Strategies of Overcoming the Physiological Barriers for Tumo

更新时间:2024-05-15 09:06:01 阅读量: 综合文库 文档下载

说明:文章内容仅供预览,部分内容可能不全。下载后的文档,内容与下面显示的完全一致。下载之前请确认下面内容是否您想要的,是否完整无缺。

Send Orders for Reprints to reprints@benthamscience.ae

6236

Current Pharmaceutical Design, 2015, 21, 6236-6245

Strategies of Overcoming the Physiological Barriers for Tumor-Targeted Nano-Sized Drug Delivery Systems

Yufang Pi 1, Jinge Zhou1, Jing Wang1, Jian Zhong2, Lin Zhang3, Yiting Wang1, Lei Yu1,* and Zhiqiang Yan1,*

Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal university, Shanghai 200062, P.R. China; 2College of Food Science & Technology, Shanghai Ocean University, Shanghai 201306, Shanghai 200241, P.R. China; 3Department of Pharmacy, Shaoxing People’s Hospital, Shaox-ing Hospital of ZheJiang University, Shaoxing 312000, P.R. China

Abstract: Nano drug delivery systems (NDDSs) have been widely used in tumor-targeted therapy since they can effectively reduce the side effects of traditional antitumor drugs and improve the anti-tumor effect. We divided the in vivo process of tumor-targeted NDDSs into seven steps: blood circulation, tumor accumulation, tumor tissue penetration, target cells internalization, lysosome escape, drug release and drug response. In each step, NDDSs will encounter different types of barriers preventing their effective delivery or response. The researchers have been making efforts to find different strategies of overcoming the corresponding barriers for NDDSs. Hence, we here

reviewed the recent progress of NDDSs in breaking the physiological barriers for more effective in vivo anti-tumor effect, in order to shed a new perspective on the development of tumor-targeted NDDSs.

1

Keywords: Nano drug delivery systems, tumor-targeted nanomedicines, pathophysiological basis, physiological barriers. INTRODUCTION

Cancer is one of the leading causes of death in the world. Cur-rently, the main treatments in clinic for cancer are surgery, chemo-therapy and radiation therapy. Although traditional chemotherapy can effectively control the growth of tumor, it could cause severe toxic effects on healthy tissues. Compared with traditional che-motherapeutic drugs, NDDSs have demonstrated great superiority due to their ability of improving the efficacy of chemotherapy and reducing the side effects [1-3].

The most common types of NDDSs include nanoparticles, liposomes, micelles, dendrimers, etc. [4]. According to the tumor targeting mechanism, the NDDSs can be divided into passive tar-geting and active targeting NDDSs. The passive targeting mecha-nism mainly involves the “Enhanced permeability and retention (EPR) effect” arisen from the abnormal, leaky and highly heteroge-neous microvasculature and the dysfunction of lymphatic drainage in tumor tissues [5]. The active targeting mechanism refers to the interaction of specific receptors overexpressed on target cells in tumor tissues and the targeting moieties conjugated to the surface of nanocarriers [6, 7]. Large amounts of reports on NDDSs have proved the effectiveness of the passive targeting and active target-ing mechanism on improving the therapeutic outcomes of NDDSs and reducing the side effect to normal tissues [8-12].

The NDDSs need to overcome a number of anatomical and physiological barriers before they can effectively arrive at target site and release drugs in tumor. We divide the systemic journey of tumor-targeted NDDSs into seven steps: blood circulation, tumor accumulation, tumor tissue penetration, target cell internalization, lysosome escape, drug release and drug response. In each step, there are different types of barriers which prevent the effective delivery or response of NDDSs. In order to improve the therapeutic outcomes, researchers have attempted many strategies on NDDSs design to overcome the physiological barriers in each step of the

*Address correspondence to these authors at the NO. 3663 Zhongshan Road, Shanghai 200062, P.R. China; E-mails: zqyan@sat.ecnu.edu.cn; yulei@nbic.ecnu.edu.cn

systemic journey. Hence, based on the analysis of physiological basis of tumor targeting, we here reviewed the different physiologi-cal barriers which may impede the systemic transport and response of tumor-targeted NDDSs, and the corresponding strategies of overcoming these barriers reported recently.

PATHOPHYSIOLOGICAL BASIS OF TUMOR TARGET-ING

Tumor grows and progresses in an intricate and complicated tumor microenvironment (TME). The TME contains a variety of components including vascular systems such as blood and lym-phatic vascular endothelial cells, dense extracellular matrix (includ-ing hyaluronic acid, smooth muscle actin, collagen fibers and pro-teolytic enzymes), hypoxia and oxidosis environment, a large num-ber of tumor cells and cancer stem cells (CSCs) [13]. As a unique microenvironment, it has the properties of leaky vasculatures, low pH, and high interstitial fluid pressure within the tumor, which have important influences on tumor proliferation, metastasis and deterio-ration [14]. The pathophysiological features of tumor tissue is the basis of designing the tumor-targeted NDDSs [15].

Tumor Cells

Different from normal cells, tumor cells allow themselves to grow out of control and become invasive. In order to meet the un-limited proliferation of tumor cells, a variety of receptors become over-expressed on the cell surface, such as transferrin receptors, folate receptors, low density lipoprotein receptors and tumor necro-sis factor receptor family [16]. Besides, there is also over-expressed P-glycoprotein (P-gp) on the surface of tumor cells. It can efflux antitumor drugs outside the cell, causing the decrease of intracellu-lar drug concentration and further the failure of chemotherapy, which is called tumor multidrug resistance (MDR) [17]. Almost all human tumor cells express different levels of P-glycoprotein, and those with insensitivity to chemotherapy tend to have higher ex-pression levels.

Vasculature

Tumor angiogenesis is the main source of nutrients and oxygen of tumor cells. When the tumor grows to a diameter of 1 to 2mm,

? 2015 Bentham Science Publishers

1873-4286/15 $58.00+.00

Overcoming Physiological Barriers of Tumor-targeted Nanomedicines the rapid and defective angiogenesis was induced in order to meet the demand of unlimited proliferation [18]. It requires tumor cells or stromal cells to secret a variety of stimulus factors, for example VEGF(vascular endothelial growth factor), to cause the prolifera-tion of endothelial cells and degradation of basement membrane and extracellular matrix, then the new capillaries founded with the form of migration and ‘sprouting’ of endothelial cells [19]. In con-trast to normal tissues, the vascular structure of tumor tissues is tortuous and aberrant, which is marked by a heterogeneous distribu-tion of vessel sizes and shapes [20]. The leaky blood vessels and the abnormally impaired lymphatic drainage make nanomedicines eas-ily accumulated and retention in the tumor tissues, which is so-called enhanced permeability and retention (EPR) effect. The EPR effect is the most important pharmacokinetic principle when de-signing tumor-targeted NDDSs [21].

Tumor Extracellular Matrix

Tumor extracellular matrix (ECM) is the structural support of the entire tumor, composed of hyaluronic acid (HA), tumor-associated fibroblasts (TAFs), smooth muscle actin (SMA), and collagen fibers, etc. [22]. The ECM of tumor is denser than normal tissue, causing a high intra-tumoral fluidic pressure, providing a significant transport barriers for nanomedicines penetrating into the tumor tissues [23]. In the process of tumor invasion and metastasis, the ECM is subjected to degradation and remodeling in order to degrade the basement membrane and the majority of proteins in the extracellular matrix [24]. This process is mainly regulated by many extracellular proteinases [25], such as matrix metalloproteinases (MMPs), a type of frequently used cleavable enzyme associated with tumor [26, 27].

Hypoxia and Oxidosis

Hypoxia and oxidosis are two basic characteristics of tumor tissues due to the abnormal nutrient metabolism. Intra-tumoral hy-poxia is generated as a result of oxygen consumption by the rapidly proliferating tumor cells, insufficient blood supply and poor lymph drainage. It has an extensive effect on a number of cellular path-ways and is one of the major contributors to drug resistance of nanomedicines [26, 28]. Besides, the anaerobic glycolysis in the hypoxia tissues can produce lactic acid. Due to the lack of tumor vascular systems, lactic acid cannot be fully discharged, thereby causing the tumor oxidosis [29]. Hypoxia and oxidosis are impor-tant events in tumor because they are related to a more aggressive phenotype with enhanced proliferation and metastasis formation, and poorer survival [30].

Cancer Stem Cells

According to the theory of tumor classification, there are two types of cells in tumor tissue: the first type is common tumor cells, which does not have the ability of tumor genesis and can be treated by chemotherapy; another is CSCs, which have the characteristics of self-renewal, proliferation and high tumorigenicity and show high resistance to drug treatment [31]. Many researches have proved that the CSCs is the main reason for tumor genesis, devel-opment, recurrence and metastasis, even if there are very few CSCs in tumor tissues. The insensitivity of CSCs to radiotherapy and chemotherapy may be resulted from that most of CSCs stay in sta-tionary phase, express a variety of ATP binding cassette transport-ers, and have DNA repair functions. This may be the key reason why tumors cannot be completely eliminated by the current clinical treatments [32].

PHYSIOLOGICAL BARRIERS OF NDDSs AND THE SOLV-ING STRATEGIES

As we mentioned above, there are seven steps in the systemic journey of tumor-targeted NDDSs before the drugs take effect. Each step is corresponding to one type of physiological barrier for the transport or response of NDDSs. Researchers have been finding

Current Pharmaceutical Design, 2015, Vol. 21, No. 42 6237

solutions of overcoming these barriers and achieving more potent anti-tumor effect. We describe each physiological barrier and the corresponding strategies as follows See Fig. (1) and Table 1. Blood Circulation

NDDSs have the characteristics of nano-size, large specific surface area and high surface activity. Thus, after intravenous injec-tion into the blood, NDDSs could adsorb a variety of ingredients on its surface such as apolipoproteins and complement proteins, form-ing what is known as the protein corona [33]. Protein corona can be easily identified by phagocytic cells, causing the opsonization and leading themselves to be involved in phagocytosis and rapidly cleared from the blood. It is essential for NDDSs to either bypass electrostatic interaction with charged proteins in the blood or avoid phagocytosis by special functional material modifications. Several surface modification platforms have been built in order to overcome limitations of NDDSs in the blood circulation. First, the presence of poly(ethylene glycol) (PEG) at the surface of nanocarriers has showed to clearly extend circulation lifetime in blood [34]. The extended circulation lifetime that PEG affords is attributed to the repulsive interaction with plasma components resulted from the hydrophilic shell surrounding the NDDSs [35]. Large amount of studies have been carried out on PEGylated NDDSs. For example, for brain tumor-targeted therapy, the PEG modified magnetic iron oxide nanoparticles (PEG-MNPs) achieved long circulating life-time, reduced concentrations in liver and spleen and increased drug accumulation in the brain compared with non-modified magnetic iron oxide nanoparticles (MNPs) [36, 37]. Other PEGylated NDDSs including PEG modified liposomes, PEG grafted copolymers, PEG coated polymer micelles and PEGylated dendrimers also showed prolonged circulation time for tumor therapy [35, 38-42]. Besides, other hydrophilic polymers such as hyaluronic acid (HA), Pluronic copolymer L61 and polyglycerol (PG) have also been proved to be able to prolong the blood circulation time of NDDSs [43-46].

Despite the effectiveness of PEGylation, PEG has been exten-sively observed as the cause of an unexpected immunogenic re-sponse known as the “accelerated blood clearance (ABC) phe-nomenon” [45]. Accordingly, researchers tried to use endogenous materials, for instance red blood cells (RBC) membrane, to enable active immune evasion of NDDSs [47]. Liangfang Zhang et al. prepared the RBC membrane coated PLGA(poly(lactic-co-glycolic acid) nanoparticles, which showed significantly longer retention time in the blood 72h post injection compared with PEGylated PLGA nanoparticles [48]. The RBC membrane obtained the long circulation time through the immunomodulatory proteins such as CD47 on the surface [49]. RBC membrane-cloaked polymeric nanoparticles represent a promising nanocarrier platform with ex-tended circulation time in vivo [50, 51].

Tumor Accumulation

Inefficiency of drug accumulation in tumor tissue is one of the obstacles in the process of successful treatment of tumors. The ac-cumulation of NDDSs is mainly achieved by exploiting the en-hanced permeability and retention (EPR) effect of tumor caused by the leaky vasculature and dysfunctional lymphatic drain [52]. How-ever, the heterogeneities of vascular permeability among different tumor type or location make the NDDSs accumulation unstable [53]. Thus nanomedicines should be further optimized into smaller scale in order to achieve both efficient blood delivery and high tumor site accumulation through the EPR effect [54].

Angiogenesis, induced by a range of angiogenesis factors, is one of the hallmarks of tumor [55]. As tumor vasculature could be directly accessible for NDDSs by specific receptors overexpressed on the endothelial cells, tumor vasculature targeting is becoming an effective strategy for tumor accumulation [56]. The expression of specific receptors on the surface of endothelial cells enables their recognition by ligand-modified nanomedicines, thus increased con-

6238 Current Pharmaceutical Design, 2015, Vol. 21, No. 42 Pi et al.

Fig. (1). Illustration of the biological barriers for tumor targeted NDDSs and the corresponding strategies of overcoming these barriers.

centration of nanomedicines accumulating on the tumor site. A large number of studies have focused on NDDSs modified by ligand which can specifically bind with the over-expressed recep-tors on endothelial cells. For example, since vascular endothelial growth factor receptor-2 (VEGFR2) is a overexpressed receptor on tumor endothelial cells, anti-VEGFR2 antibodies conjugated multi-stage nano-vectors (MSV) was developed and displayed a 4-fold increased targeting efficiency towards VEGFR2 expressing cells, whereas exhibiting minimal adherence to control cells [52]. Be-sides, as NGR(Asn-Gly-Arg) tripeptide ligand possess high affinity to CD13, a receptor up-regulated on the tumor vasculature and perivascular cells, NGR-functionalized nanoparticles were formu-

lated and showed high accumulation and deepest penetration at the glioma sites [55, 56]. Therefore, a growing body of evidence sug-gests that tumor accumulation of NDDSs by targeting receptors on the tumor vasculature is an effective strategy for tumor targeted drug delivery.

Tumor Tissue Penetration

Following penetrating tumor vessel walls and entering tumor tissue, NDDSs must penetrate through a thick tumor ECM to arrive at deep tumor tissue and be exposed to tumor cells. Solid tumors are characterized by an unusually dense and complex ECM composed of fibroblasts, hyaluronic acid (HA), and endothelial cells, etc. [57].

Overcoming Physiological Barriers of Tumor-targeted Nanomedicines The ECM often generates high interstitial fluid pressures (IFPs) and presents substantial barriers for NDDSs penetration, causing most NDDSs to be retained near the blood vessels and leaving most of tumor cells in deep tissue be exposed to few drugs [58]. Research-ers have attempted several strategies to increase the tumor tissue penetration of NDDSs, such as breaking up components of tumor stroma (such as TAFs, HA, etc.) and penetrating the ECM by tu-mor-penetrating peptide modification.

HA provides the primary barrier for transport of NDDSs in tumor stroma. In order to overcome this barrier, hyaluronidase agents have been used to loose the intercellular connective matrix of hyaluronic acid and ablate stroma from the dense ECM [57, 59]. Co-administration of hyaluronidase and antitumor drugs can cause ECM degradation and facilitate drug penetration into tumor tissues [59-61]. Besides, TAFs or myofibroblasts are a type of cells that plays a key role in the deposition of ECMs and constitutes a major portion of tumor stroma [62]. These fibroblasts present substantial barriers for NDDSs transport, especially in pancreatic cancer and bladder carcinomas [60, 62]. In order to penetrate the ECM of blad-der carcinomas, Leaf Huang group developed complex nanoparti-cles(ComboNP) which were able to break up the thick ECM, and showed a significantly enhanced anti-tumor effect [63].

Recently, tumor penetrating peptides (TPPs) have displayed their special advantages on the mediation of NDDSs penetration in tumor tissue. TPPs usually contain a key sequence motif R/KXXR/K at the C terminal, which allows their binding to the neuropilin-1 receptor overexpressed on tumor cells and tumor endo-thelial cells [64]. Many reports have demonstrated that TPP modifi-cation or co-administration can effectively increase the tumor pene-tration of NDDSs [14, 65-67]. For instance, co-administration of iRGD, a TPP, signi??cantly improved the therapeutic index of doxorubicin and doxorubicin liposomes on tumor [68]. We previ-ously prepared RGERPPR or RPARPAR peptide conjugated liposomes, which could penetrate into the whole tumor tissue, whereas plain liposome could only stay at the vicinity of tumor vessels [66, 69]. Therefore, TPP modification is an effective strat-egy for increasing tumor tissue penetration of NDDSs.

Target Cell Internalization

Following arriving at deep tumor tissues, NDDSs must to be internalized by tumor cells in order to play a good anti-tumor effect. There are mainly four strategies developed to facilitate NDDSs internalization: 1) cell penetrating peptide decorated NDDSs; 2) receptor-targeted NDDSs; 3) fusogenic NDDSs; 4) co-administration of P-gp inhibitors with NDDSs [70-73].

Cell-penetrating peptides (CPPs) are a type of peptides typically with a high relative abundance of positively charged amino acids or has sequences that contain an alternating pattern of polar/charged amino acids and non-polar/hydrophobic amino acids [74, 75]. Large numbers of traditional cell-penetrating peptides such as TAT pep-tide (AYGRKKRRQRRR), and R8 (RRRRRRRR) could be able to transport different molecules or anti-miRNAs through micelles, liposomes, complexes or other nanocarriers across biological barri-ers to be taken up by various cell lines [76-79]. Nowadays, re-searchers have developed a series of cell-penetrating peptides for intracellular cargo delivery. For example, RLW(RLWMRWYSPR TRAYG) linked nanoparticles (RNPs) showed a significantly en-hanced delivery in A549 lung cancer xenografts [71]. VG-21(VTPHHVLDEYTGEWVDSQFK), a CPP derived from vesicu-lar stomatitis virus glycoprotein(G), was conjugated to gold nanoparticles to enhance the cellular internalization, being suitable for various biomedical applications [80]. CPPs can efficiently de-liver hydrophilic and macromolecular cargos inside cells [81]. Another method is to induce cell membrane fusion with fu-sogenic NDDSs, which contain fusogenic molecules on the surface [82]. For instance, dioleoyl phosphatidylethanolamine (DOPE) is widely used as a fusogenic lipid to prepare fusogenic liposomes,

Current Pharmaceutical Design, 2015, Vol. 21, No. 42 6239

which usually showed increased cell internalization [83]. Besides, fusogenic liposomes can also be prepared by mixing the lipid com-ponents and complexes which generally have a net positive charge to facilitates their interaction with the negatively charged cellular membrane [84]. The main components of liposomes, analogous series of cationic lipids such as DODAP(1,2-dioleoyl-3-dimethyl-aminopropane), DSDMA(1,2-distearyloxy-N,N-dimethyl-3-amino-propane), DODMA(1,2-dioleyloxy-NN,-dimethyl-3-aminopro-pane), DLinDMA(1,2-dilinoleyloxy-NN,-dimethyl-3-aminopro-pane), DLenDMA(1,2-dilinolenyloxy-NN,-dimethyl-3-aminopro-pane), would affect the fusogenicity and efficiency of cell internali-zation [85]. Besides, some fusogenic peptides such as HA2 and

KALA functionalized nanoparticles for gene transduction were proved to be able to effectively enhance cellular uptake and bio-logical effects [86, 87]. Therefore, fusogenic NDDSs is an effective strategy of overcoming the tumor cell internalizing barrier.

However, a significant drawback of CPPs and fusogenic vesi-cles is their poor selectivity, which hampered their applications in delivering drugs into specific tissues systemically [71]. Thus, the active targeting molecules with cell selectivity like monoclonal antibody, peptide and aptamer may be better choices for tumor-targeted NDDSs [76, 88]. For example, since iron can effectively cross the blood–brain barrier by transferrin receptors, a transferrin-bearing generation 3-polypropylenimine dendrimer allowed the transport of plasmid DNA to the brain at least 3-fold higher than the unmodified dendrimers [73]. The folate receptor (FR), which is absent in most normal tissues but at a high level in human malig-nancies, is an attractive tumor-selective receptor and has been used on folate-derivatized adenoviruses, cationic polymers, liposomes, etc. [89, 90]. Peptide such as angiopep-2, a ligand of LRP receptor, was also used to modify nanoparticles for successfully treatment of glioma [89]. Through specific binding of receptors on tumor tis-sues, active targeting could effectively enhance drug internalization and limit the adverse side effects of tumor chemotherapy on healthy organs [91, 92].

As mentioned above, P-glycoprotein (P-gp) is a highly ex-pressed multidrug transporter which is often responsible for multidrug resistance (MDR), causing the failure of many chemical treatments against tumor [17]. Recently, some excipients which have a modulating effect on P-gp have been used on NDDSs [93]. For example, an P-gp inhibitor TPGS(d-??-tocopheryl polyethylene glycol 1000 succinate) loaded docetaxel micelles exhibited signifi-cantly higher antitumor activity on resistant KBv tumors [70]. Be-sides, both chitosan and anti-P-gp antibody conjugated PLGA nanoparticles showed increased cell internalization compared with chitosan conjugated PLGA nanoparticles [93, 94]. Owing to P-gp modulation from the inhibitors, P-gp targeting NDDSs provide an effective method for overcoming drug resistance and increasing drug internalization.

Endosomal Escape

Endolysosomal confinement is a major barrier for efficient transport of NDDSs [95]. It is necessary to perform further func-tionalization of NDDSs to enhance their escape ability from the endosome and avoid possible degradation of drugs in the endosome and lysosome. Currently, nanoparticles can escape from the en-dosome by two different mechanisms as follows: Fusion with Endosomal Membrane

Membrane fusion plays an important role not only in endocyto-sis but also in endosomal escape. Endosomal escape can be caused by the destabilization of the endosomal membrane through a series of fusogenic NDDSs [96, 97]. For example, fusogenic lipid DOPE is the most commonly used material in the preparation of NDDSs for lipid membrane fusion, because it could rapidly fuse with en-dosomal membrane based on its phase transition behavior [85]. DOPE-containing fusogenic liposomes or DOPE/PLGA nanoparti-cles have been formulated and proved to be able to facilitate

6240 Current Pharmaceutical Design, 2015, Vol. 21, No. 42 Pi et al.

Table 1. The strategies of overcoming the transport barriers of tumor-targeted NDDSs.

Physiological basis 1) 2)

The mononuclear

phagocyte system (MPS) Electrostatic interaction

1) 2)

Strategies Hydrophilic polymer nanoparticles

RBC cloak nanoparticles

2) 1)

Examples

PEG-MNPs, PEG modified nanoparticles, HA, Pluronic and polyglycerol copolymer nanoparticles

RBC encapsulating PLGA nanoparticles Smaller and compacted scale nanoparticles MSV conjugated with anti-VEGFR2 antibodies

NGR-functionalized nanoparticles Chitosan-coated liposome containing siVEGF, chemically modified heparin LHT7 nanoparticles, cRGD-linked polymeric micelles, chitosan-graft-PEI-candesartan conjugate

Complex nanoparticles(ComboNP), MMP-2 cleavable peptide inserted nanoparticles, functionalizing with hyaluronidase agent nanoparticles

Neuropilin-1-targeted liposomes, TPP functionalized PEG-PLA nanoparticles, angiopep-2 decorated gold nanoparticles, TPP fused EGFR single-domain antibody P-gp inhibitor loading micelles, anti-P-gp antibody PLGA nanoparticles, chitosan-modified or anti-P-glycoproteins conjugated PLGA nanoparticles

TAT peptide or R8 nanoparticles, RLW or VG-21 conjugated to gold nanoparticles Fusogenic liposome, HA2 and KALA functionalized gene nanoparticles Transferrin , folate or LRP receptor mediated nanoparticles

DOPE-containing fusogenic liposomes or DOPE/PLGA nanoparticles, fusogenic peptides(HA2 ,diINF-7, KALA and GALA) complexes

Polyethylenimine (PEI)-coated

mesoporous silica nanoparticles of siRNA and PAMAM dendrimer–siRNA

complexation, PAAs and PPAA as non-viral vectors

Chitosan enclosed mesoporous silica

nanoparticles(MSN), P(St-co-DMAEMA) complexes, MPEG-b-PMaIPG

nanoparticles, PELG/PEI/CAD complexes Gelatin-DOX conjugates, cationic gelatin combined polyGCeDOX, cathepsin B and cathepsin D responsive drug delivery vehicles

Delivery barriers 1)

Blood circulation

2)

Drug accumulations in tumor tissues

1) 2) 3) 4)

ECM

Vascular endothelial barrier Thick stroma

Proteolytic enzymes in the tumor

1) 2) 3)

EPR effect

Vascular targeting for accumulation

Antivascular targeting by inhibiting tumor angiogenesis

1) 2) 3) 4)

3)

Tumor tissue penetration

1) 2) 3) 4)

Low pH Tissue hypoxia EPR effect

High interstitial fluid pressure(HA, TAF, smooth muscle actin, collagen fibers)

Electric interaction need to stride over the cell

membrane to enter inside the cells

1)

Breaking up components of tumor stroma (TAFs, HA, etc.)

TPP functionalized nanoparticles

1)

2)

2)

4)

Drugs internalization into the targeted cells

1) 1) 2)

P-gp inhibitors combining nanoparticles

Tumor homing cell penetrating peptide

decorated nanomedicines Encapsulated cargo

internalized by fusogenic vesicles

Receptor-targeted nanocarriers

Fusion in the endosomal membrane

The proton sponge effect

1)

2) 3) 4)

3)

4)

5) Endosomal escape 1) 2)

Cellular uptake Endosomal escape

1) 2)

1)

2)

6)

Drug release 1)

Acidic environment and specific enzymes present in CSCs in the tumor

1) 2)

PH-sensitive nanoparticles Enzymes sensitive nanparticles

1)

2)

Overcoming Physiological Barriers of Tumor-targeted Nanomedicines Current Pharmaceutical Design, 2015, Vol. 21, No. 42 6241

(Table 1) Contd….

Delivery barriers 7)

Drug response

1)

Physiological basis Based on summarized pathophysiological basis

1) 2) 3)

Strategies Directly target and kill CSCs

Synergistic combination of two or more drugs Multi-functional targeted delivery

3) 1)

Examples

NPDAC combined with NPDOX, SAL-SWNTCHI-HA complexes, phenformin-loaded polymeric micelles

Combination of different chemotherapy and combination of chemotherapy and gene therapy

Liposome system functionalized with PEG, RGD and TAT, DGlueNP/PTX nanoparticles, octa-functional PLGA nanoparticles

2)

endosomal escape and enhance the biological response of various drugs [82, 98-101]. Besides, based on the fusion process occurring between the viral envelope and the endosomal membrane of host cell, some influenza-derived fusogenic peptides such as HA2 and diINF-7 have been used to increase the endosomal escape of siRNA and further the gene silencing efficiency [84]. Other synthetic fu-sogenic peptides such as KALA and GALA, capable of forming ??-helical structure for destabilization of endosomal membrane, have also been used to increase the endosomal escape of NDDSs [102, 103].

The Proton Sponge Effect

Unlike fusion systems that relied on the fusogenic property of the lipid or peptide to mediate endosomal escape, some nanocarri-ers, usually composed of cationic polymers and drugs, are supposed to use the so-called “proton sponge” effect to enhance the endoso-mal escape. Proton sponge effect is mainly caused by cationic polymers coated on the NDDSs that promote endosome osmotic swelling, rupture of the endosome membrane and intracellular re-lease of loaded drugs. Based on the mechanism, the cationic poly-mers with ‘proton-sponge’ nature such as polyethylenimine(PEI) and Polyamidoamine(PAMAM) dendrimers have been extensively studied as plasmid DNA (pDNA) delivery vehicles [104]. Cationic polymers containing NDDSs, such as PEI-coated and siRNA-loaded mesoporous silica nanoparticles and PAMAM dendrimer-siRNA complexation, were proved to possess the capability of initi-ating effectively endosomal escape before the degradation of the packaged siRNA in endolysosomes [95, 105]. Other chemical agents such as poly(amido amine)s (PAAs) and poly(propylacrylic acid) (PPAA) also have the function of endosomal escape and have been proved to improve the transfection efficiency of genetic drugs [97, 106]. Therefore, “proton sponge” agents are a promising can-didate for cytosolic drug delivery.

Drugs Release

As the last line of defense, an optimal drug release profile can be realized through biological stimuli-responsive NDDSs based on the acidic environment and specific enzymes present in tumor tissue or endolysosomes, thereby enhancing the killing effect on tumor cells [107].

pH-sensitive materials are more and more used to promote drug release in tumor stroma or tumor cells based on the low pH in tu-mor tissues(~6.5) or endolysosomes (4.5-6.5) compared with nor-mal tissues (pH7.4) [108, 109]. Thus various pH-responsive nano-carriers such as liposomes, nanoparticles, nanogels, polymer-drug conjugates and micelles have been extensively designed and re-ported [110]. As the conformational change can be exploited to trigger the drug release, a pH-sensitive charge-conversion system including micelles and polycarboxylates nanospheres was recently designed and the drug could be released in a pH-dependent manner

[109, 110]. Beside, both inorganic nanoscale materials like chitosan enclosed mesoporous silica nanoparticles and hybrid organic nano-materials such as P(St-co-DMAEMA) complexes(poly(styrene-co-N,N'-dimethylaminoethyl methacrylate) nanoparticles) and MPEG-b-PMaIPG(methoxy-polyethylene glycols (PEG)-b-poly (d-galactopyranose)nanoparticles have been proved to show a sensitive response to narrow pH changes and a good drug release behavior, exhibiting a high antitumor activity [109, 111-113].

Enzyme-responsive NDDSs were another effective system de-veloped to promote the drug release based on the specific enzymes present in tumor. For example, gelatin can be easily hydrolyzed into its sub-compounds by gelatinase, which is an endogenous prote-olytic enzyme and usually over-expressed in tumor tissues but not in normal tissues [114]. Gelatin-DOX (gelatin-doxorubicin) conju-gates or cationic gelatin/polyGC-DOX complexes can be specifi-cally digested when exposed to gelatinase and release doxorubicin [114, 115]. Some lysosomal cysteine proteases such as cathepsin B and cathepsin D, which play important roles in tumor progression and metastasis, also provided the possibility of designing enzyme-responsive drug delivery vehicles [116, 117]. Appropriate substrate which effectively degraded upon exposure to these cysteine prote-ases had been used as the linker of bioconjugates of antitumor drugs and polymers, permitting intra-lysosomal drug release after endocy-tosis [116, 117]. Nowadays, some esterases are found to be highly expressed in the intracellular compartments, and their substrates have been used as responsive self-immolative linkers in polymeric drug conjugates to trigger the drug release once taken up by tumor cells. For example, cholesterol esterase is highly expressed in en-dosome and lysosome, and the poly(ethylene carbonate), which can be degraded when exposed to cholesterol esterase, enabled release of bovine serum albumin (BSA) in an enzyme-responsive manner [118, 119].

Drug Response

Synergistic Antitumor Effect of Two or More Drugs

The use of single chemotherapeutic drug has shown limitations in anti-tumor treatment, such as development of drug resistance, high toxicity and low therapeutic index [120]. Thus combined ad-ministration of different drugs is emerging as a promising approach to achieve synergistic therapeutic efficacy [121]. Combinations of drugs with different mechanisms or targeting sites have been ex-plored to exploit either additive or synergistic effects. Some exam-ples are combination of different chemotherapy, and combination of chemotherapy and gene therapy [122]. These drugs can be simulta-neously loaded into nanocarriers such as functional polymeric mixed micelles, polyamidoamine dendrimer and copolymers, which have been proved to be helpful inhibiting tumor growth compared to single drug therapy [121, 123-125].

6242 Current Pharmaceutical Design, 2015, Vol. 21, No. 42 Antivascular Treatment

Inhibition of tumor angiogenesis or vascular system has also been proved to be a promising strategy for tumor therapy. Many reports have proved that the elimination of endothelial cells can remarkably inhibit tumor cell growth [38]. Some gene nanomedici-nes such as chitosan-coated liposome containing siVEGF and che-mically modified heparin LHT7 nanoparticles, have been demon-strated to significantly inhibit tumor angiogenesis, and further the tumor growth [126, 127]. Doxorubicin loaded TPP(RGERPPR) modified liposomes were able to destroy the tumor vessels while killing the tumor cells, which would signi??cantly amplify the anti-tumor effect [66]. Furthermore, the destruction of tumor vessels is considered to improve the inhibition of not only primary tumors but also metastatic solid tumors. This is a great advantage since hema-togenous metastasis is the most important way for tumor metastasis [38].

CSC Targeted System

Conventional tumor chemotherapy often fails as most anti-tumor drugs are not effective against drug-resistant CSCs [128]. As we mentioned above, although accounting for only a small part of the bulk tumors, CSCs have the ability of self-renewing, proliferat-ing, differentiating and producing new tumor bulks [129, 130]. Specific markers on CSCs could be used as the targets for CSCs targeted NDDSs [129, 131]. In general, combination of traditional chemotherapy with CSC targeted therapy could produce a synergis-tic antitumor effect [129, 132]. For example, best inhibitory effect for breast cancer was achieved by combining low-dose DAC (decit-abine) loaded nanoparticles and doxorubicin loaded nanoparticles with the lowest proportion of ALDHhi CSCs and the highest pro-portion of apoptotic tumor cells [133]. Due to the innate drug resis-tance of CSCs, effective killing of CSCs has been believed to be a key factor in eradicating tumor by NDDSs [134]. Multi-Functional Targeting Delivery System

At present, the most promising application of NDDSs is the multifunctional targeting delivery systems, which are formed via various modifications in order to overcome several barriers at the same time. For example, a multistage liposome system functional-ized with PEG, RGD and TAT has been developed and demon-strated its synergistic antitumor effect from the following three aspects: 1) increase blood circulation from PEG, 2) active malig-nant tumor targeting from RGD for integrin binding, and 3) effi-cient internalization from TAT peptide [76]. Based on the facilita-tive glucose transporter (GLUT) over-expression on both blood-brain barrier (BBB) and glioma cells, 2-deoxy-D-glucose modi??ed poly(ethylene glycol)-co-poly(trimethylene carbonate) nanoparti-cles (DGlueNP) were recently developed as a dual-targeted drug delivery system for enhancing the BBB penetration and improving the drug accumulation in the glioma via GLUT-mediated endocyto-sis [135, 136].

In order to enhance the antitumor effect of NDDSs, combined administration, anti-vascular treatment, CSCs targeted delivery system, and multi-functional targeting delivery system are more and more used for achieving additive or synergistic effects, representing a promising direction of tumor targeted therapy.

CONCLUSION AND FUTURE PERSPECTIVES

In this review, we divided the in vivo journey of NDDSs for tumor therapy into seven steps. In each step, we analyzed the trans-port barriers and reviewed the recently reported corresponding strategies of overcoming these barriers. It is essential for NDDSs to go through all the barriers to achieve a successful and effective anti-tumor therapy. A problem in any step will lead to the failure of the tumor treatment, which is in accordance with the \effect. Therefore, NDDSs should be rationally designed based on their whole systemic process in order to break up the seven barriers as we mentioned above. However, we should also keep in mind that

Pi et al.

the multi-functionalization is guaranteed to increase the complexity and decrease the druggability of NDDSs. There is still a long way to go to effectively kill tumors using rationally designed NDDSs. It requires the joint efforts from researchers of oncology, molecular biology, materials science, pharmaceutics and other related disci-plines.

CONFLICT OF INTEREST

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

ACKNOWLEDGEMENTS

This work was supported by National Basic Research Program of China (2013CB932500), National Natural Science Foundation of China (81202471, 51203024), Shanghai Innovation Fund for Small Technology based Firms (1402H234200), Natural Science Founda-tion of Shanghai (15ZR1430200) and the Zhejiang Provincial Natu-ral Science Foundation of China (Y14H300005). REFERENCES

[1]

Wu J-L, Wang C-Q, Zhuo R-X, Cheng S-X. Multi-drug delivery system based on alginate/calcium carbonate hybrid nanoparticles for combination chemotherapy. Colloids Surfaces B: Biointerfaces 2014; 123: 498-505.

[2] Wang J, Lei Y, Xie C, et al. Retro-Inverso CendR Peptide-Mediated Polyethyleneimine for Intracranial Glioblastoma-Targeting Gene Therapy. Bioconjug Chem 2014; 25: 414-423.

[3] Chen W, Gu B, Wang H, Pan J, Lu W, Hou H. Development and evaluation of novel itraconazole-loaded intravenous nanoparticles. Int J Pharm 2008; 362: 133-40.

[4] Daniels TR, Bernabeu E, Rodriguez JA, et al. The transferrin re-ceptor and the targeted delivery of therapeutic agents against can-cer. Biochim Biophys Acta 2012; 1820: 291-317.

[5] Nichols JW, Bae YH. EPR: Evidence and fallacy. J Control Re-lease 2014; 190: 451-64.

[6] Fokong S, Theek B, Wu Z, et al. Image-guided, targeted and trig-gered drug delivery to tumors using polymer-based microbubbles. J Control Release 2012; 163: 75-81.

[7] Bhattacharya C, Yu Z, Rishel MJ, Hecht SM. The carbamoylman-nose moiety of bleomycin mediates selective tumor cell targeting. Biochemistry 2014; 53: 3264-6.

[8]

Wen Y, Roudebush SL, Buckholtz GA, et al. Coassembly of am-phiphilic peptide EAK16-II with histidinylated analogues and im-plications for functionalization of beta-sheet fibrils in vivo. Bioma-terials 2014; 35: 5196-205.

[9]

Yan Z, Zhan C, Wen Z, et al. LyP-1-conjugated doxorubicin-loaded liposomes suppress lymphatic metastasis by inhibiting lymph node metastases and destroying tumor lymphatics. Nanotechnology 2011; 22: 415103.

[10] Yan Z, Wang F, Wen Z, et al. LyP-1-conjugated PEGylated liposomes: a carrier system for targeted therapy of lymphatic me-tastatic tumor. J Control Release 2012; 157: 118-25.

[11] Wen Y, Kolonich HR, Kruszewski KM, Giannoukakis N, Gawalt ES, Meng WS. Retaining antibodies in tumors with a self-assembling injectable system. Mol Pharm 2013; 10: 1035-44.

[12] Yu Z, Schmaltz RM, Bozeman TC, et al. Selective tumor cell tar-geting by the disaccharide moiety of bleomycin. J Am Chem Soc 2013; 135: 2883-6.

[13] Salo T, Vered M, Bello IO, et al. Insights into the role of compo-nents of the tumor microenvironment in oral carcinoma call for new therapeutic approaches. Exp Cell Res 2014; 325: 58-64.

[14] Ruan S, Yuan M, Zhang L, et al. Tumor microenvironment sensi-tive doxorubicin delivery and release to glioma using angiopep-2 decorated gold nanoparticles. Biomaterials 2015; 37: 425-35.

[15]

Zhao Y, Ji T, Wang H, Li S, Zhao Y, Nie G. Self-assembled pep-tide nanoparticles as tumor microenvironment activatable probes for tumor targeting and imaging. J Control Release 2014; 177: 11-9.

[16]

Huo M, Zou A, Yao C, et al. Somatostatin receptor-mediated tu-mor-targeting drug delivery using octreotide-PEG-deoxycholic acid conjugate-modified N-deoxycholic acid-O, N-hydroxyethylation chitosan micelles. Biomaterials 2012; 33: 6393-407.

Overcoming Physiological Barriers of Tumor-targeted Nanomedicines [17]

Nieto Montesinos R, Beduneau A, Pellequer Y, Lamprecht A. Delivery of P-glycoprotein substrates using chemosensitizers and nanotechnology for selective and efficient therapeutic outcomes. J Control Release 2012; 161: 50-61.

[18] Angarita FA, Acuna SA, Ottolino-Perry K, Zerhouni S, McCart JA. Mounting a strategic offense: fighting tumor vasculature with onco-lytic viruses. Trends Mol Med 2013; 19: 378-92.

[19] Lanitis E, Irving M, Coukos G. Targeting the tumor vasculature to enhance T cell activity. Curr Opin Immunol 2015; 33: 55-63.

[20]

Kanapathipillai M, Brock A, Ingber DE. Nanoparticle targeting of anti-cancer drugs that alter intracellular signaling or influence the tumor microenvironment. Adv Drug Deliv Rev 2014; 79-80: 107-118.

[21] Maeda H. Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity. Adv Drug Deliv Rev 2015; 91: 3-6.

[22] Cassereau L, Miroshnikova YA, Ou G, Lakins J, Weaver VM. A 3D tension bioreactor platform to study the interplay between ECM stiffness and tumor phenotype. J Biotechnol 2015; 193: 66-9.

[23] Kakizaki I, Koizumi H, Chen F, Endo M. Inhibitory effect of chon-droitin sulfate oligosaccharides on bovine testicular hyaluronidase. Carbohydr Polym 2015; 121: 362-71.

[24]

Gao W, Xiang B, Meng TT, Liu F, Qi XR. Chemotherapeutic drug delivery to cancer cells using a combination of folate targeting and tumor microenvironment-sensitive polypeptides. Biomaterials 2013; 34: 4137-49.

[25] Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regu-lators of the tumor microenvironment. Cell 2010; 141: 52-67.

[26] Ji RC. Hypoxia and lymphangiogenesis in tumor microenvironment and metastasis. Cancer Lett 2014; 346: 6-16.

[27] Shi NQ, Gao W, Xiang B, Qi XR. Enhancing cellular uptake of activable cell-penetrating peptide-doxorubicin conjugate by enzy-matic cleavage. Int J Nanomed 2012; 7: 1613-21.

[28] Das V, Stepankova J, Hajduch M, Miller JH. Role of tumor hy-poxia in acquisition of resistance to microtubule-stabilizing drugs. Biochim Biophys Acta 2015; 1855: 172-82.

[29] Ian F. Tannock, Daniela Rotin. Acid pH in tumors and its potential for therapeutic exploitation. Cancer Res ; 49: 4173-384.

[30] Seeber LM, Horree N, Vooijs MA, et al. The role of hypoxia in-ducible factor-1alpha in gynecological cancer. Crit Rev Oncol He-matol 2011; 78: 173-84.

[31] Binello E, Germano IM. Targeting glioma stem cells: a novel framework for brain tumors. Cancer Sci 2011; 102: 1958-66.

[32] Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer 2005; 5: 275-84.

[33] Walkey CD, Chan WC. Understanding and controlling the interac-tion of nanomaterials with proteins in a physiological environment. Chem Soc Rev 2012; 41: 2780-99.

[34]

C. Allen NDS, R. Gallagher GNC, Chiu Y, Shu WM, Li S, Johnstone A. Controlling the Physical Behavior and Biological Per-formance of Liposome Formulations through Use of Surface Grafted Poly(ethylene Glycol). Biosci Reports 2002; 22: 225-50. [35] Liu Y, Hu Y, Huang L. Influence of polyethylene glycol density and surface lipid on pharmacokinetics and biodistribution of lipid-calcium-phosphate nanoparticles. Biomaterials 2014; 35: 3027-34. [36]

Cole AJ, David AE, Wang J, Galban CJ, Yang VC. Magnetic brain tumor targeting and biodistribution of long-circulating PEG-modified, cross-linked starch-coated iron oxide nanoparticles. Biomaterials 2011; 32: 6291-301.

[37] Gu B, Xie C, Zhu J, He W, Lu W. Folate-PEG-CKK(2)-DTPA, a potential carrier for lymph-metastasized tumor targeting. Pharm Res 2010; 27: 933-42.

[38] Lollo G, Rivera-Rodriguez GR, Bejaud J, et al. Polyglutamic acid-PEG nanocapsules as long circulating carriers for the delivery of docetaxel. Eur J Pharm Biopharm 2014; 87: 47-54.

[39] Parveen S, Sahoo SK. Long circulating chitosan/PEG blended PLGA nanoparticle for tumor drug delivery. Eur J Pharmacol 2011; 670: 372-83.

[40] Maeda N, Takeuchi Y, Takada M, Sadzuka Y, Namba Y, Oku N. Anti-neovascular therapy by use of tumor neovasculature-targeted long-circulating liposome. J Control Release 2004; 100: 41-52. [41]

Yuji Yamamoto YN, Yukio Kato, Yuichi Sugiyama. Long-circulating poly(ethylene glycol)–poly(D,L-lactide) block copoly-mer micelles with modulated surface charge. J Control Release 2001: 27-38.

Current Pharmaceutical Design, 2015, Vol. 21, No. 42 6243 [42]

Sunoqrot S, Bugno J, Lantvit D, Burdette JE, Hong S. Prolonged blood circulation and enhanced tumor accumulation of folate-targeted dendrimer-polymer hybrid nanoparticles. J Control Re-lease 2014; 191: 115-22.

[43] Yang Y, Zhang YM, Chen Y, Chen JT, Liu Y. Targeted polysac-charide nanoparticle for adamplatin prodrug delivery. J Med Chem 2013; 56: 9725-36.

[44] Wang Y, Liu E, Sun X, et al. Pluronic L61 as a long-circulating modifier for enhanced liposomal delivery of cancer drugs. Polym Chem 2013; 4: 2958.

[45] Abu Lila AS, Kiwada H, Ishida T. The accelerated blood clearance (ABC) phenomenon: clinical challenge and approaches to manage. J Control Release 2013; 172: 38-47.

[46]

Abu Lila AS, Nawata K, Shimizu T, Ishida T, Kiwada H. Use of polyglycerol (PG), instead of polyethylene glycol (PEG), prevents induction of the accelerated blood clearance phenomenon against long-circulating liposomes upon repeated administration. Int J Pharm 2013; 456: 235-42.

[47] Hu CM, Fang RH, Luk BT, et al. 'Marker-of-self' functionalization of nanoscale particles through a top-down cellular membrane coat-ing approach. Nanoscale 2013; 5: 2664-8.

[48]

Hu C-MJ, Zhang L, Aryal S, Cheung C, Fang RH, Zhang L. Eryth-rocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc Natl Acad Sci USA 2011; 108: 10980-5.

[49] Weiwei Gao RHF, Soracha Thamphiwatana, Brian T Luk, et al. Modulating antibacterial immunity via bacterial membrane-coated nanoparticles. Letter 2015; 15: 1403-9.

[50] Aryal S, Hu C-MJ, Fang RH, et al. Erythrocyte membrane-cloaked polymeric nanoparticles for controlled drug loading and release. Nanomedicine 2013; 8: 1271-1280.

[51] Fang RH, Hu CM, Chen KN, et al. Lipid-insertion enables target-ing functionalization of erythrocyte membrane-cloaked nanoparti-cles. Nanoscale 2013; 5: 8884-8.

[52]

Martinez JO, Evangelopoulos M, Karun V, et al. The effect of multistage nanovector targeting of VEGFR2 positive tumor endo-thelia on cell adhesion and local payload accumulation. Biomateri-als 2014; 35: 9824-32.

[53]

Manzoor AA, Lindner LH, Landon CD, et al. Overcoming limita-tions in nanoparticle drug delivery: triggered, intravascular release to improve drug penetration into tumors. Cancer Res 2012; 72: 5566-75.

[54]

An S, Jiang X, Shi J, et al. Single-component self-assembled RNAi nanoparticles functionalized with tumor-targeting iNGR delivering abundant siRNA for efficient glioma therapy. Biomaterials 2015; 53: 330-40.

[55] Kang T, Gao X, Hu Q, et al. iNGR-modified PEG-PLGA nanopar-ticles that recognize tumor vasculature and penetrate gliomas. Biomaterials 2014; 35: 4319-32.

[56] Simnick AJ, Amiram M, Liu W, et al. In vivo tumor targeting by a NGR-decorated micelle of a recombinant diblock copolypeptide. J Control Release 2011; 155: 144-51.

[57]

Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 2012; 21: 418-29.

[58] Hambley TW, Hait WN. Is anticancer drug development heading in the right direction? Cancer Res 2009; 69: 1259-62.

[59]

Rajan M, Raj V, Al-Arfaj AA, Murugan AM. Hyaluronidase en-zyme core-5-fluorouracil-loaded chitosan-PEG-gelatin polymer nanocomposites as targeted and controlled drug delivery vehicles. Int J Pharm 2013; 453: 514-22.

[60] Buckway B, Wang Y, Ray A, Ghandehari H. Overcoming the stromal barrier for targeted delivery of HPMA copolymers to pan-creatic tumors. Int J Pharm 2013; 456: 202-11.

[61] Scodeller P. Hyaluronidase and other Extracellular Matrix Degrad-ing Enzymes for Cancer Therapy: New Uses and Nano-Formulations. J Carcinogenesis Mutagenesis 2014; 05.

[62] Luo G, Long J, Zhang B, et al. Stroma and pancreatic ductal ade-nocarcinoma: an interaction loop. Biochim Biophys Acta 2012; 1826: 170-8.

[63]

Zhang J, Miao L, Guo S, et al. Synergistic anti-tumor effects of combined gemcitabine and cisplatin nanoparticles in a stroma-rich bladder carcinoma model. J Control Release 2014; 182: 90-6.

6244 Current Pharmaceutical Design, 2015, Vol. 21, No. 42 [64] Teesalu T, Sugahara KN, Kotamraju VR, Ruoslahti E. C-end rule peptides mediate neuropilin-1-dependent cell, vascular, and tissue penetration. Proc Natl Acad Sci USA 2009; 106: 16157-62.

[65] Hu Q, Gao X, Gu G, et al. Glioma therapy using tumor homing and penetrating peptide-functionalized PEG-PLA nanoparticles loaded with paclitaxel. Biomaterials 2013; 34: 5640-50.

[66] Yang Y, Yan Z, Wei D, et al. Tumor-penetrating peptide function-alization enhances the anti-glioblastoma effect of doxorubicin liposomes. Nanotechnology 2013; 24: 405101.

[67] Ruoslahti E. Peptides as targeting elements and tissue penetration devices for nanoparticles. Adv Mater 2012; 24: 3747-56.

[68] Sugahara KN, Teesalu T, Karmali PP, et al. Tissue-penetrating delivery of compounds and nanoparticles into tumors. Cancer Cell 2009; 16: 510-20.

[69] Yan Z, Yang Y, Wei X, et al. Tumor-penetrating peptide media-tion: an effective strategy for improving the transport of liposomes in tumor tissue. Mol Pharm 2014; 11: 218-25.

[70]

Wang AT, Liang DS, Liu YJ, Qi XR. Roles of ligand and TPGS of micelles in regulating internalization, penetration and accumulation against sensitive or resistant tumor and therapy for multidrug resis-tant tumors. Biomaterials 2015; 53: 160-72.

[71] Gao H, Zhang Q, Yang Y, Jiang X, He Q. Tumor homing cell penetrating peptide decorated nanoparticles used for enhancing tu-mor targeting delivery and therapy. Int J Pharm 2014; 478: 240-50. [72] Mustata RC, Grigorescu A, Petrescu SM. Encapsulated cargo in-ternalized by fusogenic liposomes partially overlaps the endoplas-mic reticulum. J Cell Mol Med 2009; 13: 3110-21.

[73] Somani S, Blatchford DR, Millington O, Stevenson ML, Dufes C. Transferrin-bearing polypropylenimine dendrimer for targeted gene delivery to the brain. J Control Release 2014; 188: 78-86.

[74] Milletti F. Cell-penetrating peptides: classes, origin, and current landscape. Drug Discov Today 2012; 17: 850-60.

[75] Gao H, Zhang Q, Yu Z, He Q. Cell-penetrating peptide-based intel-ligent liposomal systems for enhanced drug delivery. Curr Pharm Biotechnol 2014; 15: 210-9.

[76] Mei L, Fu L, Shi K, et al. Increased tumor targeted delivery using a multistage liposome system functionalized with RGD, TAT and cleavable PEG. Int J Pharm 2014; 468: 26-38.

[77]

Zhang Y, Kollmer M, Buhrman JS, Tang MY, Gemeinhart RA. Arginine-rich, cell penetrating peptide-anti-microRNA complexes decrease glioblastoma migration potential. Peptides 2014; 58: 83-90.

[78] Khalil IA, Kogure K, Futaki S, Harashima H. Octaarginine-modified liposomes: enhanced cellular uptake and controlled intra-cellular trafficking. Int J Pharm 2008; 354: 39-48.

[79]

Kanazawa T, Akiyama F, Kakizaki S, Takashima Y, Seta Y. Deliv-ery of siRNA to the brain using a combination of nose-to-brain de-livery and cell-penetrating peptide-modified nano-micelles. Bioma-terials 2013; 34: 9220-6.

[80]

Tiwari PM, Eroglu E, Bawage SS, et al. Enhanced intracellular translocation and biodistribution of gold nanoparticles functional-ized with a cell-penetrating peptide (VG-21) from vesicular stoma-titis virus. Biomaterials 2014; 35: 9484-94.

[81] Jobin ML, Alves ID. On the importance of electrostatic interactions between cell penetrating peptides and membranes: a pathway to-ward tumor cell selectivity? Biochimie 2014; 107 Pt A: 154-9.

[82] Naumovska E, Ludwanowski S, Hersch N, et al. Plasma membrane functionalization using highly fusogenic immune activator liposomes. Acta Biomater 2014; 10: 1403-11.

[83]

Csiszar A, Csiszar A, Pinto JT, et al. Resveratrol encapsulated in novel fusogenic liposomes activates Nrf2 and attenuates oxidative stress in cerebromicrovascular endothelial cells from aged rats. J Gerontol A Biol Sci Med Sci 2015; 70: 303-13.

[84] Oliveira S, van Rooy I, Kranenburg O, Storm G, Schiffelers RM. Fusogenic peptides enhance endosomal escape improving siRNA-induced silencing of oncogenes. Int J Pharm 2007; 331: 211-4. [85] Heyes J, Palmer L, Bremner K, MacLachlan I. Cationic lipid satu-ration influences intracellular delivery of encapsulated nucleic ac-ids. Journal of Controlled Release 2005; 107: 276-87.

[86]

Ye SF, Tian MM, Wang TX, et al. Synergistic effects of cell-penetrating peptide Tat and fusogenic peptide HA2-enhanced cellu-lar internalization and gene transduction of organosilica nanoparti-cles. Nanomedicine 2012; 8: 833-41.

[87]

Li X, Chen Y, Wang M, Ma Y, Xia W, Gu H. A mesoporous silica nanoparticle--PEI--fusogenic peptide system for siRNA delivery in cancer therapy. Biomaterials 2013; 34: 1391-401.

Pi et al.

[88]

Li J, Gu B, Meng Q, et al. The use of myristic acid as a ligand of polyethylenimine/DNA nanoparticles for targeted gene therapy of glioblastoma. Nanotechnology 2011; 22: 435101.

[89] Ren J, Shen S, Wang D, et al. The targeted delivery of anticancer

drugs to brain glioma by PEGylated oxidized multi-walled carbon nanotubes modified with angiopep-2. Biomaterials 2012; 33: 3324-33.

[90] Yoo HS, Park TG. Folate-receptor-targeted delivery of doxorubicin

nano-aggregates stabilized by doxorubicin-PEG-folate conjugate. J Control Release 2004; 100: 247-56.

[91] Dharap SS, Wang Y, Chandna P, et al. Tumor-specific targeting of

an anticancer drug delivery system by LHRH peptide. Proc Natl Acad Sci USA 2005; 102: 12962-7.

[92] Zhan C, Gu B, Xie C, Li J, Liu Y, Lu W. Cyclic RGD conjugated

poly(ethylene glycol)-co-poly(lactic acid) micelle enhances pacli-taxel anti-glioblastoma effect. J Control Release 2010; 143: 136-42.

[93] Miao Guo, Ying Wang, Wenting Rong, Liying Ding, Yu S. Chito-san-modified PLGA nanoparticles as drug delivery carrier for

modulating P-glycoprotein of enterocytes. J Control Release 2013; 172: e112-3.

[94] Iangcharoen P, Punfa W, Yodkeeree S, et al. Anti-P-glycoprotein

conjugated nanoparticles for targeting drug delivery in cancer treatment. Arch Pharm Res 2011; 34: 1679-89.

[95] Wang M, Li X, Ma Y, Gu H. Endosomal escape kinetics of

mesoporous silica-based system for efficient siRNA delivery. Int J Pharm 2013; 448: 51-7.

[96] Lee WS, Kim YK, Zhang Q, et al. Polyxylitol-based gene carrier

improves the efficiency of gene transfer through enhanced en-dosomal osmolysis. Nanomedicine 2014; 10: 525-34.

[97] Varkouhi AK, Scholte M, Storm G, Haisma HJ. Endosomal escape

pathways for delivery of biologicals. J Control Release 2011; 151: 220-8.

[98] Mansourian M, Badiee A, Jalali SA, et al. Effective induction of

anti-tumor immunity using p5 HER-2/neu derived peptide encapsu-lated in fusogenic DOTAP cationic liposomes co-administrated with CpG-ODN. Immunol Lett 2014; 162: 87-93.

[99] Qiang Hu, Cli?ord R Shew, Marcel B Bally, Madden TD. Pro-grammable fusogenic vesicles for intracellular delivery of antisense oligodeoxynucleotides: enhanced cellular uptake and biological e?ects. Biochim Biophys Acta 2001; 1514: 1-13.

[100] Debra T Auguste, Robert K Prud' homme, Patrick L Ahl, Paul

Meers, Kohn J. Association of hydrophobically-modified poly(ethylene glycol) with fusogenic liposomes. Biochim Biophys Acta 2003; 1616: 184-95.

[101] Chhabra R, Grabrucker AM, Veratti P, et al. Characterization of

lysosome-destabilizing DOPE/PLGA nanoparticles designed for cytoplasmic drug release. Int J Pharm 2014; 471: 349-57.

[102] Guo XD, Wiradharma N, Liu SQ, et al. Oligomerized alpha-helical

KALA peptides with pendant arms bearing cell-adhesion, DNA-binding and endosome-buffering domains as efficient gene trans-fection vectors. Biomaterials 2012; 33: 6284-91.

[103] Funhoff AM, van Nostrum CF, Lok MC, Kruijtzer JA, Crommelin

DJ, Hennink WE. Cationic polymethacrylates with covalently linked membrane destabilizing peptides as gene delivery vectors. J Control Release 2005; 101: 233-46.

[104] Lachelt U, Kos P, Mickler FM, et al. Fine-tuning of proton sponges

by precise diaminoethanes and histidines in pDNA polyplexes. Nanomedicine 2014; 10: 35-44.

[105] Ouyang D, Zhang H, Parekh HS, Smith SC. The effect of pH on

PAMAM dendrimer-siRNA complexation: endosomal considera-tions as determined by molecular dynamics simulation. Biophys Chem 2011; 158: 126-33.

[106] Lee LK, Williams CL, Devore D, Roth CM. Poly(propylacrylic

acid) enhances cationic lipid-mediated delivery of antisense oli-gonucleotides. Biomacromolecules 2006; 7: 1502-8.

[107] Dan Z, Cao H, He X, et al. Biological stimuli-responsive cyclodex-trin-based host-guest nanosystems for cancer therapy. Int J Pharm 2015; 483: 63-8.

[108] Ouahab A, Cheraga N, Onoja V, Shen Y, Tu J. Novel pH-sensitive

charge-reversal cell penetrating peptide conjugated PEG-PLA mi-celles for docetaxel delivery: in vitro study. Int J Pharm 2014; 466: 233-45.

[109] Guan X, Li Y, Jiao Z, et al. A pH-sensitive charge-conversion

system for doxorubicin delivery. Acta Biomater 2013; 9: 7672-8.

Overcoming Physiological Barriers of Tumor-targeted Nanomedicines [110] Felber AE, Dufresne MH, Leroux JC. pH-sensitive vesicles, po-lymeric micelles, and nanospheres prepared with polycarboxylates. Adv Drug Deliv Rev 2012; 64: 979-92.

[111] Chen F, Zhu Y. Chitosan enclosed mesoporous silica nanoparticles

as drug nano-carriers: Sensitive response to the narrow pH range. Microporous Mesoporous Mater 2012; 150: 83-9.

[112] Hu Y, Wang J, Zhang H, Jiang G, Kan C. Synthesis and characteri-zation of monodispersed P(St-co-DMAEMA) nanoparticles as pH-sensitive drug delivery system. Mater Sci Eng C Mater Biol Appl 2014; 45: 1-7.

[113] Liu J, Huang Y, Kumar A, et al. pH-sensitive nano-systems for

drug delivery in cancer therapy. Biotechnol Adv 2014; 32: 693-710.

[114] Xue P, Cheong KK, Wu Y, Kang Y. An in-vitro study of enzyme-responsive Prussian blue nanoparticles for combined tumor chemo-therapy and photothermal therapy. Colloids Surf B Biointerfaces 2015; 125: 277-83.

[115] Dong L, Xia S, Wu K, et al. A pH/enzyme-responsive tumor-specific delivery system for doxorubicin. Biomaterials 2010; 31: 6309-16.

[116] Li N, Li N, Yi Q, et al. Amphiphilic peptide dendritic copolymer-doxorubicin nanoscale conjugate self-assembled to enzyme-responsive anti-cancer agent. Biomaterials 2014; 35: 9529-45.

[117] Mortato M, Argentiere S, De Gregorio GL, Gigli G, Blasi L. En-zyme-responsive multifunctional surfaces for controlled up-take/release of (bio)molecules. Colloids Surf B Biointerfaces 2014; 123: 89-95.

[118] Namgung R, Mi Lee Y, Kim J, et al. Poly-cyclodextrin and poly-paclitaxel nano-assembly for anticancer therapy. Nat Commun 2014; 5: 3702.

[119] Chu D, Curdy C, Riebesehl B, Zhang Y, Beck-Broichsitter M,

Kissel T. Enzyme-responsive surface erosion of poly(ethylene car-bonate) for controlled drug release. Eur J Pharm Biopharm 2013; 85: 1232-7.

[120] Wang H, Zhao Y, Wu Y, et al. Enhanced anti-tumor efficacy by

co-delivery of doxorubicin and paclitaxel with amphiphilic methoxy PEG-PLGA copolymer nanoparticles. Biomaterials 2011; 32: 8281-90.

[121] Kang L, Gao Z, Huang W, Jin M, Wang Q. Nanocarrier-mediated

co-delivery of chemotherapeutic drugs and gene agents for cancer treatment. Acta Pharmaceutica Sinica B 2015; 5: 169-175.

[122] Han L, Huang R, Li J, Liu S, Huang S, Jiang C. Plasmid pORF-hTRAIL and doxorubicin co-delivery targeting to tumor using pep-tide-conjugated polyamidoamine dendrimer. Biomaterials 2011; 32: 1242-52.

[123] Zhu L, Perche F, Wang T, Torchilin VP. Matrix metalloproteinase

2-sensitive multifunctional polymeric micelles for tumor-specific co-delivery of siRNA and hydrophobic drugs. Biomaterials 2014; 35: 4213-22.

Received: August 14, 2015 Accepted: October 26, 2015

Current Pharmaceutical Design, 2015, Vol. 21, No. 42 6245

[124]

Miao L, Guo S, Zhang J, Kim WY, Huang L. Nanoparticles with precise ratiometric co-loading and co-delivery of gemcitabine mo-nophosphate and cisplatin for treatment of bladder cancer. Adv Funct Mater 2014; 24: 6601-11.

[125]

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: 44-58.

[126]

Salva E, Turan SO, Eren F, Akbuga J. The enhancement of gene silencing efficiency with chitosan-coated liposome formulations of siRNAs targeting HIF-1alpha and VEGF. Int J Pharm 2014; 478: 147-54.

[127]

Chung SW, Bae SM, Lee M, et al. LHT7, a chemically modified heparin, inhibits multiple stages of angiogenesis by blocking VEGF, FGF2 and PDGF-B signaling pathways. Biomaterials 2015; 37: 271-8.

[128]

Krishnamurthy S, Ng VW, Gao S, Tan MH, Yang YY. Phen-formin-loaded polymeric micelles for targeting both cancer cells and cancer stem cells in vitro and in vivo. Biomaterials 2014; 35: 9177-86.

[129] Han L, Shi S, Gong T, Zhang Z, Sun X. Cancer stem cells: thera-peutic implications and perspectives in cancer therapy. Acta Pharm Sin B 2013; 3: 65-75.

[130] Yi SY, Hao YB, Nan KJ, Fan TL. Cancer stem cells niche: a target for novel cancer therapeutics. Cancer Treat Rev 2013; 39: 290-6. [131] Klonisch T, Wiechec E, Hombach-Klonisch S, et al. Cancer stem cell markers in common cancers - therapeutic implications. Trends Mol Med 2008; 14: 450-60.

[132] Zhan HX, Xu JW, Wu D, Zhang TP, Hu SY. Pancreatic cancer stem cells: new insight into a stubborn disease. Cancer Lett 2015; 357: 429-37.

[133]

Li SY, Sun R, Wang HX, et al. Combination therapy with epige-netic-targeted and chemotherapeutic drugs delivered by nanoparti-cles to enhance the chemotherapy response and overcome resis-tance by breast cancer stem cells. J Control Release 2014; 205: 7-14.

[134]

Liu C, Zhao G, Liu J, et al. Novel biodegradable lipid nano com-plex for siRNA delivery significantly improving the chemosensitiv-ity of human colon cancer stem cells to paclitaxel. J Control Re-lease 2009; 140: 277-83.

[135]

Jiang X, Xin H, Ren Q, et al. Nanoparticles of 2-deoxy-D-glucose functionalized poly(ethylene glycol)-co-poly(trimethylene carbon-ate) for dual-targeted drug delivery in glioma treatment. Biomateri-als 2014; 35: 518-29.

[136]

Zhou J, Patel TR, Fu M, Bertram JP, Saltzman WM. Octa-functional PLGA nanoparticles for targeted and efficient siRNA delivery to tumors. Biomaterials 2012; 33: 583-91.

本文来源:https://www.bwwdw.com/article/ja07.html

Top