 1 | 1 | Mitochondria | ||
| ABCB9 | MDR | 12q24.31 | 46214 | 0.045 | 0.036 | 0.037 | 4 | 3 | Liver | |||
| ABCB10 | MTABC2 | MDR | 1q42.13 | 42113 | 0.100 | 0.120 | 0.161 | 1 | 1 | Mitochondria | ||
| ABCB11 | SPGP or BSEP | MDR | 2q24.3 | 108385 | 0.173 | 0.212 | 1 | 1 | Liver, intestine | Bile salt transport | Progressive familial intrahepatic cholestasis-2 | |
| ABCC1 | MRP1 | CF/MRP | 16p13.12 | 192840 | 0.064 | 0.060 | 0.062 | 7 | 7 | Lung, testes, intestine, PBMC, kidney, brain | Drug resistance | |
| ABCC2 | MRP2 or cMOAT | CF/MRP | 10q24.2 | 69011 | 0.313 | 0.243 | 0.180 | 1 | 1 | Liver, intestine, kidney | Organic anion efflux | Dubin-johnson syndrome |
| ABCC3 | MRP3 | CF/MRP | 17q21.33 | 56836 | 0.169 | 0.164 | 0.204 | 1 | 1 | Lung, intestine, liver, kidney, placenta, pancreas, colon | Drug resistance | |
| ABCC4 | MRP4 | CF/MRP | 13q32.1 | 281594 | 0.137 | 0.099 | 0.060 | 1 | 1 | Prostate, lung, adrenal gland, ovary, testis | Nucleoside transport | |
| ABCC5 | MRP5 | CF/MRP | 3q27.1 | 97956 | 0.069 | 0.073 | 0.053 | 1 | 1 | Ubiquitous | Nucleoside transport | |
| ABCC6 | MRP6 | CF/MRP | 16p13.12 | 73325 | 0.098 | 0.149 | 0.160 | 1 | 1 | Kidney, liver | Nucleoside transport | Pseudoxanthoma elasticum |
| CFTR | ABCC7 | CF/MRP | 7q31.31 | 188699 | 0.172 | 0.172 | 0.238 | 1 | 1 | Exocrine tissue | Chloride ion channel | Cystic fibrosis CBAVD, pancreatitis, bronchiectasis |
| ABCC8 | SUR1 | CF/MRP | 11p15.1 | 84017 | 0.093 | 0.045 | 0.055 | 1 | 1 | Pancreas | Sulfonylurea receptor | Familial persistent hyperinsulinemic hypoglycemia of infancy; AD type 2 diabetes |
| ABCC9 | SUR2 | CF/MRP | 12p12.1 | 135631 | 0.021 | 0.039 | 3 | 3 | Heart, muscle | Regulatory subunit of cardiac K(ATP) channel | Dilated cardiomyopathy with ventricular tachycardia | |
| ABCC10 | MRP7 | CF/MRP | 6p21.1 | 18675 | 0.170 | 0.195 | 0.171 | 1 | 1 | Low in all tissues; a little higher in heart, skeletal, muscle, spleen, liver | ||
| ABCC11 | MRP8 | CF/MRP | 16q12.1 | 68267 | 0.304 | NA | NA | 2 | 3 | Low in all tissues, a little higher in breast, and testis | ||
| ABCC12 | MRP9 | CF/MRP | 16q12.1 | 63798 | 0.200 | 0.184 | 0.166 | 1 | 1 | Low in all tissues, a little higher in breast, testis, brain, ovary, skeletal muscle | ||
| ABCC13 | ||||||||||||
| ABCD1 | ALDP | ALD | Xq28 | 19846 | 0.045 | 0.053 | 0.058 | 1 | 1 | Peroxisomes | VLCFA transport regulation | Adrenoleukodystrophy |
| ABCD2 | ALDL1, ALDR | ALD | 12q11 | 67424 | 0.085 | 0.066 | 0.073 | 1 | 1 | Peroxisomes | ||
| ABCD3 | PXMP1, PMP70 | ALD | 1p22.1 | 100072 | 0.050 | 0.051 | 0.058 | 1 | 1 | Peroxisomes | ||
| ABCD4 | PMP69, P70R | ALD | 14q24.3 | 17540 | 0.188 | 0.112 | 0.110 | 1 | 1 | Peroxisomes | ||
| ABCE1 | OABP, RNS41 | OABP | 4q31.31 | 30851 | 0.003 | 0.061 | 0.002 | 1 | 1 | Ovary, testes, spleen | Oligoadenylate binding protein | |
| ABCF1 | ABC50 | GCN20 | 6p21.1 | 19920 | 0.022 | 0.057 | 0.055 | 1 | 1 | Ubiquitous | ||
| ABCF2 | GCN20 | 7q36.1 | 19395 | 0.036 | 0.011 | 0.010 | 2 | 2 | Ubiquitous | |||
| ABCF3 | GCN20 | 3q27.1 | 7908 | 0.027 | 0.030 | 0.035 | 1 | 1 | Ubiquitous | |||
| ABCG1 | ABC8, White | White | 21q22.3 | 97556 | 0.012 | 0.012 | 0.010 | 7 | 7 | Ubiquitous | Cholesterol transport? | |
| ABCG2 | ABCP, MXR, BCRP | White | 4q22 | 66883 | 0.269 | 0.206 | 0.202 | 1 | 1 | Placenta, intestine, brain | Toxin efflux, drug resistance | |
| ABCG4 | White2 | White | 11q23 | 13626 | 0.026 | 0.064 | 0.027 | 1 | 1 | Liver | ||
| ABCG5 | White3 | White | 2p21 | 26348 | 0.302 | 0.175 | 0.157 | 1 | 1 | Liver, intestine | Sterol transport | Sitosterolemia |
| ABCG8 | White | 2p21 | 39503 | 0.152 | 0.126 | 0.206 | 1 | 1 | Liver, intestine | Sterol transport | Sitosterolemia | |
ABC, ATP-binding cassette; HDL, high-density lipoprotein; PGY, P-glucoprotein; MDR, multidrug resistance; MXR, multixenobiotic resistance; MRP, multidrug resistance-associated protein; BCRP, breast cancer resistance protein; TAP, transporter ATP-binding cassette, ATP-binding cassette sub-family B; PC, phosphatidylcholine; MTABC, mitochondrial ATP-binding cassette; SPGP, sister of P-glycoprotein; BSEP, bile salt export pump; MRP, multiple drug resistance protein; PBMC, peripheral blood mononuclear cell; cMOAT, canalicular multispecific organic anion transporter 1; CF, cystic fibrosis; SUR, sulfonylurea receptor; ALD, adrenoleukodystrophy; ALDP, adrenoleukodystrophy protein; ALDR, adrenoleukodystrophy-related protein; PXMP, peroxisomal membrane protein; PMP, putative peroxisomal membrane protein; OABP, ATP-binding cassette, sub-family E; GCN20, ATP-binding cassette sub-family F; CBAVD, congenital bilateral absence of the vas deferens; VLCFA, very long chain fatty acid.
Application of Nanoparticle Delivery Systems to Reverse Drug Efflux-mediated Resistance
As mentioned above, overexpression of ABC transporters is the broadly known tumor survival mechanism that limits the efficacy of chemotherapeutic agents in clinical cancer treatments. Currently, there are no traditional strategies without serious side effects to completely reverse chemotherapeutic resistance in tumors. Based on their unique physical and biological properties, cancer nanotechnologies developed in recent years offer an unprecedented opportunity for rational delivery of anticancer drugs to solid tumors[20],[21]. The promise of nanotechnology lies in the ability to engineer customizable nanoscale constructs, which have more controllable surface for different modification, and to accommodate multiple types of payloads, such as cancer chemotherapeutics, chemosensitizers, or molecular imaging agents[22],[23]. Nanoparticles have been developed to prevent, detect, and treat resistant cancer cells while minimizing serious toxicity in normal cells and improving drug solubility and stability. Nanostructure platforms derive their effectiveness from adequate delivery systems, including polymers, dendrimers, nanoshells, nanotubes, micelles, liposomes, lipid-based nanoparticles, magnetic nanoparticles, and virus nanoparticles. For rigid nanoparticles, the size to support long circulation in vivo may not exceed 200 nm, a size that is not achievable with individual molecules alone or with equivalent materials at a larger scale. Nanoscale drug delivery systems in the size range of 10-100 nm penetrate preferentially through the tumor vasculature via the so-called enhanced permeability and retention (EPR) effect [24]–[26]. Multifunctionalization of hydrophilic nanoparticles can provide a long circulating half-life and prolong the exposure time of chemotherapeutic drugs. These unique properties based on nanostructure effectively increase the intracellular accumulation of anticancer drugs by controllable and efficient release at targeted regions due to their size effect (Figure 1). Furthermore, properly designed nanoparticles with a targeting element can aim at specific targeting sites actively with the therapeutic payloads to overcome tumor MDR. One example is the use of folate acid as an active target. Nanoparticle with folate acid ligands can bind to folate receptors, which were found to be overexpressed on the surface of drug-resistant tumor cells, to achieve the specific accumulation in tumors. In addition, nanoplatforms offer opportunities to coencapsulate multiple therapeutic agents into a single functional carrier and allow imaging to be combined with drug treatment to monitor therapeutic effects in real time. Efforts to co-administrate drugs with ultrasound and thermosensitive therapy or photodynamic therapy improve the nanoparticle delivery systems for synergistic and comprehensive functions of cancer nanochemotherapy[27]–[30]. In summary, the advantages of nanoparticle-based drug delivery system include narrow size distribution, low carrier toxicity, enhanced drug solubilization via protecting drugs from efflux, prevention from drug metabolism or excretion before accumulating in tumors, increased drug loading, prolonged drug circulation, specific site targeting, and controlled drug release. Cancer nanotechnology has the potential to reverse tumor MDR in a way that is not achievable with traditional strategies.
Figure 1. Unique nanoplatforms effectively increase the intracellular accumulation of anticancer drugs by controllable and efficient release at targeted regions.
These functionalized nanoparticles can be modified with anticancer drugs, image moiety, specific ligands, antibodies, and so on, to fulfill chemotherapy through increasing drug accumulation.
Cancer Nanomedicine for Overcoming Drug Efflux-mediated Resistance
An important prerequisite for reversing drug resistance in cancer is achieving a high concentration of the drug in the plasma, a high concentration and long retention time in MDR cancer cells to ensure effective intracellular accumulation. Considering different mechanisms of drug resistance in cancer, nanoparticles are always designed to inhibit or bypass efflux pumps on the membrane or to enhance endocytosis when recognizing MDR tumors. Over 50% of the anticancer drugs used in the clinic today are targeted by P-gp[31]. However, P-gp inhibitors do not have specific selectivity and also block the normal function of P-gp. Nanotechnology refines the concept of co-administering anticancer agents and P-gp inhibitors by combining them into a single drug carrier for simultaneous delivery into MDR tumor cells. In a recent review by Gottesman et al.[32], the strategies for circumventing P-gp-mediated MDR are eloquently categorized as follows: using P-gp inhibitors to block the efflux of cytotoxic agents; using drugs that are not substrates of P-gp; and exploiting the properties of MDR cells, such as receptor overexpression and collateral drug sensitivity. D-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS 1000), which functions as an effective inhibitor of P-gp, turned out to be one of the prominent surfactants that enhancing the cytotoxicity of doxorubicin, vinblastine, paclitaxel, and colchicines in G185 cells comparable to that in the parental cells. Reversal of P-gp activity was due to the effect of TPGS 1000 on transport at concentrations even below its critical micelle point of 0.02 wt%. This excipient was evaluated in a phase II clinical trial for drug resistance[33]. Pluronic block copolymer (P85) is another important and promising example of a modifying agent for P-gp. Membrane fluidization by P85 treatment led to inhibition of the P-gp ATPase drug efflux system and to interference with metabolic processes. These results indicate that both energy depletion (via decreasing ATP pool necessary for P-gp function) and increased permeability and fluidization of a broad spectrum of drugs are critical factors contributing to the activity of the block copolymer for MDR reversion[34],[35]. First-generation agents (for example, the calcium channel blocker verapamil) are limited by unacceptable toxicity to normal tissues, whereas second-generation agents (for example, valspodar and biricodar) have better tolerability but are limited by nonspecificity. Third-generation inhibitors (for example, tariquidar XR9576, LY335979, GF120918, 9576, etc.) have high potency and specificity for P-gp. Tariquidar has shown marked effectiveness in early clinical trials. GF120918 has achieved adequate P-gp inhibition in vivo without significant side effects[36],[37]. Taken together, the development of these findings suggests the necessity to consider to combine nanoparticle therapies with P-gp inhibitors.
Recent studies also demonstrated that nucleic acid (DNA, miRNA, siRNA, etc.)-based nanoparticles play a critical role in the modulation of drug resistance in tumors by effectively decreasing MDR1 expression in vivo[38]–[40]. The employed system, which uses siRNA to silence the expression of ABC transporters in combination with an appropriate anticancer drug, is a systemic administration strategy for MDR cells. Meng et al.[41] successfully achieved dual delivery of doxorubicin (Dox) and P-gp siRNA loaded in mesoporous silica nanoparticles (MSNP). P-gp gene knockdown by siRNA effectively increased the intracellular and intranuclear drug concentration. Similar results were also observed in vitro and in vivo using RGD peptide (arginine-glycine-aspartic acid)-modified liposomes containing P-gp siRNA or doxorubicin[42]. MacDiarmid et al. [43] provided a dual sequential treatment strategy for drug-resistant tumors with targeted micelles containing siRNA and a cytotoxic drug. First, the resistant tumors were treated with siRNA/shRNA-containing minicells targeted to tumors via bispecific antibodies (BsAb) for 48 h (for siRNA-containing minicells) and 144 h (for shRNA-containing minicells) to achieve substantial knockdown of MDR1. A second wave of treatment with siRNA/shRNA-containing minicells was followed with intravenous administration of BsAb-targeted minicells packaged with cytotoxic drugs to resistant tumor xenografts. Both the sequential and simultaneous approaches were effective and feasible and markedly decreased the doses of cytotoxic drug necessary for eliminating tumors. Injection of anti-MDR1 short hairpin RNA-encoding vectors into tumor cells with intravenous administration of doxorubicin completely reversed the MDR phenotype and inhibited tumor growth[44].
In addition, some nanomedicines that circumvent MDR in cancer through other targets are being tested in clinical trials (Table 2). These include taxane analogs DJ-927 (phase I/Il)[57]–[59] and ortataxel (phase II)[60],[61], as well as BMS-184476 (phase II)[62]–[64] and RPR 109881A (phase I)[65]–[67], which were purported to have a broad spectrum of activity both in sensitive and resistant tumor cell lines. Nab-paclitaxel is a novel clinical entity incorporating paclitaxel into an albumin nanoparticle, leading to increased intratumoral concentration and showing with superior response rate, longer time to tumor progression, and prolonged survival as second-line therapy in patients with gynecologic cancers[68],[69]. EGFR-targeted polymer-blend nanocarriers with a combination of paclitaxel and lonidamine were found to enhance the therapeutic index of both drugs by inhibiting the Warburg effect and promoting mitochondrial binding of pro-apoptotic Bcl-2 protein (via lonidamine), while hyperstabilizing microtubules (via paclitaxel)[70].
Table 2. Selected examples of nanoparticles used to overcome drug resistance in cancer treatment evaluated in vivo in the past three years.
| Nanotransporters | Main bioactive element | Main Mechanism | Reference |
| siRNA, minicells | Doxorubicin | P-gp inhibition | [43] |
| Dendrimer phthalocyanine-encapsulated polymeric micelle (DPc/m)-mediated PCI | Doxorubicin | P-gp inhibition | [45] |
| Poly(D,L-lactide-co-glycolide) | Paclitaxel, tariquidar | P-gp inhibition | [46] |
| Poly(D,L-lactide-co-glyco-lide) nanoparticles | Paclitaxel, P-gp targeted siRNA | P-gp inhibition | [47] |
| Albumin bound nanoparticles | Paclitaxel | Paditaxel-induced NF-κB pathway that up-regulatesVEGF-A | [48] |
| Aerosol-OT (AOT) | Doxorubicin | Prevents the accumulation of anticancer drugs | [49],[50] |
| Folate receptor-targeting nanoparticle | Heparin-folate-paclitaxel (HFT) backbone with an additional paclitaxel | P-gp inhibition | [51] |
| PLGA nanoparticles | Vincristine sulfate, verapamil hydrochloride | Enhanced permeation and retention effect | [52] |
| Cationic liposome-polycation-DNA (LPD) and anionic liposome-polycation-DNA (LPD-II) | Doxorubicin and siRNA | Avoid P-gp efflux and increase Dox uptake | [53] |
| Polymer-blend nanoparticle | Ceramide | Modulation of the apoptotic threshold | [54] |
| Albumin bound nanoparticles | Rapamycin and perifosine | Suppression of the PI3K/Ak1/mTOR pathway | [55] |
| Fe(3)O(4)-magnetic nanoparticle | Daunorubicin | Mdr-1 inhibition | [56] |
P-gp, P-glycoprotein; aerosol-OT (AOT), bis-2-ethylhexoxyl sodium sulfosuccinate; PLGA, poly(lactic-co-glycolic acid); VEGF-A, vascular endothelial growth factor A; Dox, doxorubicin; PI3K, phosphoinositide 3-kinase; Akt, protein kinase B; mTOR, mammalian target of rapamycin; Mdr-1, multidrug resistant gene-1.
Future Perspectives
MDR is a major impediment to the success of cancer chemotherapy. P-gp is the best known membrane transporter involved in MDR in tumors. Several strategies have been used to address MDR, especially P-gp-mediated drug resistance in tumors. However, clinical success has been limited, largely due to lack of efficacy and/or significant toxicity. To overcome both the dose-limiting side effects of conventional chemotherapeutic agents and the therapeutic failure resulting from MDR, cancer nanotechnology has been developed and shown its ability to target tumors based on their unique physical and biological properties. To date, nanoparticles have been investigated primarily to address P-gp and have been shown to improve anticancer efficacy, indicating that nanomedical strategies might provide a new opportunity to overcome MDR[71]. The most predominant advantage of nanomedicine is to deliver and concentrate drugs at the plasma membrane where ABC transporters are located and saturated with extra drugs. In addition, functionalized nanoparticles themselves or their metabolites can also block the function of ABC transporters such as P-gp by direct or indirect interaction and inhibition. The flexibility of nanoparticles with regard to their size and shape increases their potential to enhance drug-loading capacity, stabilize drugs and regulate their release rates, and deliver drugs to targeted sites effectively and specifically. This article provides a glimpse into the nanotechnology-based strategies being developed to overcome drug resistance. To the best of our knowledge, however, there are currently no nano-fomulations for drug delivery aimed at overcoming drug resistance that have been effective in clinical tests. Various nanocarriers for targeted delivery of anticancer drugs have already undergone in vivo testing in mouse models (Table 2) and clinical evaluation in humans. With better understanding of the physiologic properties of drug-resistant tumors and enhanced nanomaterial design, there will be more opportunities to develop multifunctional nanostructures for circumventing drug resistance. A safe and effective multifunctional nanosystem could provide a versatile platform to benefit patients with MDR tumors in the future.
Acknowledgments
This work is financially supported by the National Key Basic Research Program of China (973 projects 2009CB930200) and the program of National Natural Science Foundation of China (30970784 and 81171455). The authors are grateful for the support of the Chinese Academy of Sciences (CAS) “Hundred Talents Program” and CAS Knowledge Innovation Program.
References
- 1.The World Cancer Report—the major findings. Cent Eur J Public Health. 2003;11:177–179. [PubMed] [Google Scholar]
- 2.Chen CJ, Chin JE, Ueda K, et al. Internal duplication and homology with bacterial transport proteins in the mdr1 (P-glycoprotein) gene from multidrug-resistant human cells. Cell. 1986;47:381–389. doi: 10.1016/0092-8674(86)90595-7. [DOI] [PubMed] [Google Scholar]
- 3.Myers AM, Crivellone MD, Tzagoloff A. Assembly of the mitochondrial membrane system. MRP1 and MRP2, two yeast nuclear genes coding for mitochondrial ribosomal proteins. J Biol Chem. 1987;262:3388–3397. [PubMed] [Google Scholar]
- 4.Maliepaard M, van Gastelen MA, de Jong LA, et al. Overexpression of the BCRP/MXR/ABCP gene in a topotecan-selected ovarian tumor cell line. Cancer Res. 1999;59:4559–4563. [PubMed] [Google Scholar]
- 5.Kirkin V, Joos S, Zornig M. The role of Bcl-2 family members in tumorigenesis. Biochim Biophys Acta. 2004;1644:229–249. doi: 10.1016/j.bbamcr.2003.08.009. [DOI] [PubMed] [Google Scholar]
- 6.Viktorsson K, De Petris L, Lewensohn R. The role of p53 in treatment responses of lung cancer. Biochem Biophys Res Commun. 2005;331:868–880. doi: 10.1016/j.bbrc.2005.03.192. [DOI] [PubMed] [Google Scholar]
- 7.Chin KV, Ueda K, Pastan I, et al. Modulation of activity of the promoter of the human MDR1 gene by Ras and p53. Science. 1992;255:459–462. doi: 10.1126/science.1346476. [DOI] [PubMed] [Google Scholar]
- 8.Leonard GD, Fojo T, Bates SE. The role of ABC transporters in clinical practice. Oncologist. 2003;8:411–424. doi: 10.1634/theoncologist.8-5-411. [DOI] [PubMed] [Google Scholar]
- 9.Szakacs G, Varadi A, Ozvegy-Laczka C, et al. The role of ABC transporters in drug absorption, distribution, metabolism, excretion and toxicity (ADME-TOX) Drug Discov Today. 2008;13:379–393. doi: 10.1016/j.drudis.2007.12.010. [DOI] [PubMed] [Google Scholar]
- 10.Glavinas H, Krajcsi P, Cserepes J, et al. The role of ABC transporters in drug resistance, metabolism and toxicity. Curr Drug Deliv. 2004;1:27–42. doi: 10.2174/1567201043480036. [DOI] [PubMed] [Google Scholar]
- 11.Zhang JT. Use of arrays to investigate the contribution of ATP-binding cassette transporters to drug resistance in cancer chemotherapy and prediction of chemosensitivity. Cell Res. 2007;17:311–323. doi: 10.1038/cr.2007.15. [DOI] [PubMed] [Google Scholar]
- 12.Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002;2:48–58. doi: 10.1038/nrc706. [DOI] [PubMed] [Google Scholar]
- 13.Wang Q, Wang JR, Wang HL, et al. Down - regulation of P-glycoprotein is associated with resistance to cisplatin and VP-16 in human lung cancer cell lines. Anticancer Res. 2010;30:3593–3598. [PubMed] [Google Scholar]
- 14.Shiraki N, Hamada A, Ohmura T, et al. Increase in doxorubicin cytotoxicity by inhibition of P-glycoprotein activity with lomerizine. Biol Pharm Bull. 2001;24:555–557. doi: 10.1248/bpb.24.555. [DOI] [PubMed] [Google Scholar]
- 15.van Asperen J, van Tellingen O, Tijssen F, et al. Increased accumulation of doxorubicin and doxorubicinol in cardiac tissue of mice lacking mdr1a P-glycoprotein. Br J Cancer. 1999;79:108–113. doi: 10.1038/sj.bjc.6690019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Pires MM, Emmert D, Hrycyna CA, et al. Inhibition of P-glycoprotein-mediated paclitaxel resistance by reversibly linked quinine homodimers. Mol Pharmacol. 2009;75:92–100. doi: 10.1124/mol.108.050492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Enokida H, Gotanda T, Oku S, et al. Reversal of P-glycoprotein-mediated paclitaxel resistance by new synthetic isoprenoids in human bladder cancer cell line. Jpn J Cancer Res. 2002;93:1037–1046. doi: 10.1111/j.1349-7006.2002.tb02481.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ling V, Thompson LH. Reduced permeability in CHO cells as a mechanism of resistance to colchicine. J Cell Physiol. 1974;83:103–116. doi: 10.1002/jcp.1040830114. [DOI] [PubMed] [Google Scholar]
- 19.See YP, Carlsen SA, Till JE, et al. Increased drug permeability in Chinese hamster ovary cells in the presence of cyanide. Biochim Biophys Acta. 1974;373:242–252. doi: 10.1016/0005-2736(74)90148-5. [DOI] [PubMed] [Google Scholar]
- 20.Hede S, Huilgol N. “Nano”: the new nemesis of cancer. J Cancer Res Ther. 2006;2:186–195. doi: 10.4103/0973-1482.29829. [DOI] [PubMed] [Google Scholar]
- 21.Moghimi SM, Hunter AC, Murray JC. Nanomedicine: current status and future prospects. FASEB J. 2005;19:311–330. doi: 10.1096/fj.04-2747rev. [DOI] [PubMed] [Google Scholar]
- 22.Basu S, Chaudhuri P, Sengupta S. Targeting oncogenic signaling pathways by exploiting nanotechnology. Cell Cycle. 2009;8:3480–3487. doi: 10.4161/cc.8.21.9851. [DOI] [PubMed] [Google Scholar]
- 23.Nie S, Xing Y, Kim GJ, et al. Nanotechnology applications in cancer. Annu Rev Biomed Eng. 2007;9:257–288. doi: 10.1146/annurev.bioeng.9.060906.152025. [DOI] [PubMed] [Google Scholar]
- 24.Duncan R. The dawning era of polymer therapeutics. Nat Rev Drug Discov. 2003;2:347–360. doi: 10.1038/nrd1088. [DOI] [PubMed] [Google Scholar]
- 25.Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res. 1986;46:6387–6392. [PubMed] [Google Scholar]
- 26.Jain RK. Transport of molecules, particles, and cells in solid tumors. Annu Rev Biomed Eng. 1999;1:241–263. doi: 10.1146/annurev.bioeng.1.1.241. [DOI] [PubMed] [Google Scholar]
- 27.Leroux JC. Injectable nanocarriers for biodetoxification. Nat Nanotechnol. 2007;2:679–684. doi: 10.1038/nnano.2007.339. [DOI] [PubMed] [Google Scholar]
- 28.Haley B, Frenkel E. Nanoparticles for drug delivery in cancer treatment. Urol Oncol. 2008;26:57–64. doi: 10.1016/j.urolonc.2007.03.015. [DOI] [PubMed] [Google Scholar]
- 29.Thierry B. Drug nanocarriers and functional nanoparticles: applications in cancer therapy. Curr Drug Deliv. 2009;6:391–403. doi: 10.2174/156720109789000474. [DOI] [PubMed] [Google Scholar]
- 30.Alexis F, Pridgen EM, Langer R, et al. Nanoparticle technologies for cancer therapy. Handb Exp Pharmacol. 2010;197:55–86. doi: 10.1007/978-3-642-00477-3_2. [DOI] [PubMed] [Google Scholar]
- 31.Constantinides PP, Wasan KM. Lipid formulation strategies for enhancing intestinal transport and absorption of P-glycoprotein (P-gp) substrate drugs: in vitro/in vivo case studies. J Pharm Sci. 2007;96:235–248. doi: 10.1002/jps.20780. [DOI] [PubMed] [Google Scholar]
- 32.Szakacs G, Paterson JK, Ludwig JA, et al. Targeting multidrug resistance in cancer. Nat Rev Drug Discov. 2006;5:219–234. doi: 10.1038/nrd1984. [DOI] [PubMed] [Google Scholar]
- 33.Dintaman JM, Silverman JA. Inhibition of P-glycoprotein by D-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS) Pharm Res. 1999;16:1550–1556. doi: 10.1023/a:1015000503629. [DOI] [PubMed] [Google Scholar]
- 34.Batrakova EV, Li S, Vinogradov SV, et al. Mechanism of pluronic effect on P-glycoprotein efflux system in blood-brain barrier: contributions of energy depletion and membrane fluidization. J Pharmacol Exp Ther. 2001;299:483–493. [PubMed] [Google Scholar]
- 35.Batrakova EV, Li S, Li Y, et al. Effect of pluronic P85 on ATPase activity of drug efflux transporters. Pharm Res. 2004;21:2226–2233. doi: 10.1007/s11095-004-7675-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hyafil F, Vergely C, Du Vignaud P, et al. In vitro and in vivo reversal of multidrug resistance by GF120918, an acridonecarboxamide derivative. Cancer Res. 1993;53:4595–4602. [PubMed] [Google Scholar]
- 37.Witherspoon SM, Emerson DL, Kerr BM, et al. Flow cytometric assay of modulation of P-glycoprotein function in whole blood by the multidrug resistance inhibitor GG918. Clin Cancer Res. 1996;2:7–12. [PubMed] [Google Scholar]
- 38.Wang Z, Li Y, Ahmad A, et al. Targeting mirnas involved in cancer stem cell and EMT regulation: an emerging concept in overcoming drug resistance. Drug Resist Updat. 2010;13:109–118. doi: 10.1016/j.drup.2010.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Patil Y, Panyam J. Polymeric nanoparticles for siRNA delivery and gene silencing. Int J Pharm. 2009;367:195–203. doi: 10.1016/j.ijpharm.2008.09.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Mangala LS, Zuzel V, Schmandt R, et al. Therapeutic targeting of ATP7B in ovarian carcinoma. Clin Cancer Res. 2009;15:3770–3780. doi: 10.1158/1078-0432.CCR-08-2306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Meng H, Liong M, Xia T, et al. Engineered design of mesoporous silica nanoparticles to deliver doxorubicin and P-glycoprotein siRNA to overcome drug resistance in a cancer cell line. ACS Nano. 2010;4:4539–4550. doi: 10.1021/nn100690m. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Jiang J, Yang SJ, Wang JC, et al. Sequential treatment of drug-resistant tumors with RGD-modified liposomes containing siRNA or doxorubicin. Eur J Pharm Biopharm. 2010;76:170–178. doi: 10.1016/j.ejpb.2010.06.011. [DOI] [PubMed] [Google Scholar]
- 43.MacDiarmid JA, Amaro-Mugridge NB, Madrid-Weiss J, et al. Sequential treatment of drug-resistant tumors with targeted minicells containing siRNA or a cytotoxic drug. Nat Biotechnol. 2009;27:643–651. doi: 10.1038/nbt.1547. [DOI] [PubMed] [Google Scholar]
- 44.Walther W, Stein U, Lage H. Jet-injection of short hairpin RNA-encoding vectors into tumor cells. Methods Mol Biol. 2010;629:123–139. doi: 10.1007/978-1-60761-657-3_9. [DOI] [PubMed] [Google Scholar]
- 45.Lu HL, Syu WJ, Nishiyama N, et al. Dendrimer phthalocyanine-encapsulated polymeric micelle-mediated photochemical internalization extends the efficacy of photodynamic therapy and overcomes drug-resistance in vivo. J Control Release. 2011:12. doi: 10.1016/j.jconrel.2011.06.005. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- 46.Patil Y, Sadhukha T, Ma L, et al. Nanoparticle-mediated simultaneous and targeted delivery of paclitaxel and tariquidar overcomes tumor drug resistance. J Control Release. 2009;136:21–29. doi: 10.1016/j.jconrel.2009.01.021. [DOI] [PubMed] [Google Scholar]
- 47.Patil YB, Swaminathan SK, Sadhukha T, et al. The use of nanoparticle-mediated targeted gene silencing and drug delivery to overcome tumor drug resistance. Biomaterials. 2010;31:358–365. doi: 10.1016/j.biomaterials.2009.09.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Volk LD, Flister MJ, Chihade D, et al. Synergy of nab-paclitaxel and bevacizumab in eradicating large orthotopic breast tumors and preexisting metastases. Neoplasia. 2011;13:327–338. doi: 10.1593/neo.101490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Khdair A, Handa H, Mao G, et al. Nanoparticle-mediated combination chemotherapy and photodynamic therapy overcomes tumor drug resistance in vitro. Eur J Pharm Biopharm. 2009;71:214–222. doi: 10.1016/j.ejpb.2008.08.017. [DOI] [PubMed] [Google Scholar]
- 50.Khdair A, Chen D, Patil Y, et al. Nanoparticle-mediated combination chemotherapy and photodynamic therapy overcomes tumor drug resistance. J Control Release. 141:137–144. doi: 10.1016/j.jconrel.2009.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Chen K, Iribarren P, Hu J, et al. Activation of toll-like receptor 2 on microglia promotes cell uptake of alzheimer disease-associated amyloid beta peptide. J Biol Chem. 2006;281:3651–3659. doi: 10.1074/jbc.M508125200. [DOI] [PubMed] [Google Scholar]
- 52.Song XR, Zheng Y, He G, et al. Development of PLGA nanoparticles simultaneously loaded with vincristine and verapamil for treatment of hepatocellular carcinoma. J Pharm Sci. 2010;99:4874–4879. doi: 10.1002/jps.22200. [DOI] [PubMed] [Google Scholar]
- 53.Chen Y, Bathula SR, Li J, et al. Multifunctional nanoparticles delivering small interfering RNA and doxorubicin overcome drug resistance in cancer. J Biol Chem. 2010;285:22639–22650. doi: 10.1074/jbc.M110.125906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.van Vlerken LE, Duan Z, Little SR, et al. Augmentation of therapeutic efficacy in drug-resistant tumor models using ceramide coadministration in temporal-controlled polymer-blend nanoparticle delivery systems. AAPS J. 2010;12:171–180. doi: 10.1208/s12248-010-9174-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Cirstea D, Hideshima T, Rodig S, et al. Dual inhibition of akt/mammalian target of rapamycin pathway by nanoparticle albumin-bound-rapamycin and perifosine induces antitumor activity in multiple myeloma. Mol Cancer Ther. 2010;9:963–975. doi: 10.1158/1535-7163.MCT-09-0763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Lai BB, Chen BA, Cheng J, et al. Daunorubicin-loaded magnetic nanoparticles of Fe(3)O(4) greatly enhance the responses of multidrug-resistant K562 leukemic cells in a nude mouse xenograft model to chemotherapy. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2009;17:345–351. [PubMed] [Google Scholar]
- 57.Ono C, Takao A, Atsumi R. Absorption, distribution, and excretion of DJ-927, a novel orally effective taxane, in mice, dogs, and monkeys. Biol Pharm Bull. 2004;27:345–351. doi: 10.1248/bpb.27.345. [DOI] [PubMed] [Google Scholar]
- 58.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;94:459–466. doi: 10.1111/j.1349-7006.2003.tb01465.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Baas P, Szczesna A, Albert I, et al. Phase I/Il study of a 3 weekly oral taxane (DJ-927) in patients with recurrent, advanced non-small cell lung cancer. J Thorac Oncol. 2008;3:745–750. doi: 10.1097/JTO.0b013e31817c73ff. [DOI] [PubMed] [Google Scholar]
- 60.Minderman H, Brooks TA, O'Loughlin KL, et al. Broad-spectrum modulation of ATP-binding cassette transport proteins by the taxane derivatives ortataxel (IDN-5109, BAY 59-8862) and tRA96023. Cancer Chemother Pharmacol. 2004;53:363–369. doi: 10.1007/s00280-003-0745-2. [DOI] [PubMed] [Google Scholar]
- 61.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;8:2647–2654. [PubMed] [Google Scholar]
- 62.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;44:4577–4583. doi: 10.1021/jm0102607. [DOI] [PubMed] [Google Scholar]
- 63.Camps C, Felip E, Sanchez JM, et al. Phase II trial of the novel taxane BMS-184476 as second-line in non-small-cell lung cancer. Ann Oncol. 2005;16:597–601. doi: 10.1093/annonc/mdi120. [DOI] [PubMed] [Google Scholar]
- 64.Bilenker JH, Stevenson JP, Gallagher ML, et al. Phase I trial of the novel taxane BMS-184476 administered in combination with carboplatin every 21 days. Br J Cancer. 2004;91:213–218. doi: 10.1038/sj.bjc.6601885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Kurata T, Shimada Y, Tamura T, et al. Phase I and pharmacokinetic study of a new taxoid, RPR5 109881A, given as a 1-hour intravenous infusion in patients with advanced solid tumors. J Clin Oncol. 2000;18:3164–3171. doi: 10.1200/JCO.2000.18.17.3164. [DOI] [PubMed] [Google Scholar]
- 66.Gelmon KA, Latreille J, Tolcher A, et al. Phase I dose-finding study of a new taxane, RPR 109881A, administered as a one-hour intravenous infusion days 1 and 8 to patients with advanced solid tumors. J Clin Oncol. 2000;18:4098–4108. doi: 10.1200/JCO.2000.18.24.4098. [DOI] [PubMed] [Google Scholar]
- 67.Sessa C, Cuvier C, Caldiera S, et al. Phase I clinical and pharmacokinetic studies of the taxoid derivative RPR 109881A administered as a 1-hour or a 3-hour infusion in patients with advanced solid tumors. Ann Oncol. 2002;13:1140–1150. doi: 10.1093/annonc/mdf174. [DOI] [PubMed] [Google Scholar]
- 68.Miele E, Spinelli GP, Tomao F, et al. Albumin-bound formulation of paclitaxel (Abraxane ABI-007) in the treatment of breast cancer. Int J Nanomedicine. 2009;4:99–105. doi: 10.2147/ijn.s3061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Moreno-Aspitia A, Perez EA. Nanoparticle albumin-bound paclitaxel (ABI-007): a newer taxane alternative in breast cancer. Future Oncol. 2005;1:755–762. doi: 10.2217/14796694.1.6.755. [DOI] [PubMed] [Google Scholar]
- 70.Milane L, Duan Z, Amiji M. Development of EGFR-targeted polymer blend nanocarriers for combination paclitaxel/lonidamine delivery to treat multi-drug resistance in human breast and ovarian tumor cells. Mol Pharm. 2011;8:185–203. doi: 10.1021/mp1002653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Wang YZ, Huang B, Lv J, et al. Current status of nanotechnology applied in biomedicine. Acta Biophys Sin. 2009;25:168–174. [Google Scholar]

