+ All Categories
Home > Documents > Anticancer nanomedicine and tumor vascular permeability; Where is the missing link?

Anticancer nanomedicine and tumor vascular permeability; Where is the missing link?

Date post: 28-Nov-2016
Category:
Upload: khaled
View: 217 times
Download: 2 times
Share this document with a friend
11
Anticancer nanomedicine and tumor vascular permeability; Where is the missing link? Sebastien Taurin a , Hayley Nehoff a , Khaled Greish a, b, a Department of Pharmacology & Toxicology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand b Department of Oncology, Faculty of Medicine, Suez Canal University, Egypt abstract article info Article history: Received 30 March 2012 Accepted 8 July 2012 Available online 16 July 2012 Keywords: Enhanced permeability and retention (EPR) Nanomedicine Drug targeting Vascular permeability Tumor vessel Pharmacokinetics Anticancer nanomedicine was coined to describe anticancer delivery systems such as polymer conjugates, li- posomes, micelles, and metal nanoparticles. These anticancer delivery platforms have been developed with the enhanced permeability and retention (EPR) effect as a central mechanism for tumor targeting. EPR based nanomedicine has demonstrated, beyond doubt, to selectively target tumor tissues in animal models. However, over the last two decades, only nine anticancer agents utilizing this targeting strategy have been approved for clinical use. In this review, we systematically analyze various aspects that explain the limited clinical progress yet achieved. The inuence of nanomedicine physicochemical characteristics, animal tumor models, and variations in tumor biology, on EPR based tumor targeting is closely examined. Further- more, we reviewed results from over one hundred publications to construct patterns of factors that can inu- ence the transition of EPR based anticancer nanomedicine to the clinic. © 2012 Elsevier B.V. All rights reserved. 1. Introduction The discovery of the EPR effect originates from pioneering work that started more than 100 years ago. In 1907, E. Goldman described tumor vasculature as The normal blood vessels of the organs in which the tumor is developing are disturbed by chaotic growth, there is a dilatation and spiraling of the affected vessels, marked cap- illary budding and new vessel formation, particularly at the advanced border[1]. The immunological hypothesis developed by Paul Ehrlich at the beginning of the 20th century which evolved to what became known as Ehrlich's magic bulletwherein a drug would specically target the organism responsible for a disease [2]. Later, Judah Folkman demonstrated the ability of tumors to stimulate their own vascularization [3]. Subsequent studies established that these tumor blood vessels harbored higher permeability compared to normal blood vessels [47]. These newly formed vessels usually have irregu- lar and incomplete structures in conjunction with modied physio- logical responses [8]. Further work led to the identication of the tumor vascular permeability factor (VPF) which was later found to be identical to vascular endothelial growth factor (VEGF), a major de- terminant for enhanced vessel permeability [9]. The growth factors of the VEGF family are endothelial mitogens as well as potent mediators of vascular permeability. Ultrastructural studies have provided information on the mechanisms by which endothelial cells respond to VEGF [10]. Through alternative splicing, tumors generally overexpress multiple isoforms of VEGF [11] and these are known to bind VEGFR-1/Flt, VEGFR-2/Flk and some to neuropilin. Each isoform can induce vascular permeability but as they differ in their ability to interact with matrix proteins, this results in structural aberrations of blood vessels (for review see [12]). Further research revealed some of the mechanisms by which VEGF inuences vascular permeability in pathological conditions (for review see [13]). Finally, the observa- tion that macromolecules and lipids selectively permeate the tumor vasculature and remain in the tumor interstitium for an extended pe- riod of time led to the characterization of the tumor-selective EPR ef- fect [14]. The EPR effect explains the distinctive increase in vascular permeability, both in tumor vasculature [15] and in inammatory tis- sues [16]. The discovery of this unique phenomenon in solid tumors is considered a landmark for anticancer nanomedicine development. In the early 1980s, Maeda invented the rst known anticancer nanomedicine, styrene co-maleic acid conjugated neoCarzinostatin (SMANCS) based on the EPR concept [17]. Following the pioneering work of Maeda, many investigators have developed EPR based anti- cancer nanosystems. The main advantage of EPR-based anticancer nanomedicines is their altered pharmacokinetics caused by their hy- drodynamic diameter that can reach 7 nm in size. A diameter superi- or to 7 nm will escape renal ltration and urinary excretion [18], due to the slit diaphragms at the level of the podocyte foot of the glomerula which prevent the ltration of globular plasma proteins above this size [19]. Therefore, high nanometer sized particles can ex- hibit prolonged circulatory half-life, high area under concentration/ time curve (AUC) and higher partition into tumor tissues [20]. Suc- cess of an effective drug delivery system is thus dependent on the size [21] as well as the shape [22] of the nanomedicine to evade Journal of Controlled Release 164 (2012) 265275 Corresponding author at: Adams Building, 18 Frederick Street, Level 2 Room 238, Dunedin, New Zealand. Tel.: +64 3 479 4095; fax: +64 3 479 9140. E-mail address: [email protected] (K. Greish). 0168-3659/$ see front matter © 2012 Elsevier B.V. All rights reserved. doi:10.1016/j.jconrel.2012.07.013 Contents lists available at SciVerse ScienceDirect Journal of Controlled Release journal homepage: www.elsevier.com/locate/jconrel
Transcript
Page 1: Anticancer nanomedicine and tumor vascular permeability; Where is the missing link?

Journal of Controlled Release 164 (2012) 265–275

Contents lists available at SciVerse ScienceDirect

Journal of Controlled Release

j ourna l homepage: www.e lsev ie r .com/ locate / jconre l

Anticancer nanomedicine and tumor vascular permeability; Where is themissing link?

Sebastien Taurin a, Hayley Nehoff a, Khaled Greish a,b,⁎a Department of Pharmacology & Toxicology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealandb Department of Oncology, Faculty of Medicine, Suez Canal University, Egypt

⁎ Corresponding author at: Adams Building, 18 FredeDunedin, New Zealand. Tel.: +64 3 479 4095; fax: +64

E-mail address: [email protected] (K. Greish

0168-3659/$ – see front matter © 2012 Elsevier B.V. Alldoi:10.1016/j.jconrel.2012.07.013

a b s t r a c t

a r t i c l e i n f o

Article history:Received 30 March 2012Accepted 8 July 2012Available online 16 July 2012

Keywords:Enhanced permeability and retention (EPR)NanomedicineDrug targetingVascular permeabilityTumor vesselPharmacokinetics

Anticancer nanomedicine was coined to describe anticancer delivery systems such as polymer conjugates, li-posomes, micelles, and metal nanoparticles. These anticancer delivery platforms have been developed withthe enhanced permeability and retention (EPR) effect as a central mechanism for tumor targeting. EPRbased nanomedicine has demonstrated, beyond doubt, to selectively target tumor tissues in animal models.However, over the last two decades, only nine anticancer agents utilizing this targeting strategy have beenapproved for clinical use. In this review, we systematically analyze various aspects that explain the limitedclinical progress yet achieved. The influence of nanomedicine physicochemical characteristics, animaltumor models, and variations in tumor biology, on EPR based tumor targeting is closely examined. Further-more, we reviewed results from over one hundred publications to construct patterns of factors that can influ-ence the transition of EPR based anticancer nanomedicine to the clinic.

© 2012 Elsevier B.V. All rights reserved.

1. Introduction

The discovery of the EPR effect originates from pioneering workthat started more than 100 years ago. In 1907, E. Goldman describedtumor vasculature as “The normal blood vessels of the organs inwhich the tumor is developing are disturbed by chaotic growth,there is a dilatation and spiraling of the affected vessels, marked cap-illary budding and new vessel formation, particularly at the advancedborder” [1]. The immunological hypothesis developed by Paul Ehrlichat the beginning of the 20th century which evolved to what becameknown as ‘Ehrlich's magic bullet’ wherein a drug would specificallytarget the organism responsible for a disease [2]. Later, JudahFolkman demonstrated the ability of tumors to stimulate their ownvascularization [3]. Subsequent studies established that these tumorblood vessels harbored higher permeability compared to normalblood vessels [4–7]. These newly formed vessels usually have irregu-lar and incomplete structures in conjunction with modified physio-logical responses [8]. Further work led to the identification of thetumor vascular permeability factor (VPF) which was later found tobe identical to vascular endothelial growth factor (VEGF), a major de-terminant for enhanced vessel permeability [9]. The growth factors ofthe VEGF family are endothelial mitogens as well as potent mediatorsof vascular permeability. Ultrastructural studies have providedinformation on the mechanisms by which endothelial cells respondto VEGF [10]. Through alternative splicing, tumors generally

rick Street, Level 2 Room 238,3 479 9140.).

rights reserved.

overexpress multiple isoforms of VEGF [11] and these are known tobind VEGFR-1/Flt, VEGFR-2/Flk and some to neuropilin. Each isoformcan induce vascular permeability but as they differ in their ability tointeract with matrix proteins, this results in structural aberrations ofblood vessels (for review see [12]). Further research revealed someof the mechanisms by which VEGF influences vascular permeabilityin pathological conditions (for review see [13]). Finally, the observa-tion that macromolecules and lipids selectively permeate the tumorvasculature and remain in the tumor interstitium for an extended pe-riod of time led to the characterization of the tumor-selective EPR ef-fect [14]. The EPR effect explains the distinctive increase in vascularpermeability, both in tumor vasculature [15] and in inflammatory tis-sues [16]. The discovery of this unique phenomenon in solid tumors isconsidered a landmark for anticancer nanomedicine development.In the early 1980s, Maeda invented the first known anticancernanomedicine, styrene co-maleic acid conjugated neoCarzinostatin(SMANCS) based on the EPR concept [17]. Following the pioneeringwork of Maeda, many investigators have developed EPR based anti-cancer nanosystems. The main advantage of EPR-based anticancernanomedicines is their altered pharmacokinetics caused by their hy-drodynamic diameter that can reach 7 nm in size. A diameter superi-or to 7 nm will escape renal filtration and urinary excretion [18], dueto the slit diaphragms at the level of the podocyte foot of theglomerula which prevent the filtration of globular plasma proteinsabove this size [19]. Therefore, high nanometer sized particles can ex-hibit prolonged circulatory half-life, high area under concentration/time curve (AUC) and higher partition into tumor tissues [20]. Suc-cess of an effective drug delivery system is thus dependent on thesize [21] as well as the shape [22] of the nanomedicine to evade

Page 2: Anticancer nanomedicine and tumor vascular permeability; Where is the missing link?

266 S. Taurin et al. / Journal of Controlled Release 164 (2012) 265–275

renal filtration and to limit clearance. Furthermore, nanodrug carrierscan be functionalized by a variety of moieties to prolong their circula-tory half-life in the blood beyond the advantage of size [23]. Aprolonged circulation will favor high plasma AUC compared to thefree drug [20,24,25] and significantly increase drug accumulation intumor tissue [21].

The Food and Drug Administration (FDA) and other internationalequivalents have approved over 30 nano-therapeutics for clinicaluse, of which 11 are used for the treatment or detection of various can-cers (Table 1). EPR effect has certainly influenced the design, develop-ment and improvement of different delivery platforms. Nevertheless,the rate of transition of EPR principles into the clinic has been up tonow disappointingly slow. In this article, we will discuss the currentstatus of EPR based drugs in the clinic; followed by a review of the cur-rent literature to identify factors that might adversely influence thedevelopment of EPR effect based anticancer nanomedicine.

2. EPR in the clinic

Proof of the EPR effect concept was clearly demonstrated byMaeda, 26 years ago, using Evans blue dye (EBD) injected intrave-nously into rodents. The dye was bound to albumin and selectivelyconcentrated in tumor tissue [15]. The EPR effect is a time dependentphenomenon. It usually takes more than 6 h of high plasma concen-tration of the drug to achieve accumulation in tumor tissue that lastfor an extended period of time ranging from hours to several days[38,39]. Maeda's work demonstrated that the rate of accumulationof macromolecules and lipids in tumor was inversely proportionalto their clearance rate. Based on these findings, several nano-sizedrug carriers were developed. SMANCS was the first clinical polymer-ic conjugate which was approved in Japan in 1993 for the treatmentof hepatocellular carcinoma (Table 1). Following this landmarkapplication of the EPR principle for tumor targeting, severalnanomedicines were developed for anticancer chemotherapy in thefollowing years. Liposomes have achieved significant success com-pared to other nanomedicines (see Table 1). For example, Doxilwhich is a PEGylated (polyethylene glycol coated) liposome that en-capsulates doxorubicin, was approved for the treatment of Kaposisarcoma and late stage ovarian cancer in 1995 [40]. Other polymericor micellar second or third generation drugs are currently beingassessed in clinical trials (phase I–III).

The size and shape of nanoparticles are critical for nanomedicineapplications. Extensive studies have shown that both criteria willdetermine their longevity in the blood circulation and theirbiodistribution when administrated to a patient. To be effective, in-travascularly administrated particles must avoid recognition by thereticulo-endothelial system (RES) composed of macrophages pres-ent in the liver, spleen, bone marrow and lymph nodes [23]. Gener-ally, particles larger than 100 nm are rapidly eliminated from the

Table 1Clinically approved nanomedicines for cancer treatment or imaging.

Type of nanoparticle Name Therapeutic agent

Liposomes Doxil/Caelyx Doxorubicin

DaunoXome DoxorubicinDepocyt CytarabineMyocet Doxorubicin

MEPACT Muramyl tripeptide phosphatidyl ethanolaminePolymeric micelles Oncaspar PEG-L-asparaginase

Genexol-PM PaclitaxelConjugate SMANCS NeocarzinostatinAlbumin Abraxane PaclitaxelMetal Feridex Superparamagnetic iron-oxide

GastroMARK Superparamagnetic iron-oxide

circulation by the RES [41,42]. To avoid this, several strategies havebeen developed, for instance, the addition of chemical modificationsby synthetic polymers such as polyethylene glycol (PEG) sterically hin-der interaction with plasma proteins [43] and reduced opsonization[44]. Gold nanorods have a prolonged circulation time and are lessphagocytosed compared to their spherical counterparts, howeverthe biodistribution of PEGylated gold nanorods and spheres ofequivalent sizes showed a similar accumulation profiles in the liverand spleen [22].

The advantages of nanomedicine over conventional medicinesrely essentially on the EPR effect and their improved pharmacokinet-ics, loading capacity, release rates, lower systemic toxicity and abilityto bypass multidrug resistance mechanisms that involve cell-surfaceprotein pumps.

3. The advantages and limitations of the EPR effect

Unlikemost normal tissues, tumor vasculature is heterogeneous anddoes not conform to standard morphology (i.e., arterioles, capillariesand venules which form a well-organized network with dichotomousbranching and hierarchic order) [45]. The tumor microenvironment iscomposed of a variety of cell types such as tumors cells, stromal cellsand inflammatory cells which all influence the angiogenic process [3].Newly formed blood vessels are dilated, saccular, poorly aligned andheterogeneous [46]. Tumor vasculature is characterized by defectiveendothelial cells with wide fenestration ranging from 300 to 4700 nm,lack of a smooth muscle layer or innervation, a wide lumen, and im-paired functional receptors for angiotensin II [47]. Furthermore, lym-phatic drainage of tumor tissues is usually deficient [3,38]. Thesefactors alter the molecular and fluid transport dynamics in the tumorand increase the retention of nano-sized drugs. While being above therenal filtration threshold, macromolecules larger than 40 kDa, selec-tively leak from tumor vessels and accumulate in tumor tissues forprolonged periods [47]. Arnida et al. also showed that geometry (sizeand shape) as well as surface characteristics influence theirbiodistribution, circulation time and tumor uptake [22]. Additionalstudies demonstrate that the EPR effect is further accentuated by ex-travasation factors such as VEGF, bradykinin, nitric oxide (NO), prosta-glandins, peroxynitrite andmatrix metalloproteinases [48,49]. All thesefactors increase blood flow and promote diffusion and retention ofnanomedicines inside tumors.

The recognition of the EPR effect among researchers in the field ofdrug development has resulted in a considerable momentum tothe field of nanomedicine. The EPR effect served as a basis for thedevelopment of anticancer macromolecular drugs to improve its thera-peutic efficacy by targeting tumor tissues and decreasing systemic ad-verse effects compared to free drug. Most low molecular weightanticancer drugs lack specific targeting and can traverse blood vesselsfreely; the consequent distribution of the drug to healthy tissues usually

Approval (year) Treatment References

FDA (1995) Late-stage ovarian cancer [26]Advanced HIV-associated Kaposi's sarcoma [27]

FDA (1996) Advanced HIV-associated Kaposi's sarcoma [28]FDA (1999) Malignant lymphomatous meningitis [29]Europe (2000) Metastatic breast cancer in combination

with cyclophosphamide[30]

Canada (2001)Europe (2004) Osteosarcoma [31]FDA (2006) Acute lymphoblastic leukemia [32]South-Korea (2006) Metastatic breast cancer [33]Japan (1993) Hepatocellular carcinoma [34]FDA (2005) Metastatic breast cancer [35]FDA (1996) MRI contrast agent [36]FDA (1996) MRI contrast agent [37]

Page 3: Anticancer nanomedicine and tumor vascular permeability; Where is the missing link?

Fig. 1. Differences between normal and tumor tissue in relation to targeting ofnanomedicines by the enhanced permeability and retention (EPR) effect. (A) Normaltissue contains tightly connected endothelial cells which prevents the diffusion of thenanomedicine outside the blood vessel. (B) Tumor tissue contains large fenestrationbetween the endothelial cells allowing the nanomedicines to reach the matrix andthe tumor cells by the EPR effect. VEGF secreted by tumor cells, stroma cells and mac-rophages increases permeability and stimulates angiogenesis and the migration of en-dothelial cells towards the tumor. A considerable proportion of the nanomedicinenever reaches the tumor either due to entrapment or nonspecific interaction withcollagen composing the matrix or removal through macrophage endocytosis.Nanomedicines tend to concentrate at the periphery of the tumor, only a small propor-tion will diffuse to the center of the tumor.

267S. Taurin et al. / Journal of Controlled Release 164 (2012) 265–275

results in adverse effects [50]. The EPR effect was predicted in the ma-jority of human solid tumors with the exception of poorly vascularisedtumor such as prostate and pancreatic cancer [50]. The expectation thatnanotechnology would result in the development of selective antican-cer drugs utilizing the EPR effect was widespread. However, with theexception of the nine nanomedicines listed in Table 1, the transition ofnanomedicines from the laboratory to an effective clinical anticancerdrug has been disappointingly slow. A major drawback in the special-ized field of nanomedicine arises from a dissociation between differentfacets of science. Currently the ability to design and develop anticancernanomedicine far surpasses the understanding of the EPR effect and itsbiological principles. Anticancer nanomedicine has so far not succeededin fully exploiting the EPR phenomenon for optimum cancer manage-ment. The following describes some of the commonly overlooked fac-tors that could account for the slow transition of EPR based anticancerdrugs towards clinical application.

3.1. Influence of EPR based drug design on internalization, release rate,and biocompatibility

Integral to utilization of the EPR effect is the design of specific drugdelivery systemwith amolecular size larger than 7 nm. A hydrodynam-ic diameter of a nanoparticle lower than 7 nm is rapidly cleared fromthe body via renal filtration and urinary excretion [18]. The physico-chemical characteristics of engineered nanosystems should be consid-ered critically from the biological perspective in order to optimize itsefficacy. In many instances, the nature of the nanomedicine itself hasbeen a limiting factor that negatively impacted its chance of clinical suc-cess. The loading of active drug into the delivery system can result inpractical limitations to the dose needed to be delivered to tumor tissue.For example, the HPMA copolymer-paclitaxel conjugate showed insuf-ficient drug loading (≤10%) with a particle size in the range of 12–15 nm [51], and lacked ester linker stability [52]. Consequentlyinsufficient tumor tissue accumulation of the drug was evident inphase I clinical trials [53]. Another factor limiting the efficacy of thenanomedicines is a fast release rate of drug in the circulation. Forinstance, the low molecular weight HPMA copolymer-camptothecinconjugate showed a rapid release of drug and quick renal filtrationand consequent bladder toxicity in phase I clinical trials [54]. Thenanomedicinewas designedwith a labile ester linker, decreasing its sta-bility and therefore its tumor accumulation [55]. In this example, a lowmolecularweight (below renal excretory threshold) coupled to the tox-icity associated with a fast release rate resulted in the drug failing toachieve EPR based pharmacokinetics.

Following are a few considerations inherent to the design ofnanomedicine that may significantly influence the outcome of EPRbased drug targeting.

3.1.1. Intercellular internalizationThe concentration of drug inside tumor resulting from the EPR ef-

fect in a subset of highly vascularized tumors does not guarantee theefficient internalization of the drug within the cytoplasmic or nuclearcompartments of the tumor cells. Multiple factors can influence thecellular internalization process of the nanomedicine. Usually,nanoparticles and polymer-based drug delivery systems are internal-ized by endocytosis, a multistep process that culminates in the forma-tion of a late endosome which finally fuses with a lysosome [56].Malignant cells have an accelerated metabolism, a high glucose re-quirement and an increase glucose uptake characterized by the ele-vated expression of glucose transporter proteins (GLUT) [57].However, recent studies have shown that many cancer cell lines ex-hibit limited capacity for endocytosis compare to normal cells [58,59].

In addition, nanomedicine can be frequently recognized as foreignbodies resulting in its rapid uptake and elimination by specializedcells of the reticulo-endothelial system (RES).

Compared to tumor cells, macrophages usually exhibit a higher up-take of nano-sizedmolecules [22,60] as they can recognize nanomedicineeither through their Toll-like receptor 4 (TLR-4) [61] or through scaven-ger receptors [62]. Much work has therefore been devoted to the devel-opment of nanoparticles which can evade macrophage recognition,resulting in longer circulatory time and increased interaction with targettissue. On this basis, polyethylene glycol (PEG) is the polymermost com-monly used to enhance in vivo circulatory half-life [63,64]. Coatingnanoparticles with PEG results in the formation of a polymeric layerwhich sterically hinders the interaction of nanoparticleswith plasmapro-teins and cell membranes [65] preventing opsonization and phagocytosisby components of the RES [66,67]. PEG-liposome-incorporated doxorubi-cin (Doxil®) is approved by the FDA for the treatment of ovarian cancer(see Table 1). Additional polymers such as N-(2-hydroxypropyl)methylacrylamide (HPMA), polyacrylamide or poly(vinyl pyrrolidone)have also been used to improve the circulation time and steric hindranceof nanomedicines [68,69]. The main disadvantage of this strategy limitsthe interaction of (stealth) nanoconstructs with the tumor cell mem-brane and subsequently reduced internalization and uptake by tumorcells. To improve specific uptake by endocytosis, several nanoparticleshave been coated with a ligand of folate [70] or transferrin [71] to inducereceptor mediated endocytosis. These coatings increased the accumula-tion of drug inside tumor cells. However, the practical advantages in themanagement of human tumors in the clinic remain to be proven. Follow-ing intracellular internalization, active drug should be liberated from thelysosomal compartment to reach its cellular target. Mechanisms to es-cape the lysosomal compartment and improve intracellular targeted de-livery have been described by Breunig, Bauer and Goepferich [72].Another consideration relevant to relatively large sized macromolecularnanomedicine is their nonspecific interactions with the extracellular ma-trix: to reach tumor cells, nanoconstructsmustmove through thematrix,a highly interconnected network of collagen fibers that intermingle withproteins such as proteoglycans and glycosaminoglycans. This semi-solidbarrier could significantly reduce the amount of nanomedicine reachingtumor cells through nonspecific interaction (Fig. 1) or simply by imped-ing convection movements of relatively large size of nanoconstructs[73]. This could lead to nanomedicine being locally concentrated in

Page 4: Anticancer nanomedicine and tumor vascular permeability; Where is the missing link?

Fig. 2. Variation of nanocarrier release rate and accumulation profile (A) Determinationof the release rate over a 24 h period of different nanocarriers across several studies[166–175]. (B) Comparison of the ratio of free to nanoconstruct associated drugs invarious tissues normalized across studies as % of injected dose per gram of tissue.[176–191]. (C) Comparison of the ratio of free to nanoconstruct associated drugs intumor tissue normalized across studies as % of injected dose per gram of tissue relativeto the animal model used [176,177,184,189,192–195].

268 S. Taurin et al. / Journal of Controlled Release 164 (2012) 265–275

proximity to the capillary that it leaked fromwithout reaching the targettumor cells.

Recently, several studies have developed methods to circum-vent these limitations such as using of the tumor penetrating pep-tide, iRGD which has been shown to increase the delivery ofnanomedicines in solid tumors by improving its interstitial trans-port [74]. Additional therapeutic strategies aiming to normalizethe tumor vasculature and extracellular matrix in order to improvetumoral penetration of the drug have been described (for review[75,76]).

3.1.2. Release rateConjugates can be synthesized through covalent linking of drugs

to polymeric carriers such as SMANCS (Table 1) [17]. In comparison,entrapment of drug inside a micellar structure requires either cova-lent or non-covalent bonds (ionic, hydrogen bonds or hydrophobic)and involves a block polymer or copolymer. Various chemical bondssuch as amide, ester, azide, imine, hydrazone, thioether and urethaneare currently used to prepare nanomedicines [77,78]. Based on the na-ture of these chemical bonds, the release of drug from its carrier can de-pend on either pH, usually acidic pH of the lysosome [77], temperature[43] or on enzymatic cleavage [79]. Furthermore, the nature of this bondwill determine the release rate; for example an ester bond ensures arapid release of drug due to an abundance of esterases in plasmawhere-as an amide bond will show a slower release profile [54,78].

The comparison of release rates between polymer conjugates, li-posomes and micelles nanomedicines after 24 h incubation (Fig. 2A)in different studies showed a distinctive profile. Overall, polymer con-jugates and micelles have a comparable release rate which is higherthan that of liposomes (Fig. 2A). The release profile of liposomes ap-pears relatively homogeneous with a mean value of 24% (3–39%)while the release profile of conjugates and micelles appears heteroge-neous across several studies with a mean value of 39% (2.5–100%)and 41% (2.5–100%) respectively.

In order for a nanocarrier to provide tumor targeting, the carriershould have stable chemical bond with the cargo drug while in circu-lation. This prevents the rapid release of free drug and permits a ther-apeutic effect at the site of action. A rapid drug release from itsdelivery system in plasma can result in a biodistribution and toxicityprofile comparable to its related free drug. In contrast, engineering astable linkage between the drug and its carrier can result in a slow re-lease rate at the target site and inability to reach the critical therapeu-tic concentration. The release rate of nanoconstructs needs to betailored for the treatment of a specified tumor doubling time (seeTable 2). Thus, the choice of a specific linker is critical for a favorableanticancer outcome of EPR targeted nanosystems.

3.1.3. BiocompatibilityWith success of EPR based drug targeting, accumulation of active

drug inside the tumor rarely exceeds 5% of the total dose ofnanomedicine administrated by i.v. injection. The majority of injecteddose accumulates in various organs such as liver and spleen and to aminor extent kidneys and lungs [104]. Surface modification of thenanomedicine, such as PEGylation, may increase their retention inthe systemic circulation and favor tumor accumulation. However,more than 90% of PEGylated nanoparticles will still be removedthrough liver clearance within several hours of administration. Stud-ies have demonstrated that, as little as ~2% of the total i.v. adminis-tered dose was found in the tumor after 4 h [104]. Thus there is alegitimate safety concern regarding the off target accumulation ofthe drug delivery system. Ideally, the drug carrier should be eliminat-ed after drug release. But, unless the nanocarrier is biodegradable, itwill remain in the body and be dealt with as a foreign body. Theinnate elements of the immune system could be stimulated non-specifically by these foreign bodies through TLR-4 [105]. Activatedmacrophages will phagocytose and attempt to degrade the nanocarrier

in its lysosomal compartment. Failure to do so may lead to the forma-tion of foreign body giant cells caused by fusion of multiple macro-phages or monocytes [106] and ultimately to the formation of lesionsresembling granulomas [107]. This can potentially result in the patho-logical formation of a dense fibrous capsule replacing the originalfunctional tissue. Another concern in relation to the accumulation ofnon-degradable materials is the induction of malignancy resultingfrom frustrated phagocytosis and prolonged inflammation [108](Fig. 3).

Page 5: Anticancer nanomedicine and tumor vascular permeability; Where is the missing link?

Fig. 3. Schematic representation of the variables that can influence the outcome of EPRbased nanomedicine. (A) Interpretation of data obtained from animal model needscareful consideration of the immune status of the animal (1), the type of tumor cell or-igin (2), as well as the site of injection (3). (B) In translation of specific nanomedicinetowards clinical use, tumor doubling time which is usually related to high vascular den-sity and elevated expression of VEGF and its receptor, need to be considered. Hepatocel-lular carcinoma (1) and prostate cancer (2) are two extremes; hepatocellular carcinomahas a rapid tumor doubling time (42 days for themost aggressive) and a highmicrovas-cular density of 127.2. In contrast, prostate cancer is characterized by a slow tumor dou-bling time, low microvascular density of 39.2 as well as large variation in VEGFexpression. Off target accumulation (3) mainly to liver and spleen. Concern of chronictoxicity (4) associated with non-biodegradable nanoparticles and the presence ofnanoparticles in the brain such as silver (5).

Table 2Expression of VEGFR isoforms and ligands in various cancer cell lines and tumors.

Tissue Origin VEGFR-1 VEGFR-2 VEGFR-3 VEGF-A VEGF-B VEGF-C VEGF-D References

Breast Cell line MDA-MB-231 ++ + + + – +++ +/– [80–85]MCF-7 +++ +/– + + + ++ – [81–84,86,87]

Tissue Tumor 70% (28/39) [87]69% (73/106) 89% (97/109) [88]

39% (36/93) [89]Prostate Cell line DU145 + – ++ + – + [84,90,91]

LNCaP +++ + +++ ++ +/– ++ [84,90–92]PC-3 +++ ++ +++ ++ + + ++ [84,90,91,93,94]

Tissue Tumor W: 23% (16/71) W:26% (19/71) W: 26% (19/71) [95]M: 39% (28/71) M: 60% (43/71) M:39% (28/71)S: 38% (27/71) S: 14% (9/71) S:35% (24/71)

W: 25% (3/12) W: 100% (12/12) [96]M: 58% (7/12)S: 17% (2/12)

Lung Cell line A549 +/– – + + + – [97–99]Calu-3 ++ + –/+ ++ + – [100]

Tissue Non small celllung tumor

94.2% (97/103) 66% (68/103) 80.6% (83/103) 92.2% (95/102) [101]41.7% (20/48) 54.2% (26/48) 41.7% (20/48) 39.6% (19/48) 12.5% (6/48) 29.2% (14/48) 22.9% (11/48) [102]85.2% (23/27) 88.9% (24/27) [103]

W: Weak, M: Medium and S: Strong.

269S. Taurin et al. / Journal of Controlled Release 164 (2012) 265–275

To address these issues, recent work has focused on the develop-ment of biodegradable and non-immunologic drug delivery systemscontaining either enzymatically or reductively degradable spacerssuch as poly-(-D,L-lactide-co-glycoside) (PLGA) [109] or the starHPMA polymer carrier which enable a controlled degradation of thedrug carrier [110,111]. Some of these carriers demonstrate prolongedblood circulation and tumor drug accumulation but difficulties in thereproducibility of their synthesis could hamper further clinical devel-opment [110].

3.2. Validations of animal models

The EPR effect has been repeatedly proven in animal modelsthrough the use of EBD. EBD binds instantly to plasma albuminwhich results in a large molecular weight complex of about 7 nm di-ameter that can simulate the effect of a nanomedicine. After 6 h, thereis usually a distinct accumulation in tumor lesions compared to sur-rounding tissues. Similarly, many nanomedicines have been observedto accumulate in tumor tissue up to 27-fold more than free drugs(Fig. 2B). The question of whether the results of EPR-based drugtargeting in animal models can be faithfully translated to the clinic re-mains unanswered.

Production of VEGF bymacrophages and its role in cancer develop-ment is well documented [112,113]. To test the anticancer propertiesof a given nanomedicine, it is commonpractice to utilize immunocom-promisedmice to enable the use of human tumor xenografts. Howeverhuman cancer patients are rarely immunocompromised. A change inmacrophage activity in immunocompromised mice [114] can resultin less VEGF leading to a tumor with reduced vascular density,which in turn limits the access of the nanoconstruct to the tumor. Fur-thermore, results obtained from immunocompromised models differfrom results obtained in immunocompetent mice. Reviewing 22 stud-ies involving various drug delivery systems (conjugates, liposomesandmicelles), revealed an approximate two-fold increase in tumor ac-cumulation of nanoconstructs in immunocompetent mice relative toimmunocompromised ones Fig. 2C. Moreover, immunocompetentmice bear murine tumors and not human cancer cell lines which fur-ther complicate interpretation of in vivo animal data and jeopardizeits value in predicting the performance of new drugs in clinical trials.Table 2 compares the expression of VEGF and its receptors betweencommonly used human tumor cell lines and their clinically isolatedvariants. It is clearly evident that tumor cell lines have pronounced ex-pression of VEGF and its receptors with far less variability in compar-ison to clinical tumors. Relevant to this is the design of targeted

Page 6: Anticancer nanomedicine and tumor vascular permeability; Where is the missing link?

270 S. Taurin et al. / Journal of Controlled Release 164 (2012) 265–275

nanomedicine to tumors which relies on the conjugation of target li-gands that bind strongly to tumor cell surface receptors to increasecell recognition and cellular uptake. Galactosamine [115], transferrin[116] and folate [117] have been incorporated in nanomedicinebased on the preferential expression of these molecules by cancercells. Despite promising in vitro studies, these targeted nanomedicinefailed to demonstrate significant benefit at the preclinical or clinicallevel [118]. The discrepancy between the results obtained from testingspecific tumor cell lines in tumor model and the clinical trials pointsfurther to the sampling errors in generalizing the results of from spe-cific cell line to that of relevant tumors [119].

A substantial difference between tumormodels in animals and thoseof human patients is the progression rate. Animals usually develop alarge, clinically relevant tumor (>5 mm) one week following subcuta-neous (SC) tumor cell inoculation, while such a tumor volume can takeyears to develop in a human (Table 3). This rapid progression rate in an-imalmodels results in the overestimation of the targeting role of EPR ef-fect. Animal tumors developing quickly presumably produce a largequantity of VEGF and vascular mediators to support their rapid growth.In addition a one-gram tumor mass in a thirty-gram mouse is about 3%of its total weight. In humans, a comparable tumorwouldweigh 2–5 kg,which is an advanced tumor stage that is not ideal for utilization of an-ticancer nanomedicine. Finally, tumors are usually implanted SC in an-imal models, which allow the developing tumors to take advantage ofthe extensive cutaneous vascular network for extending their bloodsupply. A condition that is rarely encountered in human malignancy.

Data collected from available literature to date show that althoughthere is a trend towards higher concentration of nanoconstructs in SCmodels, the results are however not conclusive given the limitednumber of studies. Whether site of tumor development can influencethe efficacy of the EPR effect remains an unanswered question.

3.3. Tumor biology diversity

3.3.1. Tumor doubling time (TDT)Tumor doubling time (TDT) is an important factor to consider

when designing EPR based anticancer nanomedicine. In general, fast

Table 3Tumor doubling time of various types of cancers.

Histological type Doubling time(days)

Numberof tumors

References

Adenocarcinoma (lung) 1–964 15 [120]High grade astrocytoma 60–78 3 [121]Low grade astrocytoma 108–330 9 [121]

Breast cancer 11–1293 118 [122]Bronchioalveolar carcinoma 36–1092 9 [120]Cervical cancer 58–1500 61 [123]Colon cancer 195–620 27 [124]Colorectal Carcinoma 53–1570 27 [125]Primary melanoma 50–377 4 [126]Metastatic melanoma 8–212 3 [127]

Hepatocellular carcinoma 42–238 17 [127]Hepatocellular carcinomapoorly differentiated

20–78 6 [128]

Hepatocellular carcinomamoderately differentiated

94–380 6 [129]

Hepatocellular carcinomawell differentiated

38–274 19 [129]

Meningioma 133–4750 15 [130]Ovarian solid tumor 60–115 8 [131]Pancreatic carcinoma 64–255 9 [132]Pituitary adenoma 506–5378 38 [133]Prostate cancer 360–1890 43 [134]Renal Cell Carcinoma 93–1113.2 56 [135]Small cell lung cancer 61.9–120.4 13 [136]Stomach cancer 12–105 112 [137]

doubling times (DTs) provide a selection trait that is exploited bychemotherapeutics that target processes necessary for mitosis suchas DNA synthesis and cytoskeleton remodeling. Many chemothera-peutic agents fail to cope with rapidly dividing tumors as the amountof drug necessary to kill a given number of cells will double with eachtumor doubling. However, the dose that will elicit dose limiting tox-icity will remain the same. A short TDT is well known to be associatedwith an unfavorable survival prognosis [138–143].

TDT is a highly heterogeneous, both within and between differenttumor types, stages and grades. There is a large degree of variation ofTDT between tumors of different tissue origins. Pituitary adenoma, forexample, has an extremely long DT of 506–5378 days [133] whilestomach cancer may have a minimal DT of as little as 12 days [137].

Within tumor types variation is also pronounced. For instance, lungadenocarcinoma has an extremely high variation in TDT of 964-fold[120] followed by breast cancer with a variation of 117.5-fold [122](see Table 3). TDT can also differ according to the specific cellular originwithin a given tissue. Bronchoalveolar cancer, for example, has an ex-tremely varied TDT of 36–1092 days, a variation of 30.3 fold [120]while small cell lung cancer has a TDT of 61.9–120.4, amere 1.9-fold dif-ference [136]. In addition, TDT can range depending on tumor grade.Poorly differentiated hepatocellular carcinoma is highly invasive and,predictably, has a DT of 20–78 days [128] while moderately differenti-ated hepatocellular carcinoma has a significantly extended DT of 94–380 days [129]. Astrocytoma also follows this trend with DT ofhigh-grade astrocytoma at 60–78 days and the TDT of low-grade astro-cytoma at 108–330 days. The primary or metastatic status of a tumorcan also cause large fluctuations. For example primary melanoma mayhave a DT of 50–377 days [126] while metastatic melanoma may havea DT of 8–212 days [127] (see Table 3).

Scientists designing EPR based anticancer nanomedicine shouldconsider doubling time variation when planning the release mecha-nism of active chemotherapeutic agents from its nanocarrier, as wellas the internalization rate of macromolecular complexes into tumorcells. For example, a slow releasing amide bond between the polymerback bone and the drug, or slowly internalized liposome, could bothbe a good choice for tumors with a slow DT.

In contrast a fast releasing micelle or an ester bond linkage can bea better fit for rapidly dividing tumors. Generally, EPR basednanomedicine has a wider therapeutic window [144], an advantagethat can be exploited to shape dose regimens based on individual pa-tient conditions. Tumor inherent sensitivity to specific chemothera-peutic agents as well as TDT is of the upmost importance indesigning EPR based anticancer nanomedicine.

3.3.2. Variation in tumor vascular densityThe EPR effect is a phenomenon that strictly depends on the vascula-

ture of the tumor with theoretical assumption that all tumors indepen-dently of their origin, stage and organswill behave identically. However,this conception is drastically challenged by a number of reports thatshow a high diversity in angiogenesis behavior [76,145–147]. Nagy etal. have identified six structurally and functionally distinct types ofblood vessels in human cancers [147]. Vascular density may relate totumor progression. As shown in Table 4, vascular density is largely de-pendent on the type of cancer and varies largely within each tumortype. For instance, renal cell carcinoma is highly vascularized [148]while the density of micro-vessels appears low in head and neck squa-mous cell carcinoma [149] or in ovarian carcinoma [150]. In addition,higher stages of cancer are well correlated with higher microvasculardensity as observed in non-small cell lung cancer [151] while in othertypes of tumors such as renal cell carcinoma no direct correlation canbe established between tumor stage and vascular density [152]. Further-more, metastatic tumors tend to possess higher vascular densitycompared to non-metastatic tumors [153–156]. Another element re-garding the EPR effect is the secretion of VEGF by the tumor. Vascularpermeability can be altered by VEGF as well as a wide array of

Page 7: Anticancer nanomedicine and tumor vascular permeability; Where is the missing link?

Table 4Microvessel vascular density of various types of cancers.

Tissue Tumor Microvessel vascular density (vessel/field) Patient (n) Endothelial marker References

Liver Hepatocellular carcinoma 127.2 (42–246) 50 CD34 [148]Lung Non small cell lung cancer (stage I–II) 23.6 (7.4–39.8) 33 Factor VIII [151]

Non small cell lung cancer (stage III–IIIb) 65.3 (33.4–98.7) 22 Factor VIII [151]Endometrial Carcinoma 77.5 (19–189) 34 Factor VIII [157]Kidney Renal cell carcinoma (stage I–II) 75.7 (37–114) 25 CD34 [152]

Renal cell carcinoma (stage III–IV) 68 (26–110) 16 CD34 [152]Colorectal Carcinoma 77 (39–129) 21 CD31 [158]

145 (132–160) 28 CD34 [159]Breast Non-metastatic carcinoma 45 (15–100) 32 Factor VIII [153]

Metastatic carcinoma 101 (16–220) 30 Factor VIII [154]Prostate Non-metastatic carcinoma 39.2 (20.6–57.8) 45 Factor VIII [156]

Metastatic carcinoma 76.8 (32.2–121.4) 29 Factor VIII [156]Head and neck Squamous cell carcinoma 14.8 (4–39) 32 CD34 [149]Ovary Carcinoma 49.4 (16–118) 43 CD34 [150]Brain Glioblastoma 49 (27–99) 46 CD105 [160]Skin Malignant melanoma 33.6 (17.1–50.1) 17 Factor VIII [155]

Malignant melanoma metastatic 35.8 (20.3–51.3) 20 Factor VIII [155]

271S. Taurin et al. / Journal of Controlled Release 164 (2012) 265–275

inflammatory mediators [10], which can affect the extent ofnanomedicine accumulation driven by the EPR effect and the penetra-tion of nanodrug inside the tumor. As shown in Table 2, there is a largeheterogeneity in the expression of VEGF seen when comparing prostateand lung cancers.

When designing the nanocarrier, the properties of the targetedtumor tissue such as the cancer type, the microvascular density,and the secretion of permeability factors such as VEGF should thereforebe taken into account in order to take full advantage of the EPRphenomenon.

3.3.3. EPR related pharmacokineticsIn classical pharmacology, the tissue concentration of a certain

drug cannot exceed its plasma concentration. With respect to tradi-tional pharmacokinetics, any administered drug is equally distributedto different body compartments indiscriminately forming the volumeof distribution. Conventional chemotherapeutic agents usually exhib-it large volume of distribution. For example, doxorubicin has avolume of distribution of ~680 L [161] indicating that it is indiscrim-inately partitioned to various organ tissues.

As drugs reach sizes of 7 nm, classical pharmacokinetics roles can-not be accurately applied due to the acquisition of two drasticchanges. Firstly, nano-sized drugs cannot be eliminated by renal glo-meruli as they exceed the renal threshold of excretion dictated by thepore size in the glomeruli [19,162]. Secondly, their organ distributionis limited to tissues that have capillaries with large enough endothe-lial fenestrations to allow macromolecular drugs to pass through[19,20]. The EPR effect utilizes the unique characteristic of largegaps between endothelial cells that makes up tumor vessels. Usuallythese gaps can vary from few nanometers to up to 400 nm in size[151,163]. At this large size, nanomedicines can preferentially accu-mulate in tumor tissues. However, tumors are not the only organswith such large fenestration as the spleen and liver show similarcharacteristics. Liver sinusoid can have fenestration of around100 nm in humans [164] whereas the spleen has large sinusoidlumina of 5 μm that can support extravasations of aged red bloodcells [19]. With such a large fenestration size, great amounts ofnano-sized drugs are filtered by these organs. As shown if Fig. 2Bmeta-analysis of 73 studies over the last 10 years revealed that withall the EPR effect-based nanoconstructs that were used, liver andspleen were the two major organs competing with tumors for thenanoconstructs (conjugate, micelles, and liposomes). While spleenfunction can be compensated for by other lymphatic organs, liverdamage due to the concentration of cytotoxic nanomedicine remainsa potential challenge to successful anticancer drug targeting. For ex-ample, nanoconstructs of cis-platinum have reduced toxicity in thekidney compared to the free drug, but results in a dose limiting

liver toxicity [165]. To summarize, EPR related pharmacokinetic pa-rameters such as long circulatory half-life, reduced elimination, andaltered distribution could be a double edge sword. Careful consider-ation of these parameters is essential for effective and more personal-ized cancer treatments and the prevention of unexpected toxicity.

4. Conclusion

The EPR concept has been the corner stone in the developmentof a new class of anticancer drugs aiming at increased anticanceractivity and lower toxicity. Contrary to the high expectations ofEPR effect, only a few drugs based on this principle have reachedthe clinic over the last 20 years. Unjustified generalization of resultsobtained from subcutaneous animal tumor models to human coun-terparts, might have in part contributed to the slow progressionof anticancer nanomedicine. Human cancer diversity, characteristicsof commonly used animal tumor models, biodistribution of nano-sized molecules, variable release rates, slow intracellular internali-zation and biocompatibility all are important factors to take into ac-count when designing optimal nanomedicines. Wise considerationof these variables will ensure a prosperous future for EPR-basedcancer chemotherapy.

Acknowledgment

KG gratefully acknowledges the support of Professor Hiroshi Maeda.The EPR effect was first described and extensively studied by ProfessorMaeda's group in the department ofMicrobiology, Kumamoto University,Japan. This work has been supported by departmental fund no.;(PL. 108403.01.S. LM) to KG and HRC Emerging Researcher First Grant(PL. 108360.01.P.LM), UORG and AG305 for ST from the Department ofPharmacology and Toxicology, Otago University. KG thanks Dr. FlorianeImhoff, Ms Céline Bourdon and Mr SimranMaggo for proof reading thearticle.

References

[1] E. Goldmann, The growth of malignant disease in man and the lower animals,with special reference to the vascular system, Proc. R. Soc. Med. 1 (1908) 1–13.

[2] K. Strebhardt, A. Ullrich, Paul Ehrlich's magic bullet concept: 100 years of prog-ress, Nat. Rev. Cancer 8 (2008) 473–480.

[3] J. Folkman, Tumor angiogenesis: therapeutic implications, N. Engl. J. Med. 285(1971) 1182–1186.

[4] J.C. Underwood, I. Carr, The ultrastructure and permeability characteristics ofthe blood vessels of a transplantable rat sarcoma, J. Pathol. 107 (1972) 157–166.

[5] H.I. Peterson, K.L. Appelgren, Experimental studies on the uptake and rententionof labelled proteins in a rat tumour, Eur. J. Cancer 9 (1973) 543–547.

Page 8: Anticancer nanomedicine and tumor vascular permeability; Where is the missing link?

272 S. Taurin et al. / Journal of Controlled Release 164 (2012) 265–275

[6] J.D. Ward, M.G. Hadfield, D.P. Becker, E.T. Lovings, Endothelial fenestrations andother vascular alterations in primary melanoma of the central nervous system,Cancer 34 (1974) 1982–1991.

[7] N.B. Ackerman, P.A. Hechmer, Studies on the capillary permeability of experi-mental liver metastases, Surg. Gynecol. Obstet. 146 (1978) 884–888.

[8] H. Maeda, K. Greish, J. Fang, The EPR Effect And Polymeric Drugs: A ParadigmShift for Cancer Chemotherapy in the 21st Century, Springer, Heidelberg, Germany,2006.

[9] D.R. Senger, C.A. Perruzzi, J. Feder, H.F. Dvorak, A highly conserved vascular per-meability factor secreted by a variety of human and rodent tumor cell lines, Can-cer Res. 46 (1986) 5629–5632.

[10] S.M. Weis, D.A. Cheresh, Pathophysiological consequences of VEGF-induced vas-cular permeability, Nature 437 (2005) 497–504.

[11] N. Ferrara, B. Keyt, Vascular endothelial growth factor: basic biology and clinicalimplications, EXS 79 (1997) 209–232.

[12] J.A. Nagy, S.H. Chang, A.M. Dvorak, H.F. Dvorak, Why are tumour blood vesselsabnormal and why is it important to know? Br. J. Cancer 100 (2009) 865–869.

[13] J.A. Nagy, A.M. Dvorak, H.F. Dvorak, Vascular hyperpermeability, angiogenesis,and stroma generation, Cold Spring Harb. Perspect. Med. 2 (2012) a006544.

[14] H. Maeda, Y. Matsumura, Tumoritropic and lymphotropic principles of macro-molecular drugs, Crit. Rev. Ther. Drug Carrier Syst. 6 (1989) 193–210.

[15] Y. Matsumura, H. Maeda, A new concept for macromolecular therapeutics incancer chemotherapy: mechanism of tumoritropic accumulation of proteinsand the antitumor agent smancs, Cancer Res. 46 (1986) 6387–6392.

[16] H. Maeda, J. Fang, T. Inutsuka, Y. Kitamoto, Vascular permeability enhancementin solid tumor: various factors, mechanisms involved and its implications, Int.Immunopharmacol. 3 (2003) 319–328.

[17] H. Maeda, J. Takeshita, R. Kanamaru, A lipophilic derivative of neocarzinostatin.A polymer conjugation of an antitumor protein antibiotic, Int. J. Pept. ProteinRes. 14 (1979) 81–87.

[18] H.S. Choi, W. Liu, P. Misra, E. Tanaka, J.P. Zimmer, B. Itty Ipe, M.G. Bawendi, J.V.Frangioni, Renal clearance of quantumdots, Nat. Biotechnol. 25 (2007) 1165–1170.

[19] H. Sarin, Physiologic upper limits of pore size of different blood capillary typesand another perspective on the dual pore theory of microvascular permeability,J. Angiogenes. Res. 2 (2010) 14.

[20] K. Greish, Enhanced permeability and retention of macromolecular drugs insolid tumors: a royal gate for targeted anticancer nanomedicines, J. Drug Target.15 (2007) 457–464.

[21] K. Greish, Enhanced permeability and retention (EPR) effect for anticancernanomedicine drug targeting, Methods Mol. Biol. 624 (2010) 25–37.

[22] M.M. Arnida, A. Janát-Amsbury, C.M. Ray, H. Peterson, Ghandehari, Geometryand surface characteristics of gold nanoparticles influence their biodistributionand uptake by macrophages, Eur. J. Pharm. Biopharm. 77 (2011) 417–423.

[23] S.M. Moghimi, A.C. Hunter, J.C. Murray, Long-circulating and target-specificnanoparticles: theory to practice, Pharmacol. Rev. 53 (2001) 283–318.

[24] Y. Zhu, L. Che, H. He, Y. Jia, J. Zhang, X. Li, Highly efficient nanomedicines assem-bled via polymer–drug multiple interactions: tissue-selective delivery carriers,J. Control. Release 152 (2011) 317–324.

[25] M.L. Immordino, F. Dosio, L. Cattel, Stealth liposomes: review of the basic sci-ence, rationale, and clinical applications, existing and potential, Int. J.Nanomedicine 1 (2006) 297–315.

[26] A.N. Gordon, J.T. Fleagle, D. Guthrie, D.E. Parkin, M.E. Gore, A.J. Lacave, Recurrentepithelial ovarian carcinoma: a randomized phase III study of pegylated liposo-mal doxorubicin versus topotecan, J. Clin. Oncol. 19 (2001) 3312–3322.

[27] N.D. James, R.J. Coker, D. Tomlinson, J.R. Harris, M. Gompels, A.J. Pinching, J.S.Stewart, Liposomal doxorubicin (Doxil): an effective new treatment for Kaposi'ssarcoma in AIDS, Clin. Oncol. 6 (1994) 294–296.

[28] FDA approves DaunoXome as first-line therapy for Kaposi's sarcoma. Food andDrug Administration, J. Int. Assoc. Physicians AIDS Care 2 (5) (May 1996) 50–51.

[29] B.S. Chhikara, K. Parang, Development of cytarabine prodrugs and delivery sys-tems for leukemia treatment, Expert Opin. Drug Deliv. 7 (2010) 1399–1414.

[30] C.E. Swenson, L.E. Bolcsak, G. Batist, T.H. Guthrie Jr., K.H. Tkaczuk, H. Boxenbaum,L. Welles, S.C. Chow, R. Bhamra, P. Chaikin, Pharmacokinetics of doxorubicin ad-ministered i.v. as Myocet (TLC D-99; liposome-encapsulated doxorubicin cit-rate) compared with conventional doxorubicin when given in combinationwith cyclophosphamide in patients with metastatic breast cancer, AnticancerDrugs 14 (2003) 239–246.

[31] K. Ando, K. Mori, N. Corradini, F. Redini, D. Heymann, Mifamurtide for the treatmentof nonmetastatic osteosarcoma, Expert. Opin. Pharmacother. 12 (2011) 285–292.

[32] P.A. Dinndorf, J. Gootenberg, M.H. Cohen, P. Keegan, R. Pazdur, FDA drug approv-al summary: pegaspargase (oncaspar) for the first-line treatment of childrenwith acute lymphoblastic leukemia (ALL), Oncologist 12 (2007) 991–998.

[33] C. Oerlemans, W. Bult, M. Bos, G. Storm, J.F. Nijsen, W.E. Hennink, Polymeric mi-celles in anticancer therapy: targeting, imaging and triggered release, Pharm.Res. 27 (2010) 2569–2589.

[34] H. Maeda, G.Y. Bharate, J. Daruwalla, Polymeric drugs for efficient tumor-targeteddrug delivery based on EPR-effect, Eur. J. Pharm. Biopharm. 71 (2009) 409–419.

[35] W.J. Gradishar, Albumin-bound paclitaxel: a next-generation taxane, Expert.Opin. Pharmacother. 7 (2006) 1041–1053.

[36] G. Morana, E. Salviato, A. Guarise, Contrast agents for hepatic MRI, Cancer Imag-ing 7 (2007) S24–S27 (Spec No A).

[37] C.W. Jung, Surface properties of superparamagnetic iron oxideMR contrast agents:ferumoxides, ferumoxtran, ferumoxsil, Magn. Reson. Imaging 13 (1995) 675–691.

[38] Y. Noguchi, J. Wu, R. Duncan, J. Strohalm, K. Ulbrich, T. Akaike, H. Maeda, Earlyphase tumor accumulation of macromolecules: a great difference in clearancerate between tumor and normal tissues, Jpn. J. Cancer Res. 89 (1998) 307–314.

[39] L.W. Seymour, Y. Miyamoto, H. Maeda, M. Brereton, J. Strohalm, K. Ulbrich, R.Duncan, Influence of molecular weight on passive tumour accumulation of asoluble macromolecular drug carrier, Eur. J. Cancer 31A (1995) 766–770.

[40] M. Sharpe, S.E. Easthope, G.M. Keating, H.M. Lamb, Polyethylene glycol-liposomal doxorubicin: a review of its use in the management of solid andhaematological malignancies and AIDS-related Kaposi's sarcoma, Drugs 62(2002) 2089–2126.

[41] P. Decuzzi, R. Pasqualini, W. Arap, M. Ferrari, Intravascular delivery of particu-late systems: does geometry really matter? Pharm. Res. 26 (2009) 235–243.

[42] M.E. Davis, Z.G. Chen, D.M. Shin, Nanoparticle therapeutics: an emerging treat-ment modality for cancer, Nat. Rev. Drug Discov. 7 (2008) 771–782.

[43] V.P. Torchilin, Micellar nanocarriers: pharmaceutical perspectives, Pharm. Res.24 (2007) 1–16.

[44] I. Brigger, C. Dubernet, P. Couvreur, Nanoparticles in cancer therapy and diagno-sis, Adv. Drug Deliv. Rev. 54 (2002) 631–651.

[45] S.P. Herbert, D.Y. Stainier, Molecular control of endothelial cell behaviour duringblood vessel morphogenesis, Nat. Rev. Mol. Cell Biol. 12 (2011) 551–564.

[46] E.E. Graves, A. Maity, Q.T. Le, The tumor microenvironment in non-small-celllung cancer, Semin. Radiat. Oncol. 20 (2010) 156–163.

[47] J. Fang, H. Nakamura, H. Maeda, The EPR effect: unique features of tumor bloodvessels for drug delivery, factors involved, and limitations and augmentation ofthe effect, Adv. Drug Deliv. Rev. 63 (2011) 136–151.

[48] J. Wu, T. Akaike, H. Maeda, Modulation of enhanced vascular permeability in tu-mors by a bradykinin antagonist, a cyclooxygenase inhibitor, and a nitric oxidescavenger, Cancer Res. 58 (1998) 159–165.

[49] A.K. Iyer, G. Khaled, J. Fang, H. Maeda, Exploiting the enhanced permeability andretention effect for tumor targeting, Drug Discov. Today 11 (2006) 812–818.

[50] H. Maeda, Vascular permeability in cancer and infection as related to macromo-lecular drug delivery, with emphasis on the EPR effect for tumor-selective drugtargeting, Proceedings of the Japan Academy, Series B, Physical and BiologicalSciences 88 (2012) 53–71.

[51] S. Van, S.K. Das, X. Wang, Z. Feng, Y. Jin, Z. Hou, F. Chen, A. Pham, N. Jiang,S.B. Howell, L. Yu, Synthesis, characterization, and biological evaluation ofpoly(L-gamma-glutamyl-glutamine)- paclitaxel nanoconjugate, Int. J. Nanomedicine5 (2010) 825–837.

[52] R. Duncan, M.J. Vicent, F. Greco, R.I. Nicholson, Polymer–drug conjugates: to-wards a novel approach for the treatment of endrocine-related cancer, Endocr.Relat. Cancer 12 (2005) S189–S199.

[53] J.M.MeerumTerwogt,W.W. ten Bokkel Huinink, J.H. Schellens,M. Schot, I.A.Mandjes,M.G. Zurlo, M. Rocchetti, H. Rosing, F.J. Koopman, J.H. Beijnen, Phase I clinical andpharmacokinetic study of PNU166945, a novel water-soluble polymer-conjugatedprodrug of paclitaxel, Anticancer Drugs 12 (2001) 315–323.

[54] F.M. Wachters, H.J. Groen, J.G. Maring, J.A. Gietema, M. Porro, H. Dumez, E.G. deVries, A.T. van Oosterom, A phase I study with MAG-camptothecin intravenouslyadministered weekly for 3 weeks in a 4-week cycle in adult patients with solidtumours, Br. J. Cancer 90 (2004) 2261–2267.

[55] N.E. Schoemaker, C. van Kesteren, H. Rosing, S. Jansen, M. Swart, J. Lieverst, D.Fraier, M. Breda, C. Pellizzoni, R. Spinelli, M. Grazia Porro, J.H. Beijnen, J.H.Schellens, W.W. ten Bokkel Huinink, A phase I and pharmacokinetic study ofMAG-CPT, a water-soluble polymer conjugate of camptothecin, Br. J. Cancer 87(2002) 608–614.

[56] N.M. Zaki, N. Tirelli, Gateways for the intracellular access of nanocarriers: a re-view of receptor-mediated endocytosis mechanisms and of strategies in recep-tor targeting, Expert Opin. Drug Deliv. 7 (2010) 895–913.

[57] M.L. Macheda, S. Rogers, J.D. Best, Molecular and cellular regulation of glucosetransporter (GLUT) proteins in cancer, J. Cell. Physiol. 202 (2005) 654–662.

[58] Z. Hu, F. Luo, Y. Pan, C. Hou, L. Ren, J. Chen, J. Wang, Y. Zhang, Arg-Gly-Asp (RGD)peptide conjugated poly(lactic acid)-poly(ethylene oxide) micelle for targeteddrug delivery, J. Biomed. Mater. Res. A 85 (2008) 797–807.

[59] Y. Zhang, M. Yang, N.G. Portney, D. Cui, G. Budak, E. Ozbay, M. Ozkan, C.S. Ozkan,Zeta potential: a surface electrical characteristic to probe the interaction ofnanoparticles with normal and cancer human breast epithelial cells, Biomed.Microdevices 10 (2008) 321–328.

[60] T. Yu, A. Malugin, H. Ghandehari, Impact of silica nanoparticle design on cellulartoxicity and hemolytic activity, ACS Nano 5 (2011) 5717–5728.

[61] D. Landesman-Milo, D. Peer, Altering the immune response with lipid-basednanoparticles, J. Control. Release 161 (2012) 600–608.

[62] M.A. Dobrovolskaia, S.E. McNeil, Immunological properties of engineerednanomaterials, Nat. Nanotechnol. 2 (2007) 469–478.

[63] A.L. Klibanov, K.Maruyama, V.P. Torchilin, L. Huang, Amphipathic polyethyleneglycolseffectively prolong the circulation time of liposomes, FEBS Lett. 268 (1990)235–237.

[64] D. Papahadjopoulos, T.M. Allen, A. Gabizon, E. Mayhew, K. Matthay, S.K. Huang,K.D. Lee, M.C. Woodle, D.D. Lasic, C. Redemann, Sterically stabilized liposomes:improvements in pharmacokinetics and antitumor therapeutic efficacy, Proc.Natl. Acad. Sci. U. S. A. 88 (1991) 11460–11464.

[65] J. Senior, C. Delgado, D. Fisher, C. Tilcock, G. Gregoriadis, Influence of surface hy-drophilicity of liposomes on their interaction with plasma protein and clearancefrom the circulation: studies with poly(ethylene glycol)-coated vesicles,Biochim. Biophys. Acta 1062 (1991) 77–82.

[66] J.H. Senior, Fate and behavior of liposomes in vivo: a review of controlling fac-tors, Crit. Rev. Ther. Drug Carrier Syst. 3 (1987) 123–193.

[67] V.P. Torchilin, V.G. Omelyanenko, M.I. Papisov, A.A. Bogdanov Jr., V.S.Trubetskoy, J.N. Herron, C.A. Gentry, Poly(ethylene glycol) on the liposome sur-face: on the mechanism of polymer-coated liposome longevity, Biochim.Biophys. Acta 1195 (1994) 11–20.

Page 9: Anticancer nanomedicine and tumor vascular permeability; Where is the missing link?

273S. Taurin et al. / Journal of Controlled Release 164 (2012) 265–275

[68] J.G. Shiah, M. Dvořák, P. Kopečková, Y. Sun, C.M. Peterson, J. Kopeček,Biodistribution and antitumour efficacy of long-circulating N-(2-hydroxypropyl)methacrylamide copolymer–doxorubicin conjugates in nude mice, Eur. J. Cancer37 (2001) 131–139.

[69] D.D. Lasic, F.J. Martin, A. Gabizon, S.K. Huang, D. Papahadjopoulos, Sterically sta-bilized liposomes: a hypothesis on the molecular origin of the extended circula-tion times, Biochim. Biophys. Acta 1070 (1991) 187–192.

[70] V. Dixit, J. Van den Bossche, D.M. Sherman, D.H. Thompson, R.P. Andres, Synthe-sis and grafting of thioctic acid−PEG−folate conjugates onto Au nanoparticlesfor selective targeting of folate receptor-positive tumor cells, Bioconjug. Chem.17 (2006) 603–609.

[71] N.C. Bellocq, S.H. Pun, G.S. Jensen, M.E. Davis, Transferrin-containing, cyclodex-trin polymer-based particles for tumor-targeted gene delivery, Bioconjug.Chem. 14 (2003) 1122–1132.

[72] M. Breunig, S. Bauer, A. Goepferich, Polymers and nanoparticles: intelligent toolsfor intracellular targeting? Eur. J. Pharm. Biopharm. 68 (2008) 112–128.

[73] B. Haley, E. Frenkel, Nanoparticles for drug delivery in cancer treatment, Urol.Oncol. Semin. Orig. Investig. 26 (2008) 57–64.

[74] K.N. Sugahara, T. Teesalu, P.P. Karmali, V.R. Kotamraju, L. Agemy, D.R.Greenwald, E. Ruoslahti, Coadministration of a tumor-penetrating peptide en-hances the efficacy of cancer drugs, Science 328 (2010) 1031–1035.

[75] R.K. Jain, T. Stylianopoulos, Delivering nanomedicine to solid tumors, Nat. Rev.Clin. Oncol. 7 (2010) 653–664.

[76] S. Goel, D.G. Duda, L. Xu, L.L. Munn, Y. Boucher, D. Fukumura, R.K. Jain, Normal-ization of the vasculature for treatment of cancer and other diseases, Physiol.Rev. 91 (2011) 1071–1121.

[77] M.D. Howard, A. Ponta, A. Eckman, M. Jay, Y. Bae, Polymer micelles withhydrazone-ester dual linkers for tunable release of dexamethasone, Pharm.Res. 28 (2011) 2435–2446.

[78] K. Greish, J. Fang, T. Inutsuka, A. Nagamitsu, H. Maeda, Macromolecular thera-peutics: advantages and prospects with special emphasis on solid tumourtargeting, Clin. Pharmacokinet. 42 (2003) 1089–1105.

[79] R. Duncan, The dawning era of polymer therapeutics, Nat. Rev. Drug Discov. 2(2003) 347–360.

[80] B. Mezquita, J. Mezquita, M. Pau, C. Mezquita, A novel intracellular isoform ofVEGFR-1 activates Src and promotes cell invasion in MDA-MB-231 breast cancercells, J. Cell. Biochem. 110 (2010) 732–742.

[81] T.H. Lee, S. Seng, M. Sekine, C. Hinton, Y. Fu, H.K. Avraham, S. Avraham,Vascular endothelial growth factor mediates intracrine survival in humanbreast carcinoma cells through internally expressed VEGFR1/FLT1, PLoSMed. 4 (2007) e186.

[82] A.V. Timoshenko, S. Rastogi, P.K. Lala, Migration-promoting role of VEGF-C andVEGF-C binding receptors in human breast cancer cells, Br. J. Cancer 97 (2007)1090–1098.

[83] A.V. Timoshenko, C. Chakraborty, G.F. Wagner, P.K. Lala, COX-2-mediated stim-ulation of the lymphangiogenic factor VEGF-C in human breast cancer, Br. J.Cancer 94 (2006) 1154–1163.

[84] P. Laakkonen, M. Waltari, T. Holopainen, T. Takahashi, B. Pytowski, P. Steiner, D.Hicklin, K. Persaud, J.R. Tonra, L. Witte, K. Alitalo, Vascular endothelial growthfactor receptor 3 is involved in tumor angiogenesis and growth, Cancer Res. 67(2007) 593–599.

[85] T.E. Fitzpatrick, G.E. Lash, A. Yanaihara, D.S. Charnock-Jones, S.K. Macdonald-Goodfellow, C.H. Graham, Inhibition of breast carcinoma and trophoblast cellinvasiveness by vascular endothelial growth factor, Exp. Cell Res. 283 (2003)247–255.

[86] M. Weigand, P. Hantel, R. Kreienberg, J. Waltenberger, Autocrine vascular endo-thelial growth factor signalling in breast cancer. Evidence from cell lines and pri-mary breast cancer cultures in vitro, Angiogenesis 8 (2005) 197–204.

[87] A. Hoeben, B. Landuyt, M.S. Highley, H. Wildiers, A.T. Van Oosterom, E.A. DeBruijn, Vascular endothelial growth factor and angiogenesis, Pharmacol. Rev.56 (2004) 549–580.

[88] L. Ryden, M. Stendahl, H. Jonsson, S. Emdin, N.O. Bengtsson, G. Landberg,Tumor-specific VEGF-A and VEGFR2 in postmenopausal breast cancer patientswith long-term follow-up. Implication of a link between VEGF pathway and ta-moxifen response, Breast Cancer Res. Treat. 89 (2005) 135–143.

[89] M. Schmidt, H.U. Voelker, M. Kapp, J. Dietl, U. Kammerer, Expression of VEGFR-1(Flt-1) in breast cancer is associated with VEGF expression and withnode-negative tumour stage, Anticancer. Res. 28 (2008) 1719–1724.

[90] N. Ravindranath, D. Wion, P. Brachet, D. Djakiew, Epidermal growth factor mod-ulates the expression of vascular endothelial growth factor in the human pros-tate, J. Androl. 22 (2001) 432–443.

[91] Y. Zeng, K. Opeskin, J. Goad, E.D. Williams, Tumor-induced activation oflymphatic endothelial cells via vascular endothelial growth factor receptor-2is critical for prostate cancer lymphatic metastasis, Cancer Res. 66 (2006)9566–9575.

[92] M. Li, H. Chen, L. Diao, Y. Zhang, C. Xia, F. Yang, Caveolin-1 and VEGF-C promotelymph node metastasis in the absence of intratumoral lymphangiogenesis innon-small cell lung cancer, Tumori 96 (2010) 734–743.

[93] R. Bianco, R. Rosa, V. Damiano, G. Daniele, T. Gelardi, S. Garofalo, V. Tarallo, S. DeFalco, D. Melisi, R. Benelli, A. Albini, A. Ryan, F. Ciardiello, G. Tortora, Vascular en-dothelial growth factor receptor-1 contributes to resistance to anti-epidermalgrowth factor receptor drugs in human cancer cells, Clin. Cancer Res. 14(2008) 5069–5080.

[94] Y. Takei, K. Kadomatsu, Y. Yuzawa, S. Matsuo, T. Muramatsu, A small interferingRNA targeting vascular endothelial growth factor as cancer therapeutics, CancerRes. 64 (2004) 3365–3370.

[95] J. Yang, H.F. Wu, L.X. Qian, W. Zhang, L.X. Hua, M.L. Yu, Z. Wang, Z.Q. Xu, Y.G. Sui,X.R. Wang, Increased expressions of vascular endothelial growth factor (VEGF),VEGF-C and VEGF receptor-3 in prostate cancer tissue are associated with tumorprogression, Asian J. Androl. 8 (2006) 169–175.

[96] N.R. Smith, D. Baker, N.H. James, K. Ratcliffe, M. Jenkins, S.E. Ashton, G. Sproat, R.Swann, N. Gray, A. Ryan, J.M. Jurgensmeier, C.Womack, Vascular endothelial growthfactor receptors VEGFR-2 and VEGFR-3 are localized primarily to the vasculature inhuman primary solid cancers, Clin. Cancer Res. 16 (2010) 3548–3561.

[97] N. Simiantonaki, C. Jayasinghe, R. Michel-Schmidt, K. Peters, M.I. Hermanns, C.J.Kirkpatrick, Hypoxia-induced epithelial VEGF-C/VEGFR-3 upregulation in carci-noma cell lines, Int. J. Oncol. 32 (2008) 585–592.

[98] M.P. Morelli, A.M. Brown, T.M. Pitts, J.J. Tentler, F. Ciardiello, A. Ryan, J.M.Jurgensmeier, S.G. Eckhardt, Targeting vascular endothelial growth factorreceptor-1 and -3 with cediranib (AZD2171): effects on migration and invasionof gastrointestinal cancer cell lines, Mol. Cancer Ther. 8 (2009) 2546–2558.

[99] T. Niki, S. Iba, M. Tokunou, T. Yamada, Y. Matsuno, S. Hirohashi, Expression of vas-cular endothelial growth factors A, B, C, and D and their relationships to lymphnode status in lung adenocarcinoma, Clin. Cancer Res. 6 (2000) 2431–2439.

[100] E. Martinelli, T. Troiani, F. Morgillo, G. Rodolico, D. Vitagliano, M.P. Morelli, C.Tuccillo, L. Vecchione, A. Capasso, M. Orditura, F. De Vita, S.G. Eckhardt, M.Santoro, L. Berrino, F. Ciardiello, Synergistic antitumor activity of sorafenib incombination with epidermal growth factor receptor inhibitors in colorectaland lung cancer cells, Clin. Cancer Res. 16 (2010) 4990–5001.

[101] V.K. Anagnostou, D.G. Tiniakos, M. Fotinou, A. Achimastos, K.N. Syrigos,Multiplexed analysis of angiogenesis and lymphangiogenesis factors predicts out-come for non-small cell lung cancer patients, Virchows Arch. 458 (2011) 331–340.

[102] E. Carrillo de Santa Pau, F.C. Arias, E. Caso Pelaez, G.M. Munoz Molina, I. SanchezHernandez, I. Muguruza Trueba, R. Moreno Balsalobre, S. Sacristan Lopez, A.Gomez Pinillos, M. del Val Toledo Lobo, Prognostic significance of the expressionof vascular endothelial growth factors A, B, C, and D and their receptors R1, R2,and R3 in patients with nonsmall cell lung cancer, Cancer 115 (2009) 1701–1712.

[103] E. Zygalaki, E.G. Tsaroucha, L. Kaklamanis, E.S. Lianidou, Quantitative real-timereverse transcription PCR study of the expression of vascular endothelial growthfactor (VEGF) splice variants and VEGF receptors (VEGFR-1 and VEGFR-2) innon small cell lung cancer, Clin. Chem. 53 (2007) 1433–1439.

[104] Y.H. Bae, K. Park, Targeted drug delivery to tumors: myths, reality and possibility,J. Control. Release 153 (2011) 198–205.

[105] R. Kedmi, N. Ben-Arie, D. Peer, The systemic toxicity of positively charged lipidnanoparticles and the role of Toll-like receptor 4 in immune activation, Bioma-terials 31 (2010) 6867–6875.

[106] J.M. Anderson, A. Rodriguez, D.T. Chang, Foreign body reaction to biomaterials,Semin. Immunol. 20 (2008) 86–100.

[107] S. Mukhopadhyay, A.A. Gal, Granulomatous lung disease: an approach to the dif-ferential diagnosis, Arch. Pathol. Lab. Med. 134 (2010) 667–690.

[108] L.A.J. O'Neill, How frustration leads to inflammation, Science 320 (2008) 619–620.[109] J. Panyam, V. Labhasetwar, Sustained cytoplasmic delivery of drugswith intracellu-

lar receptors using biodegradable nanoparticles, Mol. Pharm. 1 (2004) 77–84.[110] T. Etrych, L. Kovář, J. Strohalm, P. Chytil, B. Říhová, K. Ulbrich, Biodegradable star

HPMA polymer–drug conjugates: Biodegradability, distribution and anti-tumorefficacy, J. Control. Release 154 (2011) 241–248.

[111] M. Dvořák, P. Kopečková, J. Kopeček, High-molecular weight HPMA copolymer–adriamycin conjugates, J. Control. Release 60 (1999) 321–332.

[112] J.L. Yu, J.W. Rak, Host microenvironment in breast cancer development: inflam-matory and immune cells in tumour angiogenesis and arteriogenesis, BreastCancer Res. 5 (2003) 83–88.

[113] J.W. Pollard, Macrophages define the invasive microenvironment in breast can-cer, J. Leukoc. Biol. 84 (2008) 623–630.

[114] T.M. Robinson-Smith, I. Isaacsohn, C.A. Mercer, M. Zhou, N. Van Rooijen, N.Husseinzadeh, M.M. McFarland-Mancini, A.F. Drew, Macrophages mediateinflammation-enhanced metastasis of ovarian tumors in mice, Cancer Res. 67(2007) 5708–5716.

[115] L.W. Seymour, D.R. Ferry, D. Anderson, S. Hesslewood, P.J. Julyan, R. Poyner, J.Doran, A.M. Young, S. Burtles, D.J. Kerr, f.t.C.R.C.P.I.I.C.T. Committee, Hepaticdrug targeting: Phase I evaluation of polymer-bound doxorubicin, J. Clin.Oncol. 20 (2002) 1668–1676.

[116] Z.M. Qian, H. Li, H. Sun, K. Ho, Targeted drug delivery via the transferrinreceptor-mediated endocytosis pathway, Pharmacol. Rev. 54 (2002) 561–587.

[117] S. Wang, P.S. Low, Folate-mediated targeting of antineoplastic drugs, imagingagents, and nucleic acids to cancer cells, J. Control. Release 53 (1998) 39–48.

[118] T. Lammers, F. Kiessling, W.E. Hennink, G. Storm, Drug targeting to tumors: princi-ples, pitfalls and (pre-) clinical progress, J. Control. Release 161 (2012) 175–187.

[119] A. Garcia-Bennett, M. Nees, B. Fadeel, In search of the Holy Grail: folate-targetednanoparticles for cancer therapy, Biochem. Pharmacol. 81 (2011) 976–984.

[120] H.T. Winer-Muram, S.G. Jennings, R.D. Tarver, A.M. Aisen, M. Tann, D.J. Conces,C.A. Meyer, Volumetric growth rate of stage I lung cancer prior to treatment: se-rial CT scanning, Radiology 223 (2002) 798–805.

[121] K. Wechsler-Jentzsch, J.H. Witt, C.R. Fitz, D.C. McCullough, L. Harisiadis,Unresectable gliomas in children: tumor-volume response to radiation therapy,Radiology 169 (1988) 237–242.

[122] T. Kuroishi, S. Tominaga, T. Morimoto, H. Tashiro, S. Itoh, H.Watanabe,M. Fukuda, J.Ota, T. Horino, T. Ishida, Tumor growth rate and prognosis of breast cancer mainlydetected by mass screening, Jpn. J. Cancer Res. 81 (1990) 454–462.

[123] G.M. Zharinov, V.A. Gushchin, The rate of tumor growth and cell loss in cervicalcancer, Vopr. Onkol. 35 (1989) 21–25.

[124] E. Strom, J.L. Larsen, Colon cancer at barium enema examination and colonoscopy:a study from the county of Hordaland, Norway, Radiology 211 (1999) 211–214.

Page 10: Anticancer nanomedicine and tumor vascular permeability; Where is the missing link?

274 S. Taurin et al. / Journal of Controlled Release 164 (2012) 265–275

[125] S. Bolin, E. Nilsson, R. Sjodahl, Carcinoma of the colon and rectum-growth rate,Ann. Surg. 198 (1983) 151–158.

[126] J.A. Carlson, Tumor doubling time of cutaneous melanoma and its metastasis,Am. J. Dermatopathol. 25 (2003) 291–299.

[127] M. Yoshino, Growth kinetics of hepatocellular carcinoma, Jpn. J. Clin. Oncol. 13(1983) 45–52.

[128] T. Nakajima, M. Moriguchi, Y. Mitsumoto, T. Katagishi, H. Kimura, H. Shintani,T. Deguchi, T. Okanoue, K. Kagawa, T. Ashihara, Simple tumor profile chartbased on cell kinetic parameters and histologic grade is useful for estimatingthe natural growth rate of hepatocellular carcinoma, Hum. Pathol. 33 (2002)92–99.

[129] Y. Saito, Y. Matsuzaki, M. Doi, T. Sugitani, T. Chiba, M. Abei, J. Shoda, N.Tanaka, Multiple regression analysis for assessing the growth of small hepa-tocellular carcinoma: the MIB-1 labeling index is the most effective parame-ter, J. Gastroenterol. 33 (1998) 229–235.

[130] S. Nakasu, Y. Nakasu, M. Nakajima, M. Yokoyama, M. Matsuda, J. Handa, Poten-tial doubling time and tumour doubling time in meningiomas and neurinomas,Acta Neurochir. (Wien) 138 (1996) 763–770.

[131] M.R. Paling, T.H. Shawker, Abdominal ultrasound in advanced ovarian carcino-ma, J. Clin. Ultrasound 9 (1981) 435–441.

[132] H. Furukawa, R. Iwata, N. Moriyama, Growth rate of pancreatic adenocarcinoma:initial clinical experience, Pancreas 22 (2001) 366–369.

[133] Y. Tanaka, K. Hongo, T. Tada, K. Sakai, Y. Kakizawa, S. Kobayashi, Growth patternand rate in residual nonfunctioning pituitary adenomas: correlations amongtumor volume doubling time, patient age, and MIB-1 index, J. Neurosurg. 98(2003) 359–365.

[134] H.P. Schmid, J.E. McNeal, T.A. Stamey, Observations on the doubling time of pros-tate cancer. The use of serial prostate-specific antigen in patients with untreateddisease as a measure of increasing cancer volume, Cancer 71 (1993) 2031–2040.

[135] S. Ozono, N. Miyao, T. Igarashi, K. Marumo, H. Nakazawa, M. Fukuda, T.Tsushima, N. Tokuda, J. Kawamura, M. Murai, Tumor doubling time of renalcell carcinoma measured by CT: collaboration of Japanese Society of Renal Can-cer, Jpn. J. Clin. Oncol. 34 (2004) 82–85.

[136] T. Arai, T. Kuroishi, Y. Saito, Y. Kurita, T. Naruke, M. Kaneko, Tumor doubling timeand prognosis in lung cancer patients: evaluation from chest films and clinicalfollow-up study. Japanese Lung Cancer Screening Research Group, Jpn. J. Clin.Oncol. 24 (1994) 199–204.

[137] Y. Takahashi, M. Mai, S. Kusama, Factors influencing growth rate of recurrentstomach cancers as determined by analysis of serum carcinoembryonic antigen,Cancer 75 (1995) 1497–1502.

[138] F.G. Blankenberg, R.L. Teplitz, W. Ellis, M.S. Salamat, B.H. Min, L. Hall, D.B.Boothroyd, I.M. Johnstone, D.R. Enzmann, The influence of volumetric tumordoubling time, DNA ploidy, and histologic grade on the survival of patientswith intracranial astrocytomas, AJNR Am. J. Neuroradiol. 16 (1995) 1001–1012.

[139] K. Usuda, Y. Saito, M. Sagawa, M. Sato, K. Kanma, S. Takahashi, C. Endo, Y. Chen,A. Sakurada, S. Fujimura, Tumor doubling time and prognostic assessment of pa-tients with primary lung cancer, Cancer 74 (1994) 2239–2244.

[140] K. Tanaka, H. Shimada, M. Miura, Y. Fujii, S. Yamaguchi, I. Endo, H. Sekido, S.Togo, H. Ike, Metastatic tumor doubling time: most important prehepatectomypredictor of survival and nonrecurrence of hepatic colorectal cancer metastasis,World J. Surg. 28 (2004) 263–270.

[141] N. Okazaki, M. Yoshino, T. Yoshida, M. Suzuki, N. Moriyama, K. Takayasu, M.Makuuchi, S. Yamazaki, H. Hasegawa, M. Noguchi, et al., Evaluation of the prog-nosis for small hepatocellular carcinoma based on tumor volume doubling time.A preliminary report, Cancer 63 (1989) 2207–2210.

[142] W. Weiss, Tumor doubling time and survival of men with bronchogenic carcino-ma, Chest 65 (1974) 3–8.

[143] E.P. Malaise, N. Chavaudra, A. Charbit, M. Tubiana, Relationship between thegrowth rate of human metastases, survival and pathological type, Eur. J. Cancer10 (1974) 451–459.

[144] M.C. Garnett, P. Kallinteri, Nanomedicines and nanotoxicology: some physiolog-ical principles, Occup. Med. (Lond.) 56 (2006) 307–311.

[145] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, Cell 144(2011) 646–674.

[146] R. Pasqualini, W. Arap, D.M. McDonald, Probing the structural and molecular di-versity of tumor vasculature, Trends Mol. Med. 8 (2002) 563–571.

[147] J.A. Nagy, S.H. Chang, S.C. Shih, A.M. Dvorak, H.F. Dvorak, Heterogeneity of thetumor vasculature, Semin. Thromb. Hemost. 36 (2010) 321–331.

[148] I.O. Ng, R.T. Poon, J.M. Lee, S.T. Fan, M. Ng, W.K. Tso, Microvessel density, vascu-lar endothelial growth factor and its receptors Flt-1 and Flk-1/KDR in hepatocel-lular carcinoma, Am. J. Clin. Pathol. 116 (2001) 838–845.

[149] F. Dunphy, B.C. Stack Jr., J.H. Boyd, T.L. Dunleavy, H.J. Kim, C.H. Dunphy,Microvessel density in advanced head and neck squamous cell carcinoma beforeand after chemotherapy, Anticancer. Res. 22 (2002) 1755–1758.

[150] H.C. Hollingsworth, E.C. Kohn, S.M. Steinberg, M.L. Rothenberg, M.J. Merino,Tumor angiogenesis in advanced stage ovarian carcinoma, Am. J. Pathol. 147(1995) 33–41.

[151] A. Yuan, P.C. Yang, C.J. Yu, Y.C. Lee, Y.T. Yao, C.L. Chen, L.N. Lee, S.H. Kuo, K.T. Luh, Tumorangiogenesis correlates with histologic type and metastasis in non-small-cell lungcancer, Am. J. Respir. Crit. Care Med. 152 (1995) 2157–2162.

[152] S.G. Sharma, N. Aggarwal, S.D. Gupta, M.K. Singh, R. Gupta, A.K. Dinda, Angiogen-esis in renal cell carcinoma: correlation of microvessel density and microvesselarea with other prognostic factors, Int. Urol. Nephrol. 43 (2011) 125–129.

[153] S. Bosari, A.K. Lee, R.A. DeLellis, B.D. Wiley, G.J. Heatley, M.L. Silverman,Microvessel quantitation and prognosis in invasive breast carcinoma, Hum.Pathol. 23 (1992) 755–761.

[154] N. Weidner, J.P. Semple, W.R. Welch, J. Folkman, Tumor angiogenesis and metas-tasis–correlation in invasive breast carcinoma, N. Engl. J. Med. 324 (1991) 1–8.

[155] C.H. Graham, J. Rivers, R.S. Kerbel, K.S. Stankiewicz, W.L. White, Extent of vascu-larization as a prognostic indicator in thin (b 0.76 mm) malignant melanomas,Am. J. Pathol. 145 (1994) 510–514.

[156] N. Weidner, P.R. Carroll, J. Flax, W. Blumenfeld, J. Folkman, Tumor angiogenesiscorrelates with metastasis in invasive prostate carcinoma, Am. J. Pathol. 143(1993) 401–409.

[157] O. Abulafia, W.E. Triest, D.M. Sherer, C.C. Hansen, F. Ghezzi, Angiogenesis in en-dometrial hyperplasia and stage I endometrial carcinoma, Obstet. Gynecol. 86(1995) 479–485.

[158] P.B. Vermeulen, D. Verhoeven, G. Hubens, E. Van Marck, G. Goovaerts, M.Huyghe, E.A. De Bruijn, A.T. Van Oosterom, L.Y. Dirix, Microvessel density, endo-thelial cell proliferation and tumour cell proliferation in human colorectal ade-nocarcinomas, Ann. Oncol. 6 (1995) 59–64.

[159] S. Svagzdys, V. Lesauskaite, D. Pavalkis, I. Nedzelskiene, D. Pranys, A. Tamelis,Microvessel density as new prognostic marker after radiotherapy in rectal can-cer, BMC Cancer 9 (2009) 95.

[160] S. Behrem, K. Zarkovic, N. Eskinja, N. Jonjic, Endoglin is a better marker than CD31in evaluation of angiogenesis in glioblastoma, Croat. Med. J. 46 (2005) 417–422.

[161] S.C. Piscitelli, K.A. Rodvold, D.A. Rushing, D.A. Tewksbury, Pharmacokinetics andpharmacodynamics of doxorubicin in patients with small cell lung cancer, Clin.Pharmacol. Ther. 53 (1993) 555–561.

[162] M. Longmire, P.L. Choyke, H. Kobayashi, Clearance properties of nano-sized parti-cles and molecules as imaging agents: considerations and caveats, Nanomedicine(Lond.) 3 (2008) 703–717.

[163] S.K. Hobbs, W.L. Monsky, F. Yuan, W.G. Roberts, L. Griffith, V.P. Torchilin, R.K.Jain, Regulation of transport pathways in tumor vessels: role of tumor typeand microenvironment, Proc. Natl. Acad. Sci. U. S. A. 95 (1998) 4607–4612.

[164] T. Horn, J.H. Henriksen, P. Christoffersen, The sinusoidal lining cells in “normal”human liver. A scanning electronmicroscopic investigation, Liver 6 (1986) 98–110.

[165] H. Uchino, Y. Matsumura, T. Negishi, F. Koizumi, T. Hayashi, T. Honda, N.Nishiyama, K. Kataoka, S. Naito, T. Kakizoe, Cisplatin-incorporating polymericmicelles (NC-6004) can reduce nephrotoxicity and neurotoxicity of cisplatin inrats, Br. J. Cancer 93 (2005) 678–687.

[166] P.E. Colombo, M. Boustta, S. Poujol, F. Pinguet, P. Rouanet, F. Bressolle, M. Vert,Biodistribution of doxorubicin-alkylated poly(L-lysine citramide imide) conju-gates in an experimental model of peritoneal carcinomatosis after intraperito-neal administration, Eur. J. Pharm. Sci. 31 (2007) 43–52.

[167] C.D. Conover, R.B. Greenwald, A. Pendri, C.W. Gilbert, K.L. Shum, Camptothecindelivery systems: enhanced efficacy and tumor accumulation of camptothecinfollowing its conjugation to polyethylene glycol via a glycine linker, CancerChemother. Pharmacol. 42 (1998) 407–414.

[168] M. Harada, J. Murata, Y. Sakamura, H. Sakakibara, S. Okuno, T. Suzuki, Carrier anddose effects on the pharmacokinetics of T-0128, a camptothecin analogue-carboxymethyl dextran conjugate, in non-tumor- and tumor-bearing rats, J.Control. Release 71 (2001) 71–86.

[169] C. Li, R.A. Newman, Q.P. Wu, S. Ke, W. Chen, T. Hutto, Z. Kan, M.D. Brannan, C.Charnsangavej, S. Wallace, Biodistribution of paclitaxel and poly(L-glutamicacid)-paclitaxel conjugate in mice with ovarian OCa-1 tumor, CancerChemother. Pharmacol. 46 (2000) 416–422.

[170] T. Schluep, J. Cheng, K.T. Khin, M.E. Davis, Pharmacokinetics and biodistributionof the camptothecin-polymer conjugate IT-101 in rats and tumor-bearing mice,Cancer Chemother. Pharmacol. 57 (2006) 654–662.

[171] J.G. Shiah, M. Dvorak, P. Kopeckova, Y. Sun, C.M. Peterson, J. Kopecek,Biodistribution and antitumour efficacy of long-circulating N-(2-hydroxypropyl)methacrylamide copolymer-doxorubicin conjugates in nude mice, Eur. J. Cancer37 (2001) 131–139.

[172] R. Song, Y. Joo Jun, J. Ik Kim, C. Jin, Y.S. Sohn, Synthesis, characterization, andtumor selectivity of a polyphosphazene-platinum(II) conjugate, J. Control. Re-lease 105 (2005) 142–150.

[173] A. Sparreboom, C.D. Scripture, V. Trieu, P.J. Williams, T. De, A. Yang, B. Beals, W.D.Figg, M. Hawkins, N. Desai, Comparative preclinical and clinical pharmacokinet-ics of a cremophor-free, nanoparticle albumin-bound paclitaxel (ABI-007) andpaclitaxel formulated in Cremophor (Taxol), Clin. Cancer Res. 11 (2005)4136–4143.

[174] S. Sugahara, M. Kajiki, H. Kuriyama, T.R. Kobayashi, Complete regression ofxenografted human carcinomas by a paclitaxel-carboxymethyl dextran conju-gate (AZ10992), J. Control. Release 117 (2007) 40–50.

[175] X. Wang, G. Zhao, S. Van, N. Jiang, L. Yu, D. Vera, S.B. Howell, Pharmacokineticsand tissue distribution of PGG-paclitaxel, a novel macromolecular formulationof paclitaxel, in nu/nu mice bearing NCI-460 lung cancer xenografts, CancerChemother. Pharmacol. 65 (2010) 515–526.

[176] Y. Bae, N. Nishiyama, S. Fukushima, H. Koyama, M. Yasuhiro, K. Kataoka, Prepa-ration and biological characterization of polymeric micelle drug carriers with in-tracellular pH-triggered drug release property: tumor permeability, controlledsubcellular drug distribution, and enhanced in vivo antitumor efficacy,Bioconjug. Chem. 16 (2005) 122–130.

[177] H. Cabral, N. Nishiyama, K. Kataoka, Optimization of (1,2-diamino-cyclohexane)platinum(II)-loaded polymeric micelles directed to improved tumor targetingand enhanced antitumor activity, J. Control. Release 121 (2007) 146–155.

[178] A.A. Gabizon, Liposome circulation time and tumor targeting: implications forcancer chemotherapy, Adv. Drug Deliv. Rev. 16 (1995) 285–294.

[179] M.L. Immordino, P. Brusa, S. Arpicco, B. Stella, F. Dosio, L. Cattel, Preparation,characterization, cytotoxicity and pharmacokinetics of liposomes containingdocetaxel, J. Control. Release 91 (2003) 417–429.

Page 11: Anticancer nanomedicine and tumor vascular permeability; Where is the missing link?

275S. Taurin et al. / Journal of Controlled Release 164 (2012) 265–275

[180] R. Jukanti, G. Devraj, A.S. Shashank, R. Devraj, Biodistribution of ascorbyl pal-mitate loaded doxorubicin pegylated liposomes in solid tumor bearing mice,J. Microencapsul. 28 (2011) 142–149.

[181] H.J. Lim, D. Masin, N.L. McIntosh, T.D. Madden, M.B. Bally, Role of drug release andliposome-mediated drug delivery in governing the therapeutic activity of liposomalmitoxantrone used to treat human A431 and LS180 solid tumors, J. Pharmacol. Exp.Ther. 292 (2000) 337–345.

[182] D.E. Lopes de Menezes, N. Hudon, N. McIntosh, L.D. Mayer, Molecular and phar-macokinetic properties associated with the therapeutics of bcl-2 antisense oli-gonucleotide G3139 combined with free and liposomal doxorubicin, Clin.Cancer Res. 6 (2000) 2891–2902.

[183] M.S. Newman, G.T. Colbern, P.K. Working, C. Engbers, M.A. Amantea, Comparativepharmacokinetics, tissue distribution, and therapeutic effectiveness of cisplatin en-capsulated in long-circulating, pegylated liposomes (SPI-077) in tumor-bearingmice, Cancer Chemother. Pharmacol. 43 (1999) 1–7.

[184] N. Nishiyama, S. Okazaki, H. Cabral, M. Miyamoto, Y. Kato, Y. Sugiyama, K. Nishio,Y. Matsumura, K. Kataoka, Novel cisplatin-incorporated polymeric micelles caneradicate solid tumors in mice, Cancer Res. 63 (2003) 8977–8983.

[185] S. Unezaki, K. Maruyama, O. Ishida, A. Suginaka, J.-i. Hosoda, M. Iwatsuru, En-hanced tumor targeting and improved antitumor activity of doxorubicin bylong-circulating liposomes containing amphipathic poly(ethylene glycol), Int.J. Pharm. 126 (1995) 41–48.

[186] J. Vaage, E. Barbera-Guillem, R. Abra, A. Huang, P. Working, Tissue distributionand therapeutic effect of intravenous free or encapsulated liposomal doxorubi-cin on human prostate carcinoma xenografts, Cancer 73 (1994) 1478–1484.

[187] X.B. Xiong, Y. Huang, W.L. Lu, X. Zhang, H. Zhang, T. Nagai, Q. Zhang, Enhancedintracellular delivery and improved antitumor efficacy of doxorubicin by steri-cally stabilized liposomes modified with a synthetic RGD mimetic, J. Control. Re-lease 107 (2005) 262–275.

[188] M. Yokoyama, T. Okano, Y. Sakurai, S. Fukushima, K. Okamoto, K. Kataoka, Selec-tive delivery of adriamycin to a solid tumor using a polymeric micelle carriersystem, J. Drug Target. 7 (1999) 171–186.

[189] H.S. Yoo, T.G. Park, Folate receptor targeted biodegradable polymeric doxorubi-cin micelles, J. Control. Release 96 (2004) 273–283.

[190] J.Q. Zhang, Z.R. Zhang, H. Yang, Q.Y. Tan, S.R. Qin, X.L. Qiu, Lyophilized paclitaxelmagnetoliposomes as a potential drug delivery system for breast carcinoma viaparenteral administration: in vitro and in vivo studies, Pharm. Res. 22 (2005)573–583.

[191] K.M. Laginha, S. Verwoert, G.J. Charrois, T.M. Allen, Determination of doxorubi-cin levels in whole tumor and tumor nuclei in murine breast cancer tumors,Clin. Cancer Res. 11 (2005) 6944–6949.

[192] C.L. Peng, P.S. Lai, F.H. Lin, S. Yueh-Hsiu Wu, M.J. Shieh, Dual chemotherapy andphotodynamic therapy in an HT-29 human colon cancer xenograft model usingSN-38-loaded chlorin-core star block copolymer micelles, Biomaterials 30(2009) 3614–3625.

[193] M. Rafi, H. Cabral, M.R. Kano, P. Mi, C. Iwata, M. Yashiro, K. Hirakawa, K.Miyazono, N. Nishiyama, K. Kataoka, Polymeric micelles incorporating(1,2-diaminocyclohexane) platinum (II) suppress the growth of orthotopic scir-rhous gastric tumors and their lymph node metastasis, J. Control. Release 159(2012) 189–196.

[194] Y. Saito, M. Yasunaga, J. Kuroda, Y. Koga, Y. Matsumura, Antitumour activity ofNK012, SN-38-incorporating polymeric micelles, in hypovascular orthotopicpancreatic tumour, Eur. J. Cancer 46 (2010) 650–658.

[195] A. Takahashi, N. Ohkohchi, M. Yasunaga, J. Kuroda, Y. Koga, H. Kenmotsu, T.Kinoshita, Y. Matsumura, Detailed distribution of NK012, an SN-38-incorporatingmicelle, in the liver and its potent antitumor effects in mice bearing liver metasta-ses, Clin. Cancer Res. 16 (2010) 4822–4831.


Recommended