REVIEW ARTICLE


Nanomedicine-Mediated Combination Drug Therapy in Tumor



Dazhong Chen1, Fangyuan Xie1, 4, Duxin Sun3, Chuan Yin2, Jie Gao1, 3, *, Yanqiang Zhong1, *
1 Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
2 Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
3 Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
4 Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai 200438, China


Article Metrics

CrossRef Citations:
2
Total Statistics:

Full-Text HTML Views: 835
Abstract HTML Views: 425
PDF Downloads: 6
ePub Downloads: 3
Total Views/Downloads: 1269
Unique Statistics:

Full-Text HTML Views: 429
Abstract HTML Views: 274
PDF Downloads: 6
ePub Downloads: 3
Total Views/Downloads: 712



© Chen et al. ; Licensee Bentham Open

open-access license: This is an open access article licensed under the terms of the Creative Commons Attribution-Non-Commercial 4.0 International Public License (CC BY-NC 4.0) (https://creativecommons.org/licenses/by-nc/4.0/legalcode), which permits unrestricted, non-commercial use, distribution and reproduction in any medium, provided the work is properly cited.

* Address correspondence to this author at the Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, Shanghai, China; Tel: 86-21-81871285; Fax: 86-21-81871285; E-mails: yqzhong68@163.com, gaojie@smmu.edu.cn, gaojiehighclea@163.com


Abstract

Background:

Combined chemotherapy has gradually become one of the conventional methods of cancer treatment due to the limitation of monotherapy. However, combined chemotherapy has several drawbacks that may lead to treatment failure because drug synergy cannot be guaranteed, achievement of the optimal synergistic drug ratio is difficult, and drug uptake into the tumor is inconsistent. Nanomedicine can be a safe and effective form of drug delivery, which may address the problems associated with combination chemotherapy.

Objective:

This review summarizes the recent research in this area, including the use of nanoparticles, liposomes, lipid-polymer hybrid nanoparticles, and polymeric micelles, and provides new approach for combined chemotherapy.

Methods:

By collecting and referring to the related literature in recent years.

Results:

Compared with conventional drugs, nanomedicine has the following advantages: it increases bioavailability of poorly soluble drugs, prolongs drug circulation time in vivo, and permits multiple drug loading, all of which could improve drug efficacy and reduce toxicity. Furthermore, nanomedicine can maintain the synergistic ratio of the drugs; deliver the drugs to the tumor at the same time, such that two or more drugs of tumor treatment achieve synchronization in time and space; and alter the pharmacokinetics and distribution profile in vivo such that these are dependent on nanocarrier properties (rather than being dependent on the drugs themselves).

Conclusion:

Therefore, nanomedicine-mediated combination drug therapy is promising in the treatment of tumors.

Keywords: Nanomedicine, Combined chemotherapy, Combination drug therapy, Tumor.



1. INTRODUCTION

1.1. Tumor Chemotherapy

Tumor chemotherapy refers to the use of chemical substances to treat cancer. It is a systemic treatment that can effectively kill tumor cells, inhibit the growth of tumors, and improve the survival rate. However, due to poor targeting, chemotherapy drugs not only kill tumor cells, but also damage the body's normal cells, resulting in a series of toxic side effects [1]. These include bone marrow suppression, cumulative cardiotoxicity, neutropenia, alopecia, vomiting, etc. Furthermore, the development of multidrug resistance(MDR) is a major obstacle for tumor chemotherapy [1, 2]. Generally, the toxic side effects and MDR of chemotherapy could be partially overcome by the combined chemotherapy.

1.2. Combined Chemotherapy

Because a single chemotherapeutic agent is ineffective in tumor treatment, the use of two or more drugs simultaneously or sequentially has become commonplace; this regimen is called combined chemotherapy [3-5]. This approach needs to abide by the following principles: (1) each chemotherapeutic drug should be effective in isolation; (2) the mechanism by which the drugs act should be different; (3) the combined effect of the chemotherapeutic agents should be additive or synergistic; (4) toxicity profiles should not overlap; and (5) the drug combination must have an acceptable therapeutic window [5, 6]. When compared with individual drug approaches, combined chemotherapy can reduce the risk of MDR and improve the therapeutic effect, as well as avoiding the side effects associated with the long-term use of a single drug [7].

To achieve the best therapeutic effect, it is necessary to determine optimal drug ratios in order to maximize drug synergy. A variety of mathematical methods have been used to calculate the interactions between drug combinations (namely synergistic, additive, or antagonistic) [8, 9]. The median-effect method of Chou and Talalay is the most common method used for combination drug analysis because it utilizes CalcuSynsoftware to evaluate the optimal drug ratio [10]. The combination index (CI) develops this median-effect method further [11], and states that: CI=(D1)/(DX)1+(D2)/(DX)2, (where (DX)1 and (DX)2 are the concentrations of drug1 and 2 that inhibit the rate of tumor cell proliferation at X% in isolation, and D1 and D2 are the concentrations of drug1 and 2 in combination that inhibit the rate of tumor cell proliferation at X%). When CI=1, the drug interactions are additive, whereas the combination drugs act synergistically when CI<1 and antagonistically when CI>1.

However, there are still some factors, which may contribute to the failure of combined chemotherapy, for example, uncertainty of drug synergy, difficulty in the achievement of the optimal drug synergistic ratio, and inconsistency of drug uptake into the tumor [12].

1.3. Nanomedicine

Nanomedicine is a safe and effective form of drug delivery. These formulations use natural or synthetic polymeric materials to encapsulate or absorb drugs, and the carrier materials include polymers, liposomes, micelles, proteins, and metallic materials. Compared with traditional drug delivery systems, nanomedicine has the following advantages: it increases the bioavailability of poorly soluble drugs, prolongs drug circulation time, increases penetrability, permits loading of a variety of drugs, improves drug efficacy and targeting, and reduces toxicity [13, 14]. These advantages have contributed to the research interest in nanomedicine-mediated combined chemotherapy and resulted in the successful development of some novel nanomedicine-anticancer drugs.

1.4. Nanomedicine-Mediated Combined Chemotherapy

As previously mentioned, combined chemotherapy has certain drawbacks, which result in failure to kill tumor cells. Nanomedicine may solve this problem by maintaining the optimal synergistic ratio of the drugs, delivering them to the tumor simultaneously, and altering the pharmacokinetic and distribution profile in vivo because these are dependent on nanocarrier properties (rather than being dependent on the drugs themselves) [15]. Thus, nanomedicine-mediated combination drug therapy appears to be very promising for tumor treatment and there are several drugs currently in clinical trials.

2. NANOPARTICLE-MEDIATED COMBINED CHEMOTHERAPY

Nanoparticles are usually prepared from natural or synthetic polymeric materials using supercritical fluid technology [16], the solvent evaporation technique [17], and high pressure homogenization [18]. Ideal polymeric materials should be biodegradable and biocompatible. Examples of natural polymers include gelatin, chitosan, alginate, gliadin, etc., while those of synthetic polymers include polylactic acid (PLA), polyglycolic acid (PGA), poly (lactic-co-glycolic) acid (PLGA), poly (alkylcyanoacrylate) (PACA), etc. However, some non-biodegradable polymeric materials can also be used for the preparation of nanoparticles such as polymethyl methacrylate (PMA) and polymethyl methacrylate (PMMA) or similar materials.

Table 1 gives several examples of recent research reports relating to nanoparticle-mediated combined chemotherapy [15, 19-25, 27-30]. For instance, co-delivery of docetaxel (DTX) and tanespimycin (17-AAG) by hyaluronic acid (HA)-modified PLGA nanoparticles had shown the highest synergistic effect in killing MCF-7, MDA-MB-231 and SCC-7 cells at the molar ratio of 2:1 (DTX:17-AGG), and this synergistic antitumor activity was also demonstrated in vivo [19]. Furthermore, Wang et al. [20] developed PEGylated PLGA nanoparticles for co-encapsulating paclitaxel (PTX) and etoposide (ETP) and demonstrated that co-delivery system enhanced cell cycle arrest and apoptosis of the human osteosarcom cells and improved cytotoxic activity of chemotherapeutic drugs.

Table 1. Nanoparticle-mediated combined chemotherapy.
Combined Chemotherapeutics Drug delivery system Type of cancer Ref.
Docetaxel and tanespimycin Hyaluronic acid-decorated PLGA nanoparticles Breast cancer, squamous cell carcinoma [19]
Paclitaxel and etoposide PEG-PLGA nanoparticles Osteosarcoma [20]
Cisplatin and paclitaxel Folic acid modified PEG-PLGA nanoparticles Non-small lung cancer [21, 22]
Cisplatin and doxorubicin Transferrin modified PEG-PLGA nanoparticles Human hepatoma carcinoma [23]
Combretastatin-A4 phosphate and doxorubicin mPEG-PLA nanoparticles Human nasopharyngeal epidermal carcinoma [24]
Paclitaxel and tetrandrine CTAB@MSN Breast cancer [25]
mPEG-PCL nanoparticles Gastric cancer [27]
PEG-b-PCL nanoparticles Gastric cancer [28]
Pirarubicin and paclitaxel Human serum albumin nanoparticles Breast cancer [15]
Hypocrellin B and paclitaxel Hyaluronic acid–ceramide nanoparticles Lung cancer [29]
Doxorubicin and paclitaxel mPEsG-b-PLG-b-PLL/DOCA nanovehicle Non-small cell lung cancer [30]

PEG-PLGA and PEG-PLA nanoparticles have also been chemically altered to improve tumor targeting. For example, He et al. [21, 22] utilized folic acid (FA)-modified PEG-PLGA nanoparticles for the co-delivery of cisplatin (CDDP) and PTX. This approach inhibited non-small cell lung cancer with a drug concentration ratio of 1:2 (CDDP:PTX). Similarly, Zhang [23] and colleagues designed transferrin-modified PEG-PLGA nanoparticles (Tf-DDP/ DOX NPs) for co-delivery of CDDP and doxorubicin (DOX). Compared with free drugs, Tf-DDP/DOX NPs demonstrated greater antitumor activity for hepatoma carcinoma both in vivo and in vitro. Finally, Zhang et al. [24] synthesized methoxy poly (ethylene glycol)-b-polylactide (mPEG-PLA) diblock copolymers by ring-opening polymerization. Combretastatin-A4 phosphate (CA4P) and DOX were loaded onto these mPEG-PLA nanoparticles, and demonstrated effective synergistic cytotoxicity, inhibiting human nasopharyngeal carcinoma angiogenesis and preventing proliferation of human nasopharyngeal epithelial cancer cells when the drug ratio of DOX:CA4P was 1:10.

In addition to using polymeric materials, inorganic materials, ceramides, and human serum albumin can also be used as nanocarriers. Human serum albumin (HSA), a protein found in the human serum, is particularly suited as a nanocarrier material due to its non-toxicity, biocompatible, and biodegradable [26]. Yi et al. [15] prepared a co-delivery system of PTX and pirarubicin (THP) by using HSA, and their results indicated that the nanoparticles had greater cytotoxicity against 4T1 breast cancer cells when compared with single or free drug delivery. Furthermore, the side effects of the drugs were significantly reduced, including bone marrow suppression, and organ or gastrointestinal toxicity.

3. LIPOSOME-MEDIATED COMBINED CHEMOTHERAPY

Liposomes are spherical envelopes with a lipoid bilayer, adapted to encapsulate drugs [31]. When compared with nanoparticles, liposomes have greater biocompatibility, and some liposome-mediated combined chemotherapy preparations have entered into clinical trials. There searchers at Celator Pharmaceuticals utilized distearoylphosphatidylglycerol (DSPG), distearoylphosphatidylcholine (DSPC), and cholesterol (Chol) to prepare liposomes for co-encapsulating cytarabine (Ara-C) and daunorubicin (DNR) at a molar ratio of 5:1. The resulting drug, CPX-351, significantly inhibited leukemia cells in vitro (while maintaining tumor cell selectivity), and the two drugs still acted synergistically in vivo, with similar anti-leukemia effects [31-33]. Furthermore, CPX-351 is in stage III of clinical trials, and the latest reports indicate that the US FDA has granted breakthrough therapy status to Celator Pharmaceuticals’ CPX-351 for the treatment of adults with therapy-related acute myeloid leukemia (t-AML) and AML with myelodysplasia-related changes (AML-MRC).

A list of recent studies concerning liposome-mediated combined chemotherapy is presented in Table 2 [34-44]. For example, Riviereet et al. [34] compared the antitumor effect of co-delivery of the synergistic drugs fluoroorotic acid (FOA) and irinotecan (IRN) in the same liposome versus delivery of the same drugs in separate liposomes. The co-delivery system was more successful in inhibiting the proliferation of C26 cells in vitro; however, the drugs did not have synergistic effects in the C26 tumor mouse model [34]. These results demonstrate the challenges associated with developing synergistic treatment protocols in vivo based on in vitro studies.

Table 2. Liposome-mediated combined chemotherapy.
Combined Chemotherapeutics Drug delivery system Type of cancer Ref.
Fluoroorotic acid and irinotecan Liposome Colorectal cancer [34]
Salinomycin and chloroquine Liposome Human hepatoma carcinoma [35]
Salinomycin and Doxorubicin Liposome Human hepatoma carcinoma [36]
Doxorubicin and erlotinib Liposome Triple negative breast cancer, Non-small cell lung cancer [37]
Omacetaxine Mepesuccinate and doxorubicin Liposome Human cervical carcinoma [38]
Combretastatin A-4 and doxorubicin Octreotide-modified stealth liposomes Non-small cell lung cancer, Breast cancer [39]
Paclitaxel and doxorubicin Liposome Lung cancer [40]
Paclitaxel and lonidamine Liposome Breast cancer [41]
Topotecan and vincristine Liposome Daoy tumors [42]
Vincristine and quercetin Liposome Breast tumor [43]
Vincristine and temozolomide Solid lipid nanoparticles Glioma [44]

Our group reported an effective strategy for targeting liver cancer cells (HepG2) and liver cancer stem cells (HepG2-TS) by developing liposomes for co-delivery of salinomycin (SAL) and chloroquine (CQ) [35]. After screening, the synergistic molar ratio of SAL:CQ was 1:5, the liposome particle size was about 120nm, and the PDI was greater. In vitro cytology results showed that combining drugs could improve the cytotoxic effect of SAL in HepG2 cells, but not in HepG2-TS. We surmised that the mechanism might be related to the basal autophagy flux, as autophagy plays a protective role in liver cancer. Therefore, we measured the basal autophagy flux of the two cell types, and demonstrated that it was significantly greater in HepG2 cells. When SAL was combined with CQ in HepG2 cells,the antitumor effect of SAL was improved because CQ could inhibit autophagy [35]. Furthermore, our group loaded SAL and DOX into liposomes separately, and in combination, to assess the subsequent antitumor activity in liver cancer. The results showed that co-delivery liposomes and separate liposomes had similar antitumor activity [36].

In addition, Morton et al. used the chemical properties of liposomes and drugs to create a controlled sequence of drug release [37]. The hydrophobic drug erlotinib and the hydrophilic drug DOX were encapsulated in the hydrophobic compartment and the hydrophilic interior of liposomes, respectively. This enabled them to achieve the desired time sequence of drug release in which erlotinib first inhibited EGFR activity in cancer cells and changed the intracellular apoptotic pathways, thus improving the sensitivity of breast cancer cells to the later release of DOX. Encouragingly, this time-staggered drug delivery could potentially enhance antitumor effects in vivo [37].

4. LIPID-POLYMER HYBRID NANOPARTICLE-MEDIATED COMBINED CHEMOTHERAPY

Lipid-polymer hybrid nanoparticles are prepared using polymeric and lipid materials which exhibit a core-shell structure [45]. They possess a wide range of potential applications, as they overcome some of the drawbacks associated with liposome nanoparticles: they are less physically and chemically unstable and they are easier to store, thus reducing drug leakage into the circulation. Furthermore, they have greater drug loading capacity than polymer nanoparticles, and exhibit better tumor targeting [45-47]. The recent research regarding lipid-polymer hybrid nanoparticle-mediated combined chemotherapy is summarized in Table 3 [48-52].

Table 3. Lipid-polymer hybrid nanoparticle-mediated combined chemotherapy.
Combined Chemotherapeutics Drug delivery system Type of cancer Ref.
Doxorubicin and curcumin Lipid nanoparticles Hepatoma carcinoma [48]
Doxorubicin and sorafenib Lipid-polymer hybrid nanoparticles Hepatocellular carcinoma [49]
Axitinib and celastrol PEGylated lipid bilayer-supported mesoporous silica nanoparticles Squamous carcinoma, breast cancer, neuroblastoma [50]
Paclitaxel and curcumin PEGylated hybrid liposomes-albumin nanoparticles Breast cancer, melanoma cells [51]
Sorafenib and quercetin RGD peptide modified lipid coated nanoparticles Hepatocellular carcinoma [52]

Of particular note, Zhao [48] and colleagues employed lipid-polymer hybrid nanoparticles to encapsulate curcumin (CUR) and DOX to investigate their antitumor activity on diethylnitrosamine (DEN)-induced hepatocellular carcinoma in mice. The results showed that co-delivery of CUR and DOX in this way enhanced their antitumor effects, promoting apoptosis of tumor cells and inhibiting both tumor cell proliferation and angiogenesis. Further analysis also indicated that DOX/CUR-NPs had greater antitumor effects than DOX-NPs, which was related to regulation of the levels of hypoxia-associated mRNA and protein, as well as drug resistance [48]. In another study, Zhang et al. [49] successfully bound iRGD peptide to lipid-polymer hybrid nanoparticles in order to enhance the antitumor efficacy of DOX and sorafenib (SOR). Compared with the free drugs and nanoparticles without iRGD peptide modification, DOX+SOR/iRGD-NPs had greater synergistic cytotoxic effects; they also prolonged the cycle time, improved the biocompatibility, and enhanced endocytosis and drug accumulation in the tumor. In a mouse model of liver cancer, the tumor inhibition effect of these DOX+SOR/iRGD-NPs was even more significant [49].

Furthermore, although lipid-polymer hybrid nanoparticles are generally prepared using polymeric and lipid materials, they have also been produced from inorganic materials and liposomes. In a recent research by Choi et al. [50], combined ACML nanoparticles were formed by loading axitinib (AXT) in PEGylated lipid bilayers and celastrol (CST) in MSN. Cytological experiments demonstrated that ACML could inhibit both angiogenesis and mitochondrial function and induce apoptosis in several cell types (SCC-7, BT-474 breast cancer cells and SH-SY5Y neuroblastoma cells). Notably, the tumor inhibition effects were even more significant in mouse tumor models.

5. POLYMER MICELLE-MEDIATED COMBINED CHEMOTHERAPY

Another promising form of nanocarrier is the polymer micelle, and several drug-loaded micelles have entered into clinical trials (for example, the PTX-loaded polymer micelles, NK105, and the CDDP-loaded polymer micelles, NC-6004) [53-55]. Table 4 provides an overview of recent studies using polymer micelle-mediated combined chemotherapy [56-62].

Table 4. Polymer micelle-mediated combined chemotherapy.
Combined Chemotherapeutics Drug delivery system Type of cancer Ref.
Cisplatin and paclitaxel PEG–PGlu90–PPhe25 hybrid micelles Ovarian cancer [56]
Linear-dendritic telodendrimer micelles Ovarian cancer [57]
Doxorubicin and disulfiram Polymeric micelles Breast cancer [58]
Doxorubicin and curcumin Polymeric micelles Breast cancer [59]
Doxorubicin and paclitaxel Pluronic mixed micelles Breast cancer [60]
Docetaxel and chloroquine PEO–PPO–PCL/TPGS micelles Breast cancer [61]
Paclitaxel and curcumin PEG-PE and vitamin E micelles Ovarian cancer [62]

In order to eradicate ovarian cancer, Desale et al. [56] synthesized biodegradable triblock copolymers containing ethylene glycol, glutamic acid, and phenylalanine (PEG–PGlu–PPhe). These copolymers self-assembled to form micelles for the co-delivery of CDDP and PTX, and the resulting nanomedicine mounted strong tumor suppression effects both in vitro and in vivo. Similarly, Cai et al. [57]. used polymeric micelles containing CDDP and PTX for the treatment of ovarian cancer. They developed three-layered linear-dendritic telodendrimer micelles and loaded drugs at a 2:1 molar ratio of CDDP:PTX. Compared with the free drug combination and loading in separate micelles, the co-delivery micelles provided greater tumor targeting and prolonged drug half-life. Moreover, the antitumor effect was significant and it could enhance the survival of mice [57].

Finally, Duan et al. [58] developed a pH-sensitive polymeric micelle system to achieve the temporal release of two drugs. The researchers conjugated DOX to a poly (styrene-co-maleic anhydride) (SMA) derivative with adipic dihydrazide (ADH) through an acid-cleavable hydrazone bond. They then loaded disulfiram (DSF) into the polymer micelles through the self-assembly of a SMA-ADH-DOX (SAD) conjugate [58]. This nanomedicine rapidly released DSF to inhibit the activity of P-glycoprotein and then slowly released DOX to increase its accumulation within tumor cells and enhance the antitumor response.

6. OTHER NANOMEDICINE-MEDIATED COMBINED CHEMOTHERAPY

Apart from the previously mentioned nanocarriers, microemulsions and microspheres are also traditional drug carriers. Owing to the relatively large particle size, these drug carriers have some unique properties such as a high drug-loading capacity and reduced drug leakage. Noh et al. [63] designed and prepared multi-prodrug nanocarriers (MPDNCs), with PTX being conjugated with polylysine carboxylate to form a cationic polymer (PLL-PTX), and HA and gemcitabine (GEM) combined to form an anionic polymer (HA-GEM). The two types of polymer then formed the multi-prodrug nanocarrier through electrostatic interaction. Because HA binds to the CD44 receptor, these MPDNCs targeted the CD44 over expression of the biliary cancer cell lines, HuCCT1, and therefore markedly inhibited cancer cell proliferation. Furthermore, they also induced cancer cell apoptosis and improved anti-tumor activity [63].

Similarly, Lee et al. [64] prepared double-layered microparticles using DOX loaded into the PLGA shell and PTX loaded into the poly (L-lactic acid) (PLLA) core. By changing the polymer ratio, the burst release of DOX was controlled and the drug release time prolonged to 2 months. The double-layered microparticles significantly reduced the rate of formation of microspheres and had a good antitumor effect when incubated with MCF-7 microspheres [64].

7. SEPARATE NANOMEDICINE-MEDIATED COMBINED CHEMOTHERAPY

As discussed above, co-delivery of chemotherapeutic agents in nanoparticles can improve the effects of combined chemotherapy for the treatment of tumors. However, there is also evidence that loading single drugs into separate nanoparticles can also achieve the desired antitumor effect. These preparations overcome certain difficulties associated with co-delivery, namely that the drug combinations are flexible, the synergistic ratio of the drugs is maintained, and drugs with disparate chemical properties can be effectively delivered within their own uniquely tailored carrier particles. Nevertheless, it is difficult to control the synergistic ratio and the concentration of the two drugs at the target site in vivo, so the success of this strategy is dependent upon the specific circumstances in which it is used.

In general, two kinds of chemotherapeutic drugs are respectively loaded into the carriers to target both cancer stem cells and tumor cells. Cancer stem cells are a form of undifferentiated or primitive cells, with the capacity for multipotent differentiation and self-renewal ability [65-69]. Our group has used this strategy previously to target MCF-7 and MCF-7 microspheres [70]. In this study, we used silane coupling agent-modified MSN to separately encapsulate 8-hydroxyquinoline (8-HQ) and DTX. The DTX-loaded MSN and 8-HQ-loaded MSN were then covered with lipid bilayers and HA-bound lipid bilayers, respectively, producing DTX-MSS and 8-HQ-HA-MSS. Our results demonstrated that DTX-MSS was more effective in inhibiting MCF-7 than MCF-7 microspheres, whereas the role of 8-HQ-HA-MSS was just the opposite . Therefore, combining the two forms of nanomedicine resulted in better antitumor activity and reduced drug toxicity in normal cells [70].

In another example of this approach, Ke et al. [71] used polymer micelles to target tumor cells and cancer stem cells. The polymer micelles were prepared using two diblock copolymers, one of PEG and urea-functionalized polycarbonate (PEG-PUC), and another of PEG and acid-functionalized polycarbonate (PEG-PAC) in a 1:1 molar ratio. Thioridazine (THZ) and DOX were loaded into these polymer micelles, respectively (THZ-MM and DOX-MM). It has been reported that THZ could kill cancer stem cells [72].The results confirmed that the free drug THZ and THZ-MM had inhibitory effect on cancer stem cells, and both the free drugs in combination and the polymer micelle combination could effectively inhibit the proliferation of cancer stem cells. However, the polymer micelle combination was found to have the most significant antitumor effect in a BT-474 nude mice tumor model [71].

Finally, Li et al. [73] demonstrated that decitabine (DAC) and DOX-loaded nanoparticles had the best inhibitory effect on cancer stem cells.

CONCLUSION

Cancer is one of the main threats to human health; therefore, the question of how to cure it has become an important research topic worldwide. Currently, chemotherapy is a common mode of treatment, and the combination of two or more chemotherapeutic agents greatly improves the therapeutic effect. However, this combined strategy has significant drawbacks, namely drug toxicity and difficulty maintaining the synergistic ratio of the drugs, which leads to treatment failure. Nanomedicine may provide a solution for this problem by increasing the antitumor activity of combined chemotherapy through novel delivery methods. At present, however, few preparations progress to the clinical research stage, and the scientific community faces challenges in terms of enhancing the antitumor effects, overcoming practical application problems, and expanding the scope of nanomedicine-mediated combined chemotherapy. We believe that with scientific and technological progress, these challenges are not insurmountable and that nanomedicine-mediated combined chemotherapy will succeed in the treatment of cancer.

CONFLICT OF INTEREST

The authors confirm that this article content has no conflict of interest.

ACKNOWLEDGEMENTS

This work was supported by the National Natural Science Foundation of China (Grant No. 81573376, 81472829).

REFERENCES

[1] Ge Y, Ma Y, Li L. The application of prodrug-based nano-drug delivery strategy in cancer combination therapy. Colloids Surf B Biointerfaces 2016; 146: 482-9.
[2] Asghar U, Meyer T. Are there opportunities for chemotherapy in the treatment of hepatocellular cancer? J Hepatol 2012; 56(3): 686-95.
[3] Rodon J, Perez J, Kurzrock R. Combining targeted therapies: practical issues to consider at the bench and bedside. Oncologist 2010; 15(1): 37-50.
[4] Ma L, Kohli M, Smith A. Nanoparticles for combination drug therapy. ACS Nano 2013; 7(11): 9518-25.
[5] Woodcock J, Griffin JP, Behrman RE. Development of novel combination therapies. N Engl J Med 2011; 364(11): 985-7.
[6] Guidance for industry: codevelopment of two or more unmarketed investigational drugs for use in combination. Food and Drug Administration 2013. Available from: http://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ ucm236669.pdf
[7] Zhao X, Chen Q, Li Y, Tang H, Liu W, Yang X. Doxorubicin and curcumin co-delivery by lipid nanoparticles for enhanced treatment of diethylnitrosamine-induced hepatocellular carcinoma in mice. Eur J Pharm Biopharm 2015; 93: 27-36.
[8] Berenbaum MC. Isobolographic, algebraic, and search methods in the analysis of multiagent synergy. Int J Toxicol 1988; 7(7): 927-38.
[9] Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul 1984; 22: 27-55.
[10] Greco WR, Bravo G, Parsons JC. The search for synergy: a critical review from a response surface perspective. Pharmacol Rev 1995; 47(2): 331-85.
[11] Chou TC, Talalay P. Generalized equations for the analysis of inhibitions of Michaelis-Menten and higher-order kinetic systems with two or more mutually exclusive and nonexclusive inhibitors. Eur J Biochem 1981; 115(1): 207-16.
[12] Dicko A, Mayer LD, Tardi PG. Use of nanoscale delivery systems to maintain synergistic drug ratios in vivo. Expert Opin Drug Deliv 2010; 7(12): 1329-41.
[13] Lu B, Huang X, Mo J, Zhao W. Drug delivery using nanoparticles for cancer Stem-Like cell targeting. Front Pharmacol 2016; 7(65103): 84.
[14] Shen S, Xia JX, Wang J. Nanomedicine-mediated cancer stem cell therapy. Biomaterials 2016; 74: 1-18.
[15] Yi X, Lian X, Dong J, et al. Co-delivery of Pirarubicin and Paclitaxel by human serum albumin nanoparticles to enhance antitumor effect and reduce systemic toxicity in breast cancers. Mol Pharm 2015; 12(11): 4085-98.
[16] York P, Kompella UB, Shekunov BY. Supercritical fluid technology for drug product development. New York: Marcel Dekker 2004.
[17] Lee M, Cho YW, Park JH, et al. Size control of self-assembled nanoparticles by an emulsion/solvent evaporation method. Colloid Polym Sci 2006; 284(5): 506-12.
[18] Wan X, Zheng X, Pang X, et al. The potential use of lapatinib-loaded human serum albumin nanoparticles in the treatment of triple-negative breast cancer. Int J Pharm 2015; 484(1-2): 16-28.
[19] Pradhan R, Ramasamy T, Choi JY, et al. Hyaluronic acid-decorated poly(lactic-co-glycolic acid) nanoparticles for combined delivery of docetaxel and tanespimycin. Carbohydr Polym 2015; 123: 313-23.
[20] Wang B, Yu XC, Xu SF, Xu M. Paclitaxel and etoposide co-loaded polymeric nanoparticles for the effective combination therapy against human osteosarcoma. J Nanobiotechnology 2015; 13(1): 22.
[21] He Z, Huang J, Xu Y, et al. Co-delivery of cisplatin and paclitaxel by folic acid conjugated amphiphilic PEG-PLGA copolymer nanoparticles for the treatment of non-small lung cancer. Oncotarget 2015; 6(39): 42150-68.
[22] He Z, Shi Z, Sun W, et al. Hemocompatibility of folic-acid-conjugated amphiphilic PEG-PLGA copolymer nanoparticles for co-delivery of cisplatin and paclitaxel: treatment effects for non-small-cell lung cancer. Tumour Biol 2016; 37(6): 7809-21.
[23] Zhang X, Li J, Yan M. Targeted hepatocellular carcinoma therapy: transferrin modified, self-assembled polymeric nanomedicine for co-delivery of cisplatin and doxorubicin. Drug Dev Ind Pharm 2016; 42(10): 1590-9.
[24] Zhu J, Xu X, Hu M, Qiu L. Co-encapsulation of combretastatin-A4 phosphate and doxorubicin in polymersomes for synergistic therapy of nasopharyngeal epidermal carcinoma. J Biomed Nanotechnol 2015; 11(6): 997-1006.
[25] Jia L, Li Z, Shen J, et al. Multifunctional mesoporous silica nanoparticles mediated co-delivery of paclitaxel and tetrandrine for overcoming multidrug resistance. Int J Pharm 2015; 489(1-2): 318-30.
[26] Kratz F. Albumin as a drug carrier: design of prodrugs, drug conjugates and nanoparticles. J Control Release 2008; 132(3): 171-83.
[27] Zhang H, Tian Y, Zhu Z, et al. Efficient antitumor effect of co-drug-loaded nanoparticles with gelatin hydrogel by local implantation. Sci Rep 2016; 6: 26546.
[28] Chang JE, Cho HJ, Yi E, Kim DD, Jheon S. Hypocrellin B and paclitaxel-encapsulated hyaluronic acid-ceramide nanoparticles for targeted photodynamic therapy in lung cancer. J Photochem Photobiol B 2016; 158: 113-21.
[29] Lv S, Tang Z, Li M, et al. Co-delivery of doxorubicin and paclitaxel by PEG-polypeptide nanovehicle for the treatment of non-small cell lung cancer. Biomaterials 2014; 35(23): 6118-29.
[30] Li X, Lu X, Xu H, et al. Paclitaxel/tetrandrine coloaded nanoparticles effectively promote the apoptosis of gastric cancer cells based on oxidation therapy. Mol Pharm 2012; 9(2): 222-9.
[31] Tardi P, Johnstone S, Harasym N, et al. In vivo maintenance of synergistic cytarabine:daunorubicin ratios greatly enhances therapeutic efficacy. Leuk Res 2009; 33(1): 129-39.
[32] Kim HP, Gerhard B, Harasym TO, Mayer LD, Hogge DE. Liposomal encapsulation of a synergistic molar ratio of cytarabine and daunorubicin enhances selective toxicity for acute myeloid leukemia progenitors as compared to analogous normal hematopoietic cells. Exp Hematol 2011; 39(7): 741-50.
[33] Lim WS, Tardi PG, Dos Santos N, et al. Leukemia-selective uptake and cytotoxicity of CPX-351, a synergistic fixed-ratio cytarabine:daunorubicin formulation, in bone marrow xenografts. Leuk Res 2010; 34(9): 1214-23.
[34] Riviere K, Kieler-Ferguson HM, Jerger K, Szoka FC Jr. Anti-tumor activity of liposome encapsulated fluoroorotic acid as a single agent and in combination with liposome irinotecan. J Control Release 2011; 153(3): 288-96.
[35] Xie F, Zhang S, Liu J, et al. Codelivery of salinomycin and chloroquine by liposomes enables synergistic antitumor activity in vitro. Nanomedicine (Lond) 2016; 11(14): 1831-46.
[36] Gong Z, Chen D, Xie F, et al. Codelivery of salinomycin and doxorubicin using nanoliposomes for targeting both liver cancer cells and cancer stem cells. Nanomedicine (Lond) 2016; 11(19): 2565-79.
[37] Morton SW, Lee MJ, Deng ZJ, et al. A nanoparticle-based combination chemotherapy delivery system for enhanced tumor killing by dynamic rewiring of signaling pathways. Sci Signal 2014; 7(325): ra44.
[38] Shim G, Lee S, Choi J, Lee S, Kim CW, Oh YK. Liposomal co-delivery of omacetaxine mepesuccinate and doxorubicin for synergistic potentiation of antitumor activity. Pharm Res 2014; 31(8): 2178-85.
[39] Dai W, Jin W, Zhang J, et al. Spatiotemporally controlled co-delivery of anti-vasculature agent and cytotoxic drug by octreotide-modified stealth liposomes. Pharm Res 2012; 29(10): 2902-11.
[40] Wang Y, Zhang H, Hao J, et al. Lung cancer combination therapy: co-delivery of paclitaxel and doxorubicin by nanostructured lipid carriers for synergistic effect. Drug Del 2015; pp. 1-6.
[41] Wang Y, Zhang H, Hao J, Li B, Li M, Xiuwen W. Lung cancer combination therapy: co-delivery of paclitaxel and doxorubicin by nanostructured lipid carriers for synergistic effect. Drug Deliv 2016; 23(4): 1398-403.
[42] Zucker D, Andriyanov AV, Steiner A, Raviv U, Barenholz Y. Characterization of PEGylated nanoliposomes co-remotely loaded with topotecan and vincristine: relating structure and pharmacokinetics to therapeutic efficacy. J Control Release 2012; 160(2): 281-9.
[43] Wong MY, Chiu GN. Liposome formulation of co-encapsulated vincristine and quercetin enhanced antitumor activity in a trastuzumab-insensitive breast tumor xenograft model. Nanomedicine (Lond) 2011; 7(6): 834-40.
[44] Wu M, Fan Y, Lv S, et al. Vincristine and Temozolomide combined chemotherapy for the treatment of glioma: a comparison of solid lipid nanoparticles and nanostructured lipid carriers for dual drugs delivery. Drug Del 2015; pp. 1-6.
[45] Hadinoto K, Sundaresan A, Cheow WS. Lipid-polymer hybrid nanoparticles as a new generation therapeutic delivery platform: a review. Eur J Pharm Biopharm 2013; 85(3 Pt A): 427-43.
[46] Mandal B, Bhattacharjee H, Mittal N, et al. Core-shell-type lipid-polymer hybrid nanoparticles as a drug delivery platform. Nanomedicine (Lond) 2013; 9(4): 474-91.
[47] Wu XY. Strategies for optimizing polymer-lipid hybrid nanoparticle-mediated drug delivery. Expert Opin Drug Deliv 2016; 13(5): 609-12.
[48] Zhao X, Chen Q, Li Y, Tang H, Liu W, Yang X. Doxorubicin and curcumin co-delivery by lipid nanoparticles for enhanced treatment of diethylnitrosamine-induced hepatocellular carcinoma in mice. Eur J Pharm Biopharm 2015; 93: 27-36.
[49] Zhang J, Hu J, Chan HF, Skibba M, Liang G, Chen M. iRGD decorated lipid-polymer hybrid nanoparticles for targeted co-delivery of doxorubicin and sorafenib to enhance anti-hepatocellular carcinoma efficacy. Nanomedicine (Lond) 2016; 12(5): 1303-11.
[50] Choi JY, Ramasamy T, Kim SY, et al. PEGylated lipid bilayer-supported mesoporous silica nanoparticle composite for synergistic co-delivery of axitinib and celastrol in multi-targeted cancer therapy. Acta Biomater 2016; 39: 94-105.
[51] Ruttala HB, Ko YT. Liposomal co-delivery of curcumin and albumin/paclitaxel nanoparticle for enhanced synergistic antitumor efficacy. Colloids Surf B Biointerfaces 2015; 128: 419-26.
[52] Wang C, Su L, Wu C, Wu J, Zhu C, Yuan G. RGD peptide targeted lipid-coated nanoparticles for combinatorial delivery of sorafenib and quercetin against hepatocellular carcinoma. Drug Dev Ind Pharm 2016; 42(12): 1938-44.
[53] Cabral H, Kataoka K. Progress of drug-loaded polymeric micelles into clinical studies. J Control Release 2014; 190: 465-76.
[54] Hamaguchi T, Kato K, Yasui H, et al. A phase I and pharmacokinetic study of NK105, a paclitaxel-incorporating micellar nanoparticle formulation. Br J Cancer 2007; 97(2): 170-6.
[55] Plummer R, Wilson RH, Calvert H, et al. A Phase I clinical study of cisplatin-incorporated polymeric micelles (NC-6004) in patients with solid tumours. Br J Cancer 2011; 104(4): 593-8.
[56] Desale SS, Cohen SM, Zhao Y, Kabanov AV, Bronich TK. Biodegradable hybrid polymer micelles for combination drug therapy in ovarian cancer. J Control Release 2013; 171(3): 339-48.
[57] Cai L, Xu G, Shi C, Guo D, Wang X, Luo J. Telodendrimer nanocarrier for co-delivery of paclitaxel and cisplatin: A synergistic combination nanotherapy for ovarian cancer treatment. Biomaterials 2015; 37: 456-68.
[58] Duan X, Xiao J, Yin Q, et al. Smart pH-sensitive and temporal-controlled polymeric micelles for effective combination therapy of doxorubicin and disulfiram. ACS Nano 2013; 7(7): 5858-69.
[59] Lv L, Qiu K, Yu X, et al. Amphiphilic Copolymeric Micelles for Doxorubicin and Curcumin Co-Delivery to reverse multidrug resistance in breast cancer. J Biomed Nanotechnol 2016; 12(5): 973-85.
[60] Chen Y, Zhang W, Huang Y, Gao F, Sha X, Fang X. Pluronic-based functional polymeric mixed micelles for co-delivery of doxorubicin and paclitaxel to multidrug resistant tumor. Int J Pharm 2015; 488(1-2): 44-58.
[61] Shi C, Zhang Z, Shi J, Wang F, Luan Y. Co-delivery of docetaxel and chloroquine via PEO-PPO-PCL/TPGS micelles for overcoming multidrug resistance. Int J Pharm 2015; 495(2): 932-9.
[62] Abouzeid AH, Patel NR, Torchilin VP. Polyethylene glycol-phosphatidylethanolamine (PEG-PE)/vitamin E micelles for co-delivery of paclitaxel and curcumin to overcome multi-drug resistance in ovarian cancer. Int J Pharm 2014; 464(1-2): 178-84.
[63] Noh I, Kim HO, Choi J, et al. Co-delivery of paclitaxel and gemcitabine via CD44-targeting nanocarriers as a prodrug with synergistic antitumor activity against human biliary cancer. Biomaterials 2015; 53: 763-74.
[64] Lee WL, Guo WM, Ho VH, et al. Delivery of doxorubicin and paclitaxel from double-layered microparticles: The effects of layer thickness and dual-drug vs. single-drug loading. Acta Biomater 2015; 27: 53-65.
[65] Song H, Su X, Yang K, et al. CD20 Antibody-Conjugated immunoliposomes for targeted chemotherapy of melanoma cancer initiating cells. J Biomed Nanotechnol 2015; 11(11): 1927-46.
[66] Su X, Song H, Niu F, et al. Co-delivery of doxorubicin and PEGylated C16-ceramide by nanoliposomes for enhanced therapy against multidrug resistance. Nanomedicine (Lond) 2015; 10(13): 2033-50.
[67] Yin C, Xie WF. Hepatocellular Carcinoma: Basic and transitional research. Gastrointest Tumors 2014; 1(2): 76-83.
[68] Yin C, Lin Y, Zhang X, et al. Differentiation therapy of hepatocellular carcinoma in mice with recombinant adenovirus carrying hepatocyte nuclear factor-4α gene. Hepatology 2008; 48(5): 1528-39.
[69] Yin C, Xie WF. Differentiation therapy with transcription factors might present as an ideal strategy for the treatment of cancer. Hepatology 2009; 50(6): 2046-7.
[70] Wang D, Huang J, Wang X, et al. The eradication of breast cancer cells and stem cells by 8-hydroxyquinoline-loaded hyaluronan modified mesoporous silica nanoparticle-supported lipid bilayers containing docetaxel. Biomaterials 2013; 34(31): 7662-73.
[71] Ke XY, Lin Ng VW, Gao SJ, Tong YW, Hedrick JL, Yang YY. Co-delivery of thioridazine and doxorubicin using polymeric micelles for targeting both cancer cells and cancer stem cells. Biomaterials 2014; 35(3): 1096-108.
[72] Sachlos E, Risueño RM, Laronde S, et al. Identification of drugs including a dopamine receptor antagonist that selectively target cancer stem cells. Cell 2012; 149(6): 1284-97.
[73] Li SY, Sun R, Wang HX, et al. Combination therapy with epigenetic-targeted and chemotherapeutic drugs delivered by nanoparticles to enhance the chemotherapy response and overcome resistance by breast cancer stem cells. J Control Release 2015; 205: 7-14.