You are on page 1of 14

Journal of Controlled Release 244 (2016) 108–121

Contents lists available at ScienceDirect

Journal of Controlled Release

journal homepage: www.elsevier.com/locate/jconrel

Review article

To exploit the tumor microenvironment: Since the EPR effect fails in the
clinic, what is the future of nanomedicine?
F. Danhier
Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73 bte B1 73.12, 1200 Brussels, Belgium

a r t i c l e i n f o a b s t r a c t

Article history: Tumor targeting by nanomedicine-based therapeutics has emerged as a promising approach to overcome the
Received 19 September 2016 lack of specificity of conventional chemotherapeutic agents and to provide clinicians the ability to overcome
Received in revised form 26 October 2016 shortcomings of current cancer treatment. The major underlying mechanism of the design of nanomedicines
Accepted 7 November 2016
was the Enhanced Permeability and Retention (EPR) effect, considered as the “royal gate” in the drug delivery
Available online 18 November 2016
field. However, after the publication of thousands of research papers, the verdict has been handed down: the
Keywords:
EPR effect works in rodents but not in humans! Thus the basic rationale of the design and development of
EPR effect nanomedicines in cancer therapy is failing making it necessary to stop claiming efficacy gains via the EPR effect,
Tumor targeting while tumor targeting cannot be proved in the clinic. It is probably time to dethrone the EPR effect and to ask the
Nanomedicine question: what is the future of nanomedicines without the EPR effect? The aim of this review is to provide a gen-
Drug delivery eral overview on (i) the current state of the EPR effect, (ii) the future of nanomedicine and (iii) the strategies of
Cancer modulation of the tumor microenvironment to improve the delivery of nanomedicine.
© 2016 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
2. Tumor targeting. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
2.1. Passive targeting: Background of the EPR effect . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
2.2. Active targeting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
2.3. Clinically used nanomedicines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
3. Heterogeneity in the EPR effect: a new clinically relevant EPR effect model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112
3.1. The heterogeneity or lack of fenestrations in the tumor endothelium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112
3.2. Hypoxic areas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112
3.3. The lower and heterogeneous pericyte coverage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112
3.4. Heterogeneous basement membrane . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
3.5. The higher and heterogeneous density of the ECM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
4. What is the future for nanomedicines without EPR effect?. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 114
4.1. Toxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 114
4.2. Local delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 114
4.3. Nanomedicines as a tool for imaging the tumor microenvironment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 114
5. Strategies to improve delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116
5.1. Vascular mediators involved in the EPR effect . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116
5.2. Microenvironment modifications/modulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
5.2.1. Direct permeability enhancement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
5.2.2. Indirect permeability enhancement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
5.3. Multifunctional nanomedicines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
5.3.1. Tumor-penetrating peptides. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
5.3.2. Stimuli-responsive nanomedicine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119

E-mail address: fabienne.danhier@uclouvain.be.

http://dx.doi.org/10.1016/j.jconrel.2016.11.015
0168-3659/© 2016 Elsevier B.V. All rights reserved.
F. Danhier / Journal of Controlled Release 244 (2016) 108–121 109

6. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119

1. Introduction hypertension. This uniformly elevated interstitial fluid pressure (IFP)


reduces convective transport, while the dense extracellular matrix hin-
Tumor targeting by nanomedicine-based therapeutics has emerged ders diffusion [2].
as a promising approach to overcome the lack of specificity of conven- In 1979, Maeda et al. reported the first synthesis of the anticancer
tional chemotherapeutic agents and to provide clinicians the ability to protein neocarzinostatin (NCS) conjugated with a polymer (styrene
overcome shortcomings of current cancer treatment [1–3]. Drug deliv- maleic acid copolymer, SMA) which has been called SMANCS [12]. In
ery systems can be used to specifically target tumors and improve the 1986, SMANCS was found to accumulate to a greater degree than NCS
therapeutic index and the pharmacokinetic profile of anticancer drugs. in tumor tissues. Later, other plasma proteins, larger than 40 kDa,
Nanomedicines are submicrometer-sized carrier materials which in- showed selective tumor accumulation. These findings led to generaliz-
tend to improve the biodistribution of systemically administered che- ing the concept of the EPR effect [10]. Typically, the EPR effect was illus-
motherapeutic drugs. Nanoformulation aims at improving the balance trated using an intravenous injection of the dye Evans blue, which binds
between the efficacy and the toxicity of systemic chemotherapeutic to plasma albumin, forming a macromolecule model. 24 h after injec-
agent interventions [4]. Among others, the main examples of tion, the dye was found in tumor tissue and not in normal tissues [13].
nanomedicine formulations are liposomes [5], micelles [6], nanoparti- Scintigraphy of 67Ga complexed to transferrin (90 kDA) also provides
cles [7] and polymer-drug conjugates [8]. a demonstration of the EPR effect because of the preferential tumor ac-
In 2010, we wrote a review called “to exploit the tumor microenvi- cumulation of complexes [11].
ronment: passive and active tumor targeting of nanocarriers for anti-
cancer drug delivery” [9]. At this time, the particular tumor targeting 2.2. Active targeting
nanomedicines was in the spotlight since nanomedicine–based thera-
pies kept the promise to overcome the lack of specificity of conventional On the contrary of passive drug targeting, active drug targeting relies
chemotherapeutic agents. The major underlying mechanism of the de- on the use on specific ligands (antibodies or peptides) that can be grafted
sign of nanomedicines was the Enhanced Permeability and Retention to the nanomedicine surface and specifically bind to overexpressed recep-
(EPR) effect, considered as the “royal gate” in the drug delivery field tors at the target site [4]. In the active targeting strategy, two cellular tar-
since drug-loaded nanocarriers could enter and accumulate inside a gets can be distinguished: (i) the targeting of cancer cells (overexpression
tumor and reduce the side effects associated with conventional treat- of transferrin, folate, Epidermal Growth Factor Receptor or Glycoproteins
ments. This recycled principle of the “magic bullet” first imagined by receptors) and (ii) the targeting of the tumor endothelium (overexpres-
P. Ehrlich in 1906 was then really promising. After the publication of sion of the Vascular Endothelial Growth Factors (VEGF), αvβ3 integrins,
thousands of research papers, the verdict has been handed down: the the Vascular Cell Adhesion Molecule-1 (VCAM-1) or matrix metallopro-
EPR effect works in rodents but not in humans! Now the question is: teinases (MMP's) (Fig. 1B). Interestingly, some targets can be
what is the future of nanomedicines? overexpressed in both endothelial and cancer cells [9]. The active
In this review, I would like to highlight other possibilities that targeting strategy promised to target the cancer tissue more specifically
nanomedicines can still offer in cancer therapy even if the basic concept than with the EPR effect alone. We know now that actively targeted nano-
of EPR effect is compromised. Several reviews are available for each sec- particles are internalized via the receptor-mediated endocytosis. The in-
tions and it would go beyond the scope of this review to discuss these creased efficacy of these systems is thus due to an improved target cell
issues in detail. Rather, the aim of this review is to propose a general recognition and target cell uptake rather than to a better tumor accumu-
overview of the nanomedicine-related issues that could be addressed lation [4,14].
and to discuss for which strategy or advantage nanomedicines are
promising. These strategies should be particularly taken into account
2.3. Clinically used nanomedicines
in the future and must be considered from their early conception to clin-
ical translation.
As demonstrated by the significant nanomedicine market, the applica-
tion of nanotechnology in the field of medicine has the potential to im-
2. Tumor targeting prove the treatment of cancer [16] and several nanomedicines have
been approved by the US Food and Drug Administration (FDA) and Euro-
2.1. Passive targeting: Background of the EPR effect pean Medicines Agencies (EMA) for cancer therapy [17]. Table 1 shows an
exhaustive list of nanomedicines for cancer treatment, which are either
The EPR effect was first termed by Maeda in 1986. The fundamental approved or in clinical trials. In this table, nanomedicines are classified
features of EPR physiology are hyperpermeable tumor vasculature by nanocarrier (micelles, nanoparticles, polymer drug conjugate and lipo-
allowing the enhanced permeability of large particles—including pro- somes). The description of all these systems is beyond the scope of this re-
teins, macromolecules, liposomes, micelles and other soluble particles. view, however, some nanomedicines are discussed hereunder.
These were large enough to avoid renal clearance into the interstitial The growing presence of novel nanomedicines raises the question of
space of the tumor combined with impaired lymphatic drainage limit- whether approved nanomedicines really form nanoparticles; i.e. are im-
ing the clearance of tumor particles and causing enhanced retention of provements we observed from nanomedicines structure-related or do
those same particles (Fig. 1A) [10]. Indeed, nanocarriers (20–200 nm they result from improved formulations? Do we even have sufficient
in diameter) can extravasate and accumulate within the interstitial data to address this question? Regarding the large number of
space of tumors by transiting through the much larger endothelial nanomedicines on the market and in clinical trials, we cannot expect
pores (10 to 1000 nm in diameter). The absence of functional lymphatic to find a “one size fits all” answer.
vessels in most tumors contributes to nanocarrier entrapment and re- The first nanotechnology drug delivery systems, described in the
tention [11]. In addition, the lack of functional lymphatic vessels and 1960s, were lipid vesicles, which were later called liposomes [18,19].
the vascular hyperpermeability inside tumors results in interstitial Subsequently, a variety of drug delivery nanosystems were developed
110 F. Danhier / Journal of Controlled Release 244 (2016) 108–121

Fig. 1. (A) Schematic representation of the conceptual passive targeting (EPR effect) of nanomedicine. (B) Active targeting of nanomedicine grafted with peptide or antibody able to bind
specific receptors overexpressed by (1) cancer cells or (2) endothelial cells.
Adapted from [9,15].

and, in 1976, the first sustained release drug delivery system was de- sarcoma, (iii) Myocet®, a 150 nm non-PEGylated liposomal doxorubicin
scribed [20]. In 1980, the first example of the specific targeting of lipo- formulation approved in Europe and Canada for metastatic breast can-
some was described with a system able to respond to changes in pH cers, and (iv) MM-398®, a 100 nm liposomal formulation of irinotecan
to trigger drug release [21]. In 1987, the first long-circulating liposome recently approved for the treatment of pancreatic adenocarcinoma in
was described, introducing the concept of PEGylation, where the lipo- combination with 5-fluorouracil (5-FU) and leucovorin (Table 1).
some surface was covered with polyethylene glycol (PEG) chains reduc- While liposomes are a dominant class of nanomedicine, other
ing opsonization and premature clearance, therefore increasing blood nanomedicines have been approved, such as nanoparticles and micelles
circulation times and potentially enhancing tumor accumulation [18]. for cancer therapy. These nanoformulations include Abraxane®, 130 nm
In 1995, doxorubicin-loaded PEGylated liposomes (named Doxil® in albumin-bound paclitaxel particles, indicated for metastatic breast can-
the USA and Caelyx® in other countries) were approved for the treat- cer and recently for pancreatic adenocarcinoma. Outside of the global
ment of AIDS-associated Kaposi's sarcoma (Table 1) [22]. Doxil® was market, Genexol-PM® is a 20–50 nm polymeric micelle formulation of
the first drug carrier to reach the market in the United States, gaining paclitaxel approved in South Korea for metastatic breast cancer (Table
FDA approval only five years after its development was first reported 1) [16].
[22]. The clinical therapeutic efficacy of Doxil® was superior in ovarian Abraxane®, with an annual revenue estimated at $967 million, is one
cancers and AIDS-related Kaposi's sarcoma compared to standard ther- of the major success stories in the development of nanomedicines.
apies, while equivalent in multiple myeloma and metastatic breast can- Abraxane® (an albumin-bound nanoparticle of paclitaxel) was devel-
cer [23,24]. However, the main benefit of Doxil® in treating solid tumors oped to retain the therapeutic benefits of paclitaxel but eliminate the
was the reduced cardiotoxicity gained by limiting doxorubicin exposure toxicities associated with the emulsifier Cremophor® EL in the commer-
to cardiac tissue. However, toxicity was not reduced across the board, cialized paclitaxel formulation (Taxol®) [25]. The maximum tolerated
but was aggravated in other categories, most notably skin toxicity in dose (MTD) of Abraxane® was approximately 50% higher than reported
the form of palmar–plantar erythrodysesthesia (PPE)—hand and foot for Taxol®. Pharmacokinetic assessments also showed that paclitaxel
syndrome. This toxicity directly results from the extended circulation clearance and volume of distribution were higher for Abraxane® than
time of the PEGylated liposomes, which allows time for the kinetically for Taxol®. These differences in pharmacokinetic properties may be as-
slow process of extravasation into the skin to become significant. sociated with the higher intratumoral concentrations observed with
Doxil® formulation failed to show drastic enhanced tumor accumula- Abraxane® compared with the equivalent dose of Taxol®. This improve-
tion over free doxorubicin, indicating that a minimum EPR is unreliable ment was credited to the receptor-ligand targeting via active albumin
in human tumors. The EPR hypothesis would suggest that longer circu- transport pathways. It is difficult to decipher the exact impact of the
lation times produce higher intratumoral accumulation, which should EPR effect, however, the increase in efficacy could be easily explained
in turn improve patient outcomes. It could mean that clinical EPR is by the increased dose of available drug alone [24]. It should be noted
not as well understood as originally thought [24]. that it is unclear whether or not the nanoparticles dissolved when in-
The initial approval of Doxil® was followed by various other liposo- fused into patients. Thus, the clinical benefit from Abraxane® is probably
mal formulations including, (i) Ambisome® for the delivery of not due to its functioning as nanoparticles but to other factors such as
amphotericin B for infectious disease indications, (ii) DaunoXome®, the removal of Cremophor® EL from the formulation that causes toxic-
50 nm daunorubicin liposomes approved for AIDS-associated Kaposi's ities of its own [14,26].
F. Danhier / Journal of Controlled Release 244 (2016) 108–121 111

Table 1
Examples of clinically used nanomedicines in cancer therapy.
Clinical data are extracted from http://www.clinicaltrials.gov (2016).

Nanocarriers Drug Name Indications Status

Micelles
Paclitaxel Genexol®-PM Breast, lung, pancreatic cancer Approveda
Recurrent breast cancer
Paclitaxel Paclical Ovarian cancer III
Paclitaxel NK105 Gastric cancer III
Doxorubicin NK911 Various solid tumors II
Cisplatin NC-6004 Pancreatic cancer I/II
Epirubicin NC-6300 Various solid tumors I
Oxaliplatin NC-4016 Various solid tumors I

Nanoparticles
Albumin- paclitaxel Abraxane® Metastatic breast cancer Approved
Doxorubicin Transdrug® Hepatocarcinoma Approved
Doxorubicin BA-003 Hepatocellular carcinoma III
Mitoxantrone DHAD-PBCA-NPs Hepatocellular carcinoma II
e
Docetaxel BIND-014 NSCLCb II
Camptothecin CRLX101 NSCLCb II
Camptothecin IT-101 Solid tumors I/II
dnG1 plasmid DNA Rexin-G Various solid tumors I/II
Anti-PKNe siRNA Atu027 Various solid tumors I/II
Docetaxel ABI-008 Breast, prostate cancers I/II
Rapamycin ABI-009 Non-hematologic malignancies I/II
BikDD plasmid DNA C-Visa-BikDD Pancreatic cancer I
Paclitaxel Nanoxel® Advanced breast cancer I
Docetaxel Docetaxel-PNP Various solid tumors I
e
Anti-RRM2 siRNA CALAA-01 Various solid tumors I

Polymer drug conjugate


Asparginase Oncaspar® Leukemia III
Paclitaxel Xyotax® Breast, ovarian cancer III
(CT-2103) Advanced lung cancer III
Paclitaxel Taxoprexin® Various solid tumors II-III
Doxorubicin PK1 Breast, lung, colon II
IFN-α2a/b PegAsys/PegIntron® Melanoma/Leukemia II
Oxaliplatin ProLindac® Ovarian cancer II
Diaminocyclohexane platinium AP 5346 Ovarian cancer II
Docetaxel DEP® Advanced cancers I
CPT XMT-1001 Gastric, NSCLC cancers I

Liposomes
Doxorubicin Doxil/Caelix® Ovarian, metastatic breast cancer, Kaposi sarcoma Approved
Doxorubicin Myocet® Breast cancer Approved
Daunorubicin DaunoXome® Kaposi Sarcoma Approved
Vincristine Onco-TCS® Non-hodgkin lymphoma Approved
Cytabirine Depocyt® Lymphomatus meningitis Approved
Vincristine Marqibo® Leukemia Approved
Mifamurtide Mepact® Osteosarcoma Approved
Irinotecan Onivyde® (MM-398) Metastatic pancreas cancer Approved
Paclitaxel Lipusu® NSCLC, breast cancer Approvedc
Paclitaxel PICN® Metastatic breast cancer Approvedd
Doxorubicin Thermodox® Liver, breast cancers III
Cisplatin Lipolatin NSCLC III
9-nitrocamptothecin 9NC-LP Hepatocellular carcinoma II/III
Cisplatin SPI-077 Solid tumors I/II/III
Oxaliplatin Lipoxal Advanced cancers II
Paclitaxel EndoTAG-1 Breast, liver, pancreatic cancers II
Lutotecan OSI-211 Solid cancers II
Docetaxel LE-DT Pancreatic, prostatic cancers I/II
Paclitaxel LEP-ETU Breast cancer, leukemia I/II
PLK1 siRNA TKM-080301 Neuroendocrine tumors I/II
PKN3 siRNA Atu027 Pancreatic cancer I/II
Doxorubicin 2B3-101 Solid tumors I/II
Irinotecan NL CPT-11 Reccurent high grade glioma I
CEBPA saRNA MTL-CEBPA Liver cancer I
Topotecan TL1 Various solid tumors I
Irinotecan IHL-305 Advanced solid tumors I
Docetaxel ATI-1123 Solid tumors I
Vinorelbine Alocrest Bresat and lung cancers I
Cisplatin LiPlaCis Advanced solid tumors I
e
Doxorubicin MCC-465 Metastatic stomach cancer I
e
p53 gene SGT-53 Various solid tumors I
a
Approved in Korea.
b
Advanced Non-Small Cell Lung Cancer.
c
Approved in China.
d
Approved in India.
e
Targeted nanomedicine.
112 F. Danhier / Journal of Controlled Release 244 (2016) 108–121

For more than thirty years, great effort has been expended to devel- the lower and heterogeneous pericyte coverage, (iv) the heterogeneous
op better nanocarriers. The advantages of nanomedicines were myriad; basement membrane (BM) and (v) the higher and heterogeneous den-
they could be endlessly manipulated to take on many shapes, sizes and sity of the extracellular matrix (ECM), leading to an elevated IFP which
functionalities; they could simultaneously perform multiple tasks, such forces nanomedicines to escape the blood compartment by diffusion
as treatment and imaging; they could carry large quantities of therapeu- rather than the much more efficient convective transport [37–39].
tic agents, and they would automatically preferentially accumulate in Hence, regarding these notable differences in the tumor microenviron-
tumors by virtue of their size while sparing the rest of the body. In ment, a new schematic representation of the passive targeting (EPR ef-
spite of these advantages, of the hundreds of drug carrier designs re- fect) in human tumors is illustrated in Fig. 2. The practicalities of
ported, only few have been approved for clinical use. The two most suc- evaluating the EPR effect in human tumors are relatively costly and
cessful formulations, Doxil® and Abraxane®, are surprising due to their time-consuming. However, biological conditions driving EPR variability
simplicity. is crucial. Tumor histology is not sufficient to predict EPR effect. Imaging
The identification of new disease biomarkers is essential in the modalities (such as computer tomography, magnetic resonance imag-
choice and development of targeting ligands. While non-targeted ing (MRI) or functional ultrasound) are needed to investigate the corre-
nanomedicines have been clinically efficacious, numerous contradicting lation between vascularization (permeability, perfusion, IFP) and intra-
data regarding the benefit of adding a targeting ligand have been shown tumor accumulation. Alternatively, researchers could use more relevant
[27]. The tumor accumulation is more largely mediated by physico- in vivo tumor models, such as patient-derived tumor explant (PDX)
chemical properties of the nanoparticles compared to active targeting models, which more faithfully reflect the morphology, complexity and
[28]. Therefore, even in the absence of targeting ligands, nanomedicines heterogeneity of clinical tumors. Indeed, these tumor models are more
can be designed to better target a specific tissue, or be nonspecifically mature and less permeable than xenografts [40].
absorbed by cells, by optimizing their physico-chemical properties
[29]. However, as aforementioned, specific uptake by cancer cells is fa- 3.1. The heterogeneity or lack of fenestrations in the tumor endothelium
cilitated by active targeting. Despite a large number of publications in
this area, only a few systems with active targeting are under investiga- Because of the rapid murine tumor growth, blood vessels in mouse
tion in clinical trials (Table 1) [30–32]. These targeted systems include tumor generally do not develop properly, and they consequently tend
(i) MCC-465, doxorubicin encapsulated in immunoliposomes tagged to be much more leaky with endothelial cells presenting many fenestra-
with the F(ab′)2 fragment of human monoclonal antibody GAH, which tions [4]. Unfortunately, in humans, not all tumor vessels are leaky,
binds to neoplastic stomach tissues [33]; (ii) SGT-53, a liposomal which causes a heterogeneous distribution of pore sizes and thus, het-
nanocomplex designed for systemic, tumor-targeting delivery of the erogeneous extravasation and delivery [2].
p53 gene. In this nanodelivery system, an anti-transferrin receptor sin- An illustration of this statement consists in the difference of tumor
gle-chain antibody fragment (scFv) serves as the targeting moiety to accumulation of 5 nm-sized radiolabeled HPMA copolymers in two dif-
target cancer cells through the transferrin glycoprotein receptor [27]; ferent rat tumor models. On one side, in Dunning AT1 prostate carcino-
(iii) BIND-014, docetaxel-loaded nanoparticles targeting the Prostate- ma growing to 1 cm in diameter within 2 weeks, the preferential
Specific Membrane Antigen (PSMA); and (iv) CALAA-01, siRNA-loaded accumulation of copolymers in tumor tissue has been correlated to
nanoparticles containing human transferrin as targeting ligand for bind- poorly differentiated blood vessels and hardly any coverage with
ing transferrin receptors [34]. pericytes. On the other hand, in Dunning H prostate carcinoma growing
to 1 cm in diameter in 1 year, a lesser accumulation of copolymers in
3. Heterogeneity in the EPR effect: a new clinically relevant EPR ef- tumor tissue has been correlated to properly differentiated blood ves-
fect model sels and dense coverage with pericytes [41].

While the number of publications citing “EPR” and “nanomedicine” 3.2. Hypoxic areas
has exponentially increased, this creative flourish has largely failed to
translate into the clinic. While the EPR effect has been well documented The heterogeneity of blood flow through tissues leads to regions be-
in small animal models, clinical data is less clear. Nanomedicines have coming acidic or hypoxic: most human solid tumors contain hypoxic
almost uniformly failed to produce improved efficacy in the clinic, in areas. Hypoxia is the direct consequence not only of the chaotic prolifer-
spite of tremendously successful preclinical results (see examples of ation of cancer cells that places them at some distance from the nearest
Abraxane® and Doxil® in the Section 2.3) [24,35]. capillary, but also of the abnormal structure of the neovasculature net-
Models being used to study EPR in laboratories are not sufficiently work resulting in transient blood flow [42]. The subsequent hypoxia in
accurate and require reconsideration. Indeed, murine tumor models tumor cells not only induces resistance to radiotherapy, but also causes
drastically differ from human cancers in many aspects including the resistance to several cytotoxic drugs, while also inducing genetic insta-
rate of development, the size relative to host, metabolic rates and host bility and selecting for more malignant tumor cells with potentially
lifespan. More particularly, human tumors present many differences in metastatic properties. Additionally, another consequence of the hetero-
their tumor microenvironment compared with murine tumors simply geneity in human cancer relating to the abnormal coagulation or
because most rodent tumors grow much faster. A subcutaneous tumor clotting in tumor vessels is the resulting fibrinolysis or thrombocytope-
grows to ±1 cm (≈0.5 g) within 2–4 weeks. In human, this would rep- nia that can affect the EPR effect [43].
resent a ± 20 cm (≈ 1–2 kg) growth [4]. The large tumor-to-body
weight ratio in mice compared to human patients may significantly 3.3. The lower and heterogeneous pericyte coverage
alter the pharmacokinetics of drug carriers. Murine tumors are often ob-
served to be as much as 10% of the mouse's body weight. The volume of The relationship between pericyte coverage and nanomedicine ex-
an equivalent tumor in a 70 kg human would be the size of a basketball travasation has been investigated in detail. In murine models, all
[36]. Such large tumors filter a significant proportion of the injected tumor microvessels have low and loose pericyte coverage. In human tu-
dose of a drug and act as a repository for the drug, effectively accentuat- mors, emerging evidence has demonstrated heterogeneous pericyte
ing the efficacy while mitigating the toxicity. Moreover, tumor microen- coverage within one single tumor. By defining pericytes as alpha-
vironment in human cancers present some major differences in smooth muscle actin (α-SMA) positive cells attached to endothelial
comparison with murine tumors: (i) the heterogeneity or lack of fenes- cells, malignant tumors have been classified into high pericyte coverage
trations in the tumor endothelium, (ii) the heterogeneity of blood flow and low-pericyte coverage subtypes. Additionally, more coverage has
through tissues, leading to regions becoming acidic or hypoxic [37], (iii) been related to a worse prognosis and more fibrotic interstitium for
F. Danhier / Journal of Controlled Release 244 (2016) 108–121 113

Fig. 2. Schematic representation of the passive targeting of nanomedicine (EPR effect) in human tumors. Tumor microenvironment in human cancers present some major differences in
comparison with murine tumors: (1) the heterogeneity or lack of fenestrations in the tumor endothelium, (2) the heterogeneity of blood flow through tissues, leading to regions becoming
acidic or hypoxic, (3) the lower and heterogeneous pericyte coverage, (4) the heterogeneous basement membrane and (5) the higher and heterogeneous density of the extracellular
matrix (ECM), leading to an elevated interstitial fluid pressure (IFP).
Adapted from [15].

different tumors (pancreas, gastric, renal cancers or glioblastoma; 60– by mesh size and thickness of the BM. Therefore, the type IV collagen
70% coverage), when compared to low-coverage cancers with a better density, mesh size, number of layers, and association with the vessels
prognosis such as colon and ovarian cancers (10% coverage) [44,45]. form a barrier to the nanomedicine extravasation [45,48,49].
Disparate intratumoral nanoparticle transport in response to cytokine- Extravasation of 1 nm doxorubicin, 50 nm macromolecule FITC-
mediated modification of pericyte coverage has been observed in two tagged dextran and 80 nm PEGylated liposomes were evaluated on
types of murine tumors. Murine colon cancer CT26 is an example of the BM/vessel over-lapped 3LL model and the BM/vessel dissociated
low-pericyte coverage tumor, while BxPC3 pancreatic model is model 4T1. The extravasation pattern of small molecules (including
hypovascularized with pericyte coverage of 70%. Kinase inhibitors doxorubicin and dextran) was comparable, suggesting that vascular col-
were able to improve 2 MDa dextran and Doxil® penetration, leading lagen could only modulate the transport of small molecules to a limited
to increased tumor inhibition, in the BxPC3 tumor model. This observa- extent. However, the extravasation of liposomes was significantly dif-
tion is due to the blockage of pericyte proliferation induced by kinase in- ferent between these two types of tumors. Extravasation only occurred
hibitors. On the CT26 tumor model, the pericyte coverage was too low to from the vessels that were not tightly covered by collagen type IV [50].
achieve any additional effect with the kinase inhibitors [46,47]. The re-
lationship between pericyte coverage and nanoparticle extravasation
3.5. The higher and heterogeneous density of the ECM
depends on the original pericyte coverage, blood vessel stabilization
and extracellular content, since pericyte is an indispensable factor for
Elevated intratumoral IFP has been well documented not only in an-
vessel stabilization and maturation. Neither leaky, unmatured blood
imals but also in clinical tumors [1]. A direct consequence of the elevat-
vessels with little coverage, nor over-matured vessels with abundant
ed IFP levels is that the main mechanism of mass transport across the
pericyte coverage are suitable for nanomedicine delivery [45].
vessel wall is diffusion, a process that is much slower than convection.
The large size of nanoparticles along with the uniformly elevated IFP
3.4. Heterogeneous basement membrane in tumors hinder transport across the vessel walls and compromise
the benefits of the EPR effect [2]. Additionally, the diffusion coefficient
The BM is a specialized form of the ECM that serves as a scaffold for of nanoparticles is considerably hindered by interactions with the ECM.
endothelial and mural cells. The main components of the BM are lami- The ECM consists in a cross-linked network of collagen and elastin fi-
nin and type IV collagen, which form distinct sheet-like structure linked bers, glycoproteins, proteoglycans and hyaluronic acid. Contrarily to
together by nidogen and heparin sulfates [45]. Heterogeneous BM mor- BM, the ECM mainly consists in a network of collagen (Types I, II, III)
phologies exist in different regions of the same tumor or different tumor interacting with the other components. The ECM not only provides
types. Even if BM does not induce elevated IFP, the presence of BM limits structural integrity, but also helps to transport important nutrients
the penetration of nanomedicines through pole-opening on capillary and oxygen to support cell growth [51]. At the cellular level, ECM is lo-
walls such as the ECM (see the next section). Hence, diffusion of calized at two different sites, in the basement membrane and in the ma-
nanomedicines with sizes larger than 100 nm could be severely affected trix of the interstitial space [52]. Cancer-associated fibroblasts (CAFs)
114 F. Danhier / Journal of Controlled Release 244 (2016) 108–121

are the major players involved in the ECM-mediated malignant changes, carboplatin. Doxil® has also been combined very well with the
which include upregulated ECM synthesis, posttranslational modifica- alkylating agent canfosfamide in ovarian carcinoma and with the pro-
tions and matrix metalloproteinase (MMP)-induced ECM remodeling teasome inhibitor bortezimab in multiple myeloma [4].
[52,53]. Diffusion of nanoparticles across the thick interstitial matrix is
the last step to approaching tumor cells before cell internalization. For 4.2. Local delivery
tumors with less ECM, nanomedicine can easily diffuse. However, for tu-
mors with a thick interstitial matrix, this process is more intractable. By definition, the local delivery does not require EPR effect. Local
Unfortunately, the ECM is much more dense and thick in human tumors chemotherapy is of great importance in cases of solid tumors, which
than in murine ones [45]. Collagen becomes increasingly important as represents N 85% of human cancers. The local delivery of anticancer
the particle size approaches or exceeds the matrix mesh size that ranges drugs is particularly preferred if cancer cells are accumulated in parts
between 20 and 40 nm in solid tumors. Particles larger than the mesh of the body which are easily accessible from the outside, such as the
size can be prevented from diffusing from the matrix entirely, those skin (melanoma). In this case, topical iontophoretic or transdermal ad-
near the mesh size can be hindered, and small particles can pass ministrations are chosen. When the tumor site is not located superficial-
through relatively uninhibited [36]. In conclusion, the dense and hetero- ly, different regional drug delivery strategies can be used, including the
geneous structure of the ECM combined with the subsequent elevated injection of anticancer agents directly into tumor tissue, the use of in-
IFP in human tumors blocks large nanomedicine penetration and results jectable in situ forming drug carriers or injectable platforms. These
in the low and heterogeneous distribution of nanomedicine into methods can be successfully applied in the treatment of solid tumors lo-
tumors. cated in breast, lungs, liver or bones. Higher achievable drug concentra-
tions combined with minimal side effects are the two main advantages
4. What is the future for nanomedicines without EPR effect? of this kind of therapy [54,55]. Moreover, the use of nanomedicine al-
lows the sustained release of the drug and thereby enhances the efficacy
EPR effect is probably the most important concept in new anticancer of the treatment. Nevertheless, the current challenge of the develop-
drug delivery systems. Nevertheless, as aforementioned, the EPR effect ment of local drug delivery systems consists in the enhancement of
is really heterogeneous in humans. Thus the basic rationale of the design drug stability and the limitation of drug diffusion away from the
and development of nanomedicines in cancer therapy is failing and it tumor. A particularly interesting indication of local drug delivery sys-
would be necessary to stop claiming efficacy gains via the EPR effect, tems is glioblastoma (GBM), since conventional chemotherapy fails
while the tumor targeting cannot be proved in the clinic. It is probably due to the poor crossing of the blood-brain barrier (BBB). Many strate-
time to dethrone the EPR effect and to ask the question: what is the fu- gies have been developed to circumvent the BBB and reach therapeutic
ture of nanomedicines without EPR effect? concentrations of chemotherapeutic drugs in brain tumors. Among
In this section, I would like to highlight the different advantages and them, small lipophilic drugs have been used to passively pass the BBB,
possibilities that nanomedicines can offer in cancer therapy. Three main while active compounds have been modified or incorporated into
applications of nanomedicines do not require the EPR effect: (i) the de- nanocarriers in order to reach the brain parenchyma by passive
crease of toxicity of chemotherapeutic agents, (ii) the local delivery of targeting or active targeting of the BBB endothelial cells [56]. Others
anticancer agents and (iii) the use of nanomedicines as a tool for imag- have tried to modify the BBB permeability or used focused ultrasounds
ing the tumor microenvironment. to transiently open the BBB for drug delivery [57]. Recently, a specific
and innovative hydrogel uniquely formed of lipid nanocapsules (LNC)
4.1. Toxicity and lauroyl-gemcitabine (GemC12) has been developed and suggested
as local treatment for GBM. This formulation fulfills the definition of
Regarding the two major examples of nanomedicines on the market both nanomedicine and hydrogel. Indeed, the formation of the hydrogel
(Abraxane® and Doxil®), nanomedicines are able to reduce the toxicity is due to the location of the GemC12 at the oil-water interface of the LNC,
of chemotherapeutic agents, rather than to improve their efficacy. Most which allows the formation of H-bond cross-links between the drug
conventional chemotherapies are effective but their clinical use is limit- moieties and the immobilization of the water phase [58].
ed by their toxicity (neuropathy induced by paclitaxel; cardiotoxicity
induced by doxorubicin or 5-fluorouracil, etc.). Drug delivery systems 4.3. Nanomedicines as a tool for imaging the tumor microenvironment
able to reduce side effects while increasing the maximum tolerated
dose (MTD) would be in itself beneficial for patients. For example, the Nanoparticles are frequently suggested as diagnostic agents. Never-
MTD of Abraxane® was approximately 70–80% higher than that report- theless, none of these nanodiagnostics were finally approved for clinical
ed for Taxol®. In a Phase III study of 454 patients with metastatic breast use. Indeed, the long-circulating properties of these nanoparticles,
cancer given Abraxane® or Taxol® intravenously every 3 weeks, re- allowing peaks ranging from 12 and 48 h, were unable to achieve the
sponse rates were significantly greater in patients treated with rapid and highly site-specific contrast enhancement needed for an ef-
Abraxane® than those receiving Taxol®. Pharmacokinetic assessment fective diagnosis [59]. The visualization of blood vessels is of high cancer
also showed that paclitaxel clearance and volume of distribution were diagnostic relevance. As positive MR contrast agents, small paramagnet-
higher for Abraxane® than for Taxol®. Clearance was 13 L per hour per ic iron oxide (SPIO) nanoparticles were successfully applied in preclini-
m2 for Abraxane® versus 14.76 L per hour per m2 for Taxol® (p = cal and clinical trials but no product was finally approved for clinical use.
0.048). There are numerous examples of highly effective combination The only exception consists in the ultrasmall paramagnetic iron oxides
of nanomedicine with anticancer agents [54]. Nevertheless, the possibil- (USPIO) nanoparticles such as Sinerem®, Feridex®, Feraheme® and
ities of combinations are limited by the toxicity of each product. To se- Supravist®, which are used in the clinic for “vessel size imaging” only
lect a potent effective combination, the two drugs need (i) to present in one particular application: mononuclear phagocytic system (MPS)
different mechanisms of action, overcoming the problems connected imaging. The MPS consists of liver and spleen, which are the main or-
with tumor cell heterogeneity and their implication for drug resistance; gans in which nanoparticles are taken up and accumulate after intrave-
and (ii) to induce different side effects, avoiding cumulative toxicity. nous administration. For more details about the MPS imaging using
Since nanomedicine is able to decrease side effects, it can be used in nanoparticles, readers are invited to read the review of Kiessling et al.
combination with conventional chemotherapy, opening the way for [59].
other combinations. Abraxane® has been combined very well with Contrast agents and therapeutic agents can be combined within a
bevacizumab and with gemcitabine in patients suffering from metasta- single multifunctional nanomedicine, know as “theranostic
tic breast cancer, as was Genexol®-PM with cisplatin and with nanomedicine or nanotheranostics”. To be efficient, nanotheranostics
F. Danhier / Journal of Controlled Release 244 (2016) 108–121 115

need to (i) selectively accumulate within the tumor tissue and (ii) to de- demonstrated that the combination of both active strategy and magnet-
liver therapeutic dose of therapeutic agents with a detectable dose of ic targeting drastically enhanced (i) nanoparticle accumulation into the
contrast agent allowing detection of tumors at their earliest stage. The tumor tissue with an 8-fold increase compared to passive targeting
applications of nanotheranostics are various: (i) the non-invasive as- (1.12% and 0.135% of the injected dose, respectively), (ii) contrast in
sessment of the nanomedicine biodistribution and target site accumula- MRI and (iii) anti-cancer efficacy with a median survival time of
tion, (ii) the drug release monitoring, (iii) the triggered release of the 22 days compared to 13 for the passive targeting [60]. However, the suc-
drugs and contract agents via an external stimulus and (iv) the predic- cess of this strategy is limited by inadequate magnetic gradients. Many
tion of the therapeutic response, also called “personalized medicine” strategies have been developed to overcome this problem [65–68]. An-
[60]. All these approaches are extensively reviewed in the literature other prospect for imaging-guided drug delivery is for the prediction of
[60–64]. In Fig. 3, the clinical relevance of each application of the therapeutic response also called “personalized medicine” (Fig. 3C).
theranostics is proposed. Among these applications, some could have For this concept, Lammers et al. discriminate two different approaches:
a potential impact for clinical use or on the contrary some could be (i) the “biomarker approach” offers the opportunity to assist treatment
used in preclinical models as a tool to visualize the tumor microenviron- selection, response prediction and treatment monitoring; and (ii) the
ment allowing a better design of nanoparticles. Researchers developing “image guidance approach” allows planning, pre-operative guidance
nanotheranostics often perform the in vivo non-invasive assessment of and follow up of the therapeutic action [62]. Few clinical studies using
the nanomedicine biodistribution and the drug release monitoring to nanotheranostics have provided proof-of-principle for personalized
demonstrate the promising potential of their systems (Fig. 3A). I believe medicine. However, convincing clinical data are only available on tu-
that clinicians and pharmaceutical companies aiming at commercializ- mors associated with a strong EPR effect, such as Kaposi sarcoma [59].
ing nanomedicines should in parallel develop nanotheranostics as a For example, in a cohort of 7 patients, scintigraphic imaging with
tool for earliest phases of clinical trials since the initial biodistribution Caelyx®-99mTc-DTPA showed an increased (2.8 times higher)
and target site of accumulation, as well as the monitoring of the tumor intratumoral drug accumulation compared to the surrounding healthy
response, should be correlated. Nanotheranostics should therefore tissue [69]. Hence, we can again legitimately question the clinical rele-
allow a better understanding of how nanoparticles work. Alternatively, vance of this kind of system in the treatment of solid tumors. In this con-
in the specific case of nanotheranostics containing SPIO, it is possible to text, via their non-invasive biodistribution visualization and the
guide SPIO-based theranostics directly to the target tumor tissue with importance of the EPR, theranostics should address this shortcoming.
an external magnetic field, since SPIO presents superparamagnetic Assuming that there will be a clear correlation between tumor concen-
properties (Fig. 3B). When a permanent magnet is applied externally tration and therapeutic efficacy, it will be highly important to properly
near the tumor region, the magnetic gradient produced exerts attractive differentiate between low and high levels of target site accumulation.
forces on magnetic nanoparticles delivered via the circulation [60]. This These studies should also guide the specific use of nanomedicines in
targeting strategy could compensate the poor EPR effect of most tumors. clinical applications [60]. However, I believe that future clinical oncolo-
For example, Schleich et al. both quantify and visualize the accumula- gy needs to take advantage of these theranostic tools to (i) understand
tion of multifunctional nanoparticles into the tumors by Electron Spin the fate of nanoparticles distributed in the tumor microenvironment
Resonance spectroscopy and MRI, respectively. It has been and to (ii) adapt the treatment to each specific case and find the best

Fig. 3. Clinical relevance of the three main applications of nanotheranostics in cancer treatment. (A) The non-invasive assessment of the nanomedicine biodistribution and the drug release
monitoring. (B) Magnetic theranostics directly target tumor tissue with an external magnetic field. (C) The prediction of the therapeutic response also called “personalized medicine”
which is divided into two different approaches: (i) the “biomarker approach” and (ii) the “image guidance approach”.
Adapted from [68] for panel A and from [62] for panel C.
116 F. Danhier / Journal of Controlled Release 244 (2016) 108–121

treatment for each patient. To further promote the clinical translation of improved transport through the pores of the tumor vessel wall and in-
nanotheranostics for diagnostic and therapeutic purposes, an increased terstitial space [32,70]. Additionally, there is evidence that flexible
interdisciplinary collaboration and knowledge exchange between sci- nanoparticles of low elastic properties might have longer circulation
entists from various disciplines is needed [62]. times [32,71].

5. Strategies to improve delivery


5.1. Vascular mediators involved in the EPR effect
Since recent data supports a considerable barrier role of tumors
resulting in imperfect or inefficient EPR effect, many researchers are Vascular mediators involved in the EPR effect (Fig. 4) are the follow-
working to either increase the EPR effect or mitigate anti-EPR character- ing: (i) Bradykinin (kinin) is a major mediator of inflammation that in-
istics [52]. Strategies to modulate or to counterattack the abnormal duces extravasation and accumulation of body fluids in inflammatory
tumor microenvironment have shown promising results in the en- tissues. Kinin is also known to activate endothelial cell-derived nitric
hancement of anticancer drug delivery systems, providing therefore a oxide (NO) synthase, leading to an increase in NO (an important medi-
second wind to nanomedicines in cancer therapy. A better understand- ator of tumor vascular permeability) [11,72]. (ii) NO is produced from L-
ing of the tumor microenvironment may allow researchers to identify arginine by the inducible form of NO synthase (iNOS) in the presence of
the abnormal physiological processes that can be modulated to improve oxygen. In cancer, NO is extensively produced from an increased num-
drug penetration and efficacy in human solid tumors. Regarding formu- ber of infiltrated leukocytes and tumor cells [11,73]. (iii) Matrix metal-
lations currently in clinical use, nanoparticles with neutral charge and a loproteinases (MMPs) are zinc-dependent neutral endopeptidases that
size range of 12–50 nm would be ideal as far as transport is concerned. are highly expressed in tumor cells and play important roles in tumor
Micellar polymeric particles (i.e. NC-6004, NC-4016, NC-6300 and invasion, metastasis and angiogenesis. Activation of MMPs induces the
Genexol-PM®) as well as Abraxane® and CRLX101 reside within this remodeling of the ECM [11,74]. (iv) Prostaglandins (PGs) are lipid
range. Liposomal formulations usually have a larger size in the range compounds enzymatically derived from arachidonic acid by cyclooxy-
of 100 nm (e.g. Doxil®, MM-398, Thermodox®). In that sense, sophisti- genase (COX). Similarly to bradykinin, PGs are important mediators in
cation of nanoparticles (addition of targeting ligands) would increase inflammation and can be upregulated by inflammatory cytokines and
the size of the nanoparticles, leading to a lower entry of these targeted kinin. PGs prevent platelet aggregation, leukocyte adhesion and throm-
agents. For instance, MCC-465 and SGT-53 are targeted liposomes bosis formation, facilitating extravasation [11,75]. (v) Vascular endo-
with sizes N100 nm, while BIND-014 and CALAA-01 are polymeric thelial growth factor (VEGF) is the angiogenesis factor that is highly
nanoparticles with sizes of 100 and 75 nm, respectively. Whether in- upregulated in most tumors, which plays a crucial role in angiogenesis.
creased sophistication of nanomedicines overcomes delivery barriers VEGF was shown to exert its action via generation of NO [76]. Thus all
posed by their large size will depend not only on the nanoparticle prop- these vascular permeability factors facilitate the supply of nutrients
erties and drug-release kinetics, but also on the tumor type [32]. Elon- and oxygen, and thus angiogenesis.
gated nanoparticles with neutral charge should be preferred over The following section aims to provide an overview of the various
spherical ones since it has been demonstrated that they present an possibilities allowing nanomedicines to better enter the tumors, despite

Non-angiogenic tumor
Dormant Inhibitors Pro-activators
< 1-2 mm Hypoxia

Induction of expression of
Kinin pro-angiogenic proteins:
TNF- VEGF

Kinin-generating cascade

1. Endothelial cell activation A


N
iNOS G
I
O
MMP 2. Basement membrane degradation
COX PG G
E
N
I
3. Endothelial cell migration C

S
Vascular permeability W
NO 4. Vessel formation I
T
C
H
Normal cell COX: Cyclooxygenase 5. Angiogenic remodeling
Cancer cell PG: Prostaglandin
Dividing cell NO: Nitric oxide
Blood vessel with pericyte iNOS: Inducible form of NO synthase
Angiogenic
Apoptosing, necrotic cell TNF- : Tumor necrosis factor tumor
MMP: Matrix metalloproteinase
VEGF: Vascular endothelial growth factor

Fig. 4. Schematic representation of the factors involved in the angiogenic switch and consequently in the EPR effect.
Adapted from [77,78].
F. Danhier / Journal of Controlled Release 244 (2016) 108–121 117

Fig. 5. Strategies to modulate or to counterattack the abnormal tumor microenvironment have shown promising results in the enhancement of anticancer drug delivery systems, providing
therefore a second wind to nanomedicines in cancer therapy. ACE: angiotensin-converting enzyme; NG: nitroglycerin; NO: nitric oxide; VEGF: vascular endothelial growth factor; TGF:
transforming growth factor; MMP: Matrix metalloproteinase.

an altered and heterogeneous EPR effect. The different approaches are This concept of normalized vessels suggest that these “normal” ves-
summarized in the Fig. 5. sels could avoid the EPR effect and compromise the delivery of drugs to
tumors. R.K. Jain resolved this paradox by comparing properties of nor-
5.2. Microenvironment modifications/modulations mal, tumor and normalized vessels. It appeared that normalized vessels
remain permeable to large molecules but only in case of small NP
5.2.1. Direct permeability enhancement (b60 nm) [15,80]. Hence, normalization induces a better delivery of
Inhibitors of angiotensin-converting enzyme (ACE) (Fig. 5) inhibit the therapeutic entities by keeping the EPR effect intact and decreasing
conversion of angiotensin I to angiotensin II by carboxypeptidase. ACE the IFP (Fig. 6A). Although mostly described in mouse tumor models
inhibitors (such as enalapril or temocapril) inhibit the degradation of [81–83], several clinical trials have provided evidence of a normalized
bradykinin, thus raising the local bradykinin concentration in tumor tis- vasculature in cancer patients treated with anti-angiogenic agents [1,2].
sue, leading to activation of endothelial NOS because the level of brady- Transforming growth factor beta (TGF-β) (Fig. 5) can induce the ex-
kinin is increased. Thus, the EPR effect becomes more apparent, and pression of different key angiogenic factors, including VEGF and MMP-9
such inhibitors enhance delivery of macromolecular drugs or compo- expression in macrophages [84]. Therefore, TGF-β receptor antagonists
nents to the tumor [11,76]. (including small molecule kinase inhibitors and siRNAs) were the most
Nitroglycerin and other NO-releasing agents (Fig. 5) generate NO se- frequently used therapeutic agents to inhibit pericyte recruitment and
lectively in hypoxic tumor tissue compared with normoxic tissues. BM activation [45,85]. These inhibitors increase vessel leakiness and im-
Hence, such nitro agents facilitate the EPR effect via local NO generation prove the intratumoral penetration of sub-100 nm nanoparticles. TGF-β
in tumors, with drug delivery enhanced 2 to 3-fold with an improved inhibitors were also found to lower interstitial hypertension [45].
therapeutic effect [11,76].
One of the main purposes of the vascular normalization (Fig. 5) 5.2.2. Indirect permeability enhancement
strategy is to transform the phenotype of abnormal tumor vasculature The compression of blood vessels (Fig. 5) by proliferating tumor cells
into a phenotype that closely resembles functional normal vasculature in a confined space can reduce the diameter of blood vessels and conse-
by increasing coverage of pericytes and the basement membrane, a re- quently decrease the blood flow. Therefore the decompression of blood
duced IFP, and ultimately decreasing vessel leakiness [52]. VEGF overex- vessels can be achieved through the induction of apoptosis of specific
pression has been observed in most tumors and is well know to anticancer drugs. Successful decompression of tumor blood vessels
promote angiogenesis. Therefore, VEGF is a therapeutic target for vascu- was first reported by R.K. Jain and his colleagues, who used Taxanes
lar normalization. VEGF and other angiogenic signaling factors can be (paclitaxel and docetaxel) to induce apoptosis [86]. The term “tumor
blocked by using a variety of monoclonal antibodies (mAb). The first ap- priming” was coined by Au's group when they first describe the reduc-
proved anti-angiogenic mAb, bevacizumab (Avastin®) and its subse- tion of tumor cell density and expansion of the interstitial space, in-
quently developed derivative, ranibizumab, have been used in duced by paclitaxel [87,88]. Paclitaxel is particularly interesting since
metastatic colorectal cancer. Bevacizumab inhibits angiogenesis by its action on an increased EPR effect results in (i) enhanced perfusion
interacting with biologically active VEGF and hinders its binding to the resulting from a decreased IFP and transient vascular normalization
receptors (VEGFR-1/2) [52,79]. (anti-angiogenic effect) and (ii) the decompression of blood vessels
118 F. Danhier / Journal of Controlled Release 244 (2016) 108–121

Fig. 6. Schematic representation of the effects of (A) anti-angiogenic drugs and (B) paclitaxel on the tumor microenvironment and consequently on the EPR effect.
Adapted from [81].

resulting from the induction of apoptosis (cytotoxic effect) (Fig. 6B). VEGF-induced neovascularization [92]. Paclitaxel induces a transient
Since it has been demonstrated that paclitaxel induced apoptosis and normalization of the tumor vasculature and, thus, increased perfusion
reduced the density of intact neoplastic cells in both MCa-IV and that accounts for better delivery of chemotherapeutic agents [93,94].
HSTS-26T, the further in vivo determination of blood vessel decompres- The two requirements of effective paclitaxel tumor priming are its
sion has been assessed by measurements of diameter of tumor vessels dose and the time window, sufficient to produce ± 10% apoptosis. As
in HSTS-26T tumors implanted in transparent dorsal skin fold cham- aforementioned, the toxicity related to the high dose of paclitaxel re-
bers. At 48 and 96 h after paclitaxel administration (40 mg/kg), the di- quired to induce tumor priming can be decreased by using
ameter of tumor vessels was significantly increased. This increase in nanomedicine.
vascular diameters was associated with reductions in microvascular The ECM of solid tumors (Fig. 5) presents a transport barrier that re-
pressure and IFP, measured with the wick-in-needle technique [86]. stricts nanoparticle penetration, thereby limiting the efficacy of nano-
More recently, paclitaxel-loaded polymeric micelles (M-PTX) have sized delivery vehicles for cancer imaging and therapy. The relative ef-
been shown to enhance the blood flow and oxygenation of tumor 24 h fect of various components of the ECM most likely varies with tumor
after treatment [89]. These polymeric micelles were shown to be safe type, as ECM differs from tumors of different type and location [95]. Sev-
and effective delivery system for paclitaxel, drastically increasing the eral studies have shown that collagenase improves macromolecular
maximum tolerated dose of paclitaxel from 13,5 mg/kg to 80 mg/kg, penetration depth [96], while treatment with hyaluronidase yielded
for the commercial drug Taxol® and M-PTX, respectively [90]. When mixed results [97,98]. The enzyme hyaluronidase degrading
injected as pre-treatment 24 h after treatment, M-PTX (80 mg/kg) hyaluronidan is reported in phase I and II trials to prevent regrowth of
were shown to allow a more effective delivery into tumors of three various tumor types and to improve clinical outcome, when given adju-
anti-cancer nanomedicines: polymeric micelles (20 nm), liposomes vant to chemotherapy. The mechanism for the enhanced therapeutic ef-
(100 nm) and nanoparticles (230 nm). Qualitative intravital microscopy fect is not well understood, but degradation of the ECM is assumed to
using dorsal skin-flap window chambers provided clear evidence of en- improve the penetration of the drug [15,99]. However, the clinical utility
hanced permeability of the tumor endothelium in M-PTX pre-treated of enzymatic ECM degradation remains uncertain because (i) elevated
tumors. Quantitative studies also supported this hypothesis (Miles interstitial collagenase levels are associated with a poor patient progno-
assay, Electron Spin Resonance (ESR) spectroscopy and accumulation sis in a variety of cancers and (ii) ECM degradation may promote cancer
of radiolabeled-paclitaxel-loaded micelles [81]. This strategy has also progression and metastasis [88]. The hormone relaxin is also effective in
been supported by a clinical study of patients with breast cancer, reveal- increasing drug delivery by modifying collagen structure. Relaxin is a
ing that paclitaxel decreases IFP and improves tumor oxygenation [91]. nontoxic hormone secreted in women during pregnancy to induce up-
Downregulation of VEGF expression was suggested to be partly respon- regulation of MMPs. Relaxin reduces the collagen content by down-reg-
sible for this phenomenon. Paclitaxel was found to have broad inhibito- ulating fibroblast activity and up-regulating collagenase synthesis [52,
ry effects on angiogenesis, including the proliferation, motility and cord 100]. The proteolysis of the extracellular matrices fuels the process of
formation of endothelial cells, the angiogenic response in vivo, and angiogenesis and is mediated by a family of proteases known as MMP.
F. Danhier / Journal of Controlled Release 244 (2016) 108–121 119

Of the MMP family, MMP-2 and MMP-9 are thought to play a more specifically in tumors (in that case, the temperature should be consid-
prominent role in tumorigenesis. One of the most common strategies ered as an endogenous stimulus). However, the majority of drug deliv-
to target MMP consists in the use of tissue inhibitor of MMP (TIMP). An- ery systems using temperature as stimulus need an external heating
other approach is the development of MMP-sensitive drug delivery sys- source (Ultrasound, magnetic field, etc.) to improve the drug release.
tems [51,101,102]. (ii) Light: photo-sensitive carriers trigger drug release at the desired
target through stimulation by light. (iii) Magnetic field: magnetic
5.3. Multifunctional nanomedicines nanomedicines are guided to the target tissue under programmable ex-
posure of an external magnetic field. (iv) Ultrasound: the development
5.3.1. Tumor-penetrating peptides of ultrasonic sensitive nanocarriers for ultrasonography expands ultra-
Tumor-homing peptides (Fig. 5), identified by phage library screen- sound techniques to be unique and effective methods to capture drug
ing, have recently been recognized for their ability to enhance targeted carriers and trigger drug release by tuning the ultrasound frequency,
drug delivery into tumors [52]. The major example of this strategy is the duty cycles and time of exposure [105,106]. Exogenous-stimuli respon-
cyclic iRGD peptide. The iRGD peptide is constituted from CRGDK/RGPD, sive systems present some disadvantages: they require expensive
containing a cryptic CendR motif, RGDK/R that possesses CendR-like tis- equipments, which are not always practical. Additionally, the depth of
sue and cell penetrating activities. Intravenously injected nanomedicine tissue penetration is a major issue of these systems. For more details
coupled to iRGD bound to tumor vessels and spread into the extravascu- about these strategies, an excellent review of P. Couvreur and colleagues
lar tumor parenchyma, whereas conventional RGD peptides (CRGDC is available [107].
and RGD-4C) only delivered the cargo to the blood vessels. In a first
step, the intact peptide binds to the endothelial cell expressing αv
6. Conclusion
integrins. In a second step, a protease cleaves iRGD and exposes the
cryptic CendR motif RGDK, which can interact with the neuropilin-1 re-
EPR effect is probably the most important concept in new anticancer
ceptor and thereby increase tumor vascular permeability. Systemic in-
drug delivery systems. Nevertheless, the EPR effect is extremely hetero-
jection with iRGD improved the therapeutic index of drugs of various
geneous in humans. Thus the basic rationale of the design and develop-
compositions including small molecules ((doxorubicin), nanoparticles
ment of nanomedicines in cancer therapy is failing and it would be
(nab-paclitaxel (Abraxane®) and doxorubicin liposomes) and a mono-
necessary to stop claiming efficacy gains via the EPR effect, while the
clonal antibody (trastuzumab). Indeed, when injected with iRGD,
tumor targeting cannot be proved in the clinic. First, nanomedicine
Abraxane® accumulated 12-fold more in orthotopic BT474 human
should be studied on more clinically relevant tumor models or should
breast tumors than Abraxane® given alone, and penetrated far from
be dedicated only for tumors presenting a strong EPR effect in the clinic,
the tumor vessels. They also tested doxorubicin in a liposomal formula-
such as Kaposi sarcoma and head and neck tumors. Alternatively,
tion (particle diameter = 120 nm) on orthotopic 22Rv1 human prostate
nanomedicines can also be used for oncologic applications that do not
tumor model. The results were similar to those with free doxorubicin
require the EPR effect: the decrease of toxicity of chemotherapeutic
with regard to treatment efficacy, tumor accumulation of the drug
agents, the local delivery of anticancer agents and the use of
(14-fold), tumor penetration and toxicity. These results indicate that,
nanomedicines as a tool for imaging the tumor microenvironment. Fi-
in comparison to drug administered alone, the iRGD combination ther-
nally, recent evidence supports a considerable barrier role of tumors
apy provides equivalent or better antitumor efficacy at a three-fold
resulting in imperfect or inefficient EPR effect. In this context, strategies
lower dose of the drug, with a commensurate reduction in toxicity. Fi-
to modulate or to counterattack the abnormal tumor microenvironment
nally, they tested the iRGD combination regimen in mice bearing tu-
have shown promising results in the enhancement of anticancer drug
mors derived from BT474 human breast tumor cells, which
delivery systems, providing therefore a second wind to nanomedicines
overexpress HER2/neu/ErbB2. Co-injection of iRGD resulted in a 14-
in cancer therapy.
fold increase in trastuzumab- positive areas within the tumor. An en-
zyme-linked immunosorbent assay (ELISA) showed that iRGD en-
hanced the accumulation of trastuzumab in the tumors 40-fold. Acknowledgments
Combining trastuzumab with iRGD increased the drug potency at differ-
ent trastuzumab dose levels. At a clinical dose (9 mg/kg), the combina- This work is supported by the Fonds National de la recherché
tion eradicated all tumors in the mice, whereas the equivalent dose of Scientifique (F.R.S.-FNRS).
trastuzumab alone only slowed tumor growth. Thus, co-administration
of iRGD may be a valuable way to enhance the efficacy of anti-cancer References
drugs while reducing their side effects, a primary goal of cancer therapy
[1] C.H. Heldin, K. Rubin, K. Pietras, A. Ostman, High interstitial fluid pressure - an ob-
research [77,103,104]. stacle in cancer therapy, Nat. Rev. Cancer 4 (10) (2004) 806–813.
[2] R.K. Jain, T. Stylianopoulos, Delivering nanomedicine to solid tumors, Nat. Rev. Clin.
5.3.2. Stimuli-responsive nanomedicine Oncol. 7 (11) (2010) 653–664.
[3] A.I. Minchinton, I.F. Tannock, Drug penetration in solid tumours, Nat. Rev. Cancer 6
To enhance the preferential accumulation of nanomedicine or drug (8) (2006) 583–592.
release in tumors, efforts are increasing to develop stimuli-responsive [4] T. Lammers, F. Kiessling, W.E. Hennink, G. Storm, Drug targeting to tumors: principles,
nanomedicine (Fig. 5) that use endogenous or exogenous (also called pitfalls and (pre-) clinical progress, J. Control. Release 161 (2) (2012) 175–187.
[5] Y. Malam, M. Loizidou, A.M. Seifalian, Liposomes and nanoparticles: nanosized ve-
external) stimuli to overcome the limitation of EPR effect [105]. Endog- hicles for drug delivery in cancer, Trends Pharmacol. Sci. 30 (11) (2009) 592–599.
enous stimuli-responsive nanomedicine utilize local tumor microenvi- [6] H.S. Park, H.A. Kim, J.W. Jung, Y.K. Kim, S.K. Lee, S.S. Kim, D.H. Nahm, Metallopro-
ronment factors: (i) Changes in pH: the extracellular pH values in teinase-9 is increased after toluene diisocyanate exposure in the induced sputum
from patients with toluene diisocyanate-induced asthma, Clin. Exp. Allergy 33
solid tumors are usually below 7.0, due to the high rate of glycolysis. (1) (2003) 113–118.
(ii) Redox gradients: the redox potential in microenvironments is mul- [7] H. Hillaireau, P. Couvreur, Nanocarriers' entry into the cell: relevance to drug deliv-
tivariate in different tissues. More particularly, tumors present hypoxic ery, Cell. Mol. Life Sci. 66 (17) (2009) 2873–2896.
[8] R. Duncan, Polymer conjugates as anticancer nanomedicines, Nat. Rev. Cancer 6 (9)
areas and elevated reactive oxygen species (ROS). (iii) Enzyme concen-
(2006) 688–701.
tration: enzymes (such as glycosidases, lipase, phospholipases and [9] F. Danhier, O. Feron, V. Préat, To exploit the tumor microenvironment: passive and
MMPs) are overexpressed in tumors. On the contrary, exogenous-re- active tumor targeting of nanocarriers for anti-cancer drug delivery, J. Control. Re-
sponsive nanomedicine respond to external stimuli: (i) Temperature: lease 148 (2) (2010) 135–146.
[10] Y. Matsumura, H. Maeda, A new concept for macromolecular therapeutics in can-
considering the temperature difference between tumor and normal tis- cer chemotherapy: mechanism of tumoritropic accumulation of proteins and the
sues, some nanomedicines have been designed to be triggered antitumor agent smancs, Cancer Res. 46 (12 Pt 1) (1986) 6387–6392.
120 F. Danhier / Journal of Controlled Release 244 (2016) 108–121

[11] J. Fang, H. Nakamura, H. Maeda, The EPR effect: unique features of tumor blood HPMA copolymer-based drug delivery systems, J. Control. Release 117 (3) (2007)
vessels for drug delivery, factors involved, and limitations and augmentation of 333–341.
the effect, Adv. Drug Deliv. Rev. 63 (3) (2011) 136–151. [42] C. Michiels, C. Tellier, O. Feron, Cycling hypoxia: a key feature of the tumor micro-
[12] H. Maeda, J. Takeshita, R. Kanamaru, A lipophilic derivative of neocarzinostatin. A environment, Biochim. Biophys. Acta 1866 (1) (2016) 76–86.
polymer conjugation of an antitumor protein antibiotic, Int. J. Pept. Protein Res. [43] H. Maeda, Toward a full understanding of the EPR effect in primary and metastatic
14 (2) (1979) 81–87. tumors as well as issues related to its heterogeneity, Adv. Drug Deliv. Rev. 91
[13] K. Greish, J. Fang, T. Inutsuka, A. Nagamitsu, H. Maeda, Macromolecular therapeu- (2015) 3–6.
tics: advantages and prospects with special emphasis on solid tumour targeting, [44] A. Casazza, C.G. Di, M. Wenes, V. Finisguerra, S. Deschoemaeker, M. Mazzone,
Clin. Pharmacokinet. 42 (13) (2003) 1089–1105. Tumor stroma: a complexity dictated by the hypoxic tumor microenvironment,
[14] M.E. Davis, Z.G. Chen, D.M. Shin, Nanoparticle therapeutics: an emerging treatment Oncogene 33 (14) (2014) 1743–1754.
modality for cancer, Nat. Rev. Drug Discov. 7 (9) (2008) 771–782. [45] L. Miao, L. Huang, Exploring the tumor microenvironment with nanoparticles, Can-
[15] F. Danhier, V. Préat, Strategies to improve the EPR effect for the delivery of anti- cer Treat. Res. 166 (2015) 193–226.
cancer nanomedicines, Cancer Cell Microenviron. 2 (2015), e808. [46] M.R. Kano, Y. Bae, C. Iwata, Y. Morishita, M. Yashiro, M. Oka, T. Fujii, A. Komuro, K.
[16] R.A. Petros, J.M. DeSimone, Strategies in the design of nanoparticles for therapeutic Kiyono, M. Kaminishi, K. Hirakawa, Y. Ouchi, N. Nishiyama, K. Kataoka, K. Miyazono,
applications, Nat. Rev. Drug Discov. 9 (8) (2010) 615–627. Improvement of cancer-targeting therapy, using nanocarriers for intractable solid tu-
[17] H. Ragelle, F. Danhier, V. Préat, R. Langer, D.G. Anderson, Nanoparticle-based mors by inhibition of TGF-beta signaling, Proc. Natl. Acad. Sci. U. S. A. 104 (9) (2007)
drug delivery systems: a commercial and regulatory outlook as the field ma- 3460–3465.
tures, Expert Opin. Drug Deliv. (2016). [47] M.R. Kano, Y. Komuta, C. Iwata, M. Oka, Y.T. Shirai, Y. Morishita, Y. Ouchi, K.
[18] T.M. Allen, A. Chonn, Large unilamellar liposomes with low uptake into the reticu- Kataoka, K. Miyazono, Comparison of the effects of the kinase inhibitors imatinib,
loendothelial system, FEBS Lett. 223 (1) (1987) 42–46. sorafenib, and transforming growth factor-beta receptor inhibitor on extravasation
[19] A.D. Bangham, M.M. Standish, J.C. Watkins, Diffusion of univalent ions across the of nanoparticles from neovasculature, Cancer Sci. 100 (1) (2009) 173–180.
lamellae of swollen phospholipids, J. Mol. Biol. 13 (1) (1965) 238–252. [48] A. Glentis, V. Gurchenkov, V.D. Matic, Assembly, heterogeneity, and breaching of
[20] R. Langer, J. Folkman, Polymers for the sustained release of proteins and other mac- the basement membranes, Cell Adhes. Migr. 8 (3) (2014) 236–245.
romolecules, Nature 263 (5580) (1976) 797–800. [49] R. Kalluri, Basement membranes: structure, assembly and role in tumour angio-
[21] M.B. Yatvin, W. Kreutz, B.A. Horwitz, M. Shinitzky, pH-sensitive liposomes: possi- genesis, Nat. Rev. Cancer 3 (6) (2003) 422–433.
ble clinical implications, Science 210 (4475) (1980) 1253–1255. [50] K. Yokoi, M. Kojic, M. Milosevic, T. Tanei, M. Ferrari, A. Ziemys, Capillary-wall colla-
[22] Y. Barenholz, Doxil(R)–the first FDA-approved nano-drug: lessons learned, J. Con- gen as a biophysical marker of nanotherapeutic permeability into the tumor micro-
trol. Release 160 (2) (2012) 117–134. environment, Cancer Res. 74 (16) (2014) 4239–4246.
[23] A.N. Gordon, J.T. Fleagle, D. Guthrie, D.E. Parkin, M.E. Gore, A.J. Lacave, Recurrent epithe- [51] S.K. Sriraman, B. Aryasomayajula, V.P. Torchilin, Barriers to drug delivery in solid
lial ovarian carcinoma: a randomized phase III study of pegylated liposomal doxorubi- tumors, Tissue Barriers 2 (2014), e29528.
cin versus topotecan, J. Clin. Oncol. 19 (14) (2001) 3312–3322. [52] I.A. Khawar, J.H. Kim, H.J. Kuh, Improving drug delivery to solid tumors: priming
[24] J.W. Nichols, Y.H. Bae, EPR: evidence and fallacy, J. Control. Release 190 (2014) the tumor microenvironment, J. Control. Release 201 (2015) 78–89.
451–464. [53] M. Egeblad, E.S. Nakasone, Z. Werb, Tumors as organs: complex tissues that inter-
[25] A. Sparreboom, C.D. Scripture, V. Trieu, P.J. Williams, T. De, A. Yang, B. Beals, W.D. face with the entire organism, Dev. Cell 18 (6) (2010) 884–901.
Figg, M. Hawkins, N. Desai, Comparative preclinical and clinical pharmacokinetics [54] K. Krukiewicz, J.K. Zak, Biomaterial-based regional chemotherapy: local anticancer
of a cremophor-free, nanoparticle albumin-bound paclitaxel (ABI-007) and pacli- drug delivery to enhance chemotherapy and minimize its side-effects, Mater. Sci.
taxel formulated in Cremophor (Taxol), Clin. Cancer Res. 11 (11) (2005) Eng. C Mater. Biol. Appl. 62 (2016) 927–942.
4136–4143. [55] M. Norouzi, B. Nazari, D.W. Miller, Injectable hydrogel-based drug delivery systems
[26] I.C. Henderson, V. Bhatia, Nab-paclitaxel for breast cancer: a new formulation with an for local cancer therapy, Drug Discov. Today (2016).
improved safety profile and greater efficacy, Expert. Rev. Anticancer. Ther. 7 (7) (2007) [56] R. Karim, C. Palazzo, B. Evrard, G. Piel, Nanocarriers for the treatment of glioblasto-
919–943. ma multiforme: current state-of-the-art, J. Control. Release 227 (2016) 23–37.
[27] K.F. Pirollo, E.H. Chang, Does a targeting ligand influence nanoparticle tumor local- [57] A. Burgess, K. Shah, O. Hough, K. Hynynen, Focused ultrasound-mediated drug de-
ization or uptake? Trends Biotechnol. 26 (10) (2008) 552–558. livery through the blood-brain barrier, Expert. Rev. Neurother. 15 (5) (2015)
[28] R.K. Jain, Delivery of molecular and cellular medicine to solid tumors, Adv. 477–491.
Drug Deliv. Rev. 46 (1–3) (2001) 149–168. [58] C. Bastiancich, K. Vanvarenberg, B. Ucakar, M. Pitorre, G. Bastiat, F. Lagarce, V. Préat, F.
[29] A. Verma, O. Uzun, Y. Hu, Y. Hu, H.S. Han, N. Watson, S. Chen, D.J. Irvine, F. Stellacci, Danhier, Lauroyl-gemcitabine-loaded lipid nanocapsule hydrogel for the treatment of
Surface-structure-regulated cell-membrane penetration by monolayer-protected glioblastoma, J. Control. Release 225 (2016) 283–293.
nanoparticles, Nat. Mater. 7 (7) (2008) 588–595. [59] F. Kiessling, M.E. Mertens, J. Grimm, T. Lammers, Nanoparticles for imaging: top or
[30] J. Hrkach, H.D. Von, A.M. Mukkaram, E. Andrianova, J. Auer, T. Campbell, W.D. De, M. flop? Radiology 273 (1) (2014) 10–28.
Figa, M. Figueiredo, A. Horhota, S. Low, K. McDonnell, E. Peeke, B. Retnarajan, A. [60] N. Schleich, F. Danhier, V. Préat, Iron oxide-loaded nanotheranostics: major obsta-
Sabnis, E. Schnipper, J.J. Song, Y.H. Song, J. Summa, D. Tompsett, G. Troiano, H.T. Van cles to in vivo studies and clinical translation, J. Control. Release 198 (2015) 35–54.
Geen, J. Wright, P. LoRusso, P.W. Kantoff, N.H. Bander, C. Sweeney, O.C. Farokhzad, R. [61] N. Ahmed, H. Fessi, A. Elaissari, Theranostic applications of nanoparticles in cancer,
Langer, S. Zale, Preclinical development and clinical translation of a PSMA-targeted do- Drug Discov. Today 17 (17–18) (2012) 928–934.
cetaxel nanoparticle with a differentiated pharmacological profile, Sci. Transl. Med. 4 [62] S.C. Baetke, T. Lammers, F. Kiessling, Applications of nanoparticles for diagnosis and
(128) (2012) 128ra39. therapy of cancer, Br. J. Radiol. 88 (1054) (2015) 20150207.
[31] C. Mamot, R. Ritschard, A. Wicki, G. Stehle, T. Dieterle, L. Bubendorf, C. Hilker, S. Deuster, [63] T. Lammers, L.Y. Rizzo, G. Storm, F. Kiessling, Personalized nanomedicine, Clin. Can-
R. Herrmann, C. Rochlitz, Tolerability, safety, pharmacokinetics, and efficacy of doxoru- cer Res. 18 (18) (2012) 4889–4894.
bicin-loaded anti-EGFR immunoliposomes in advanced solid tumours: a phase 1 dose- [64] L.Y. Rizzo, B. Theek, G. Storm, F. Kiessling, T. Lammers, Recent progress in
escalation study, Lancet Oncol. 13 (12) (2012) 1234–1241. nanomedicine: therapeutic, diagnostic and theranostic applications, Curr. Opin.
[32] T. Stylianopoulos, R.K. Jain, Design considerations for nanotherapeutics in oncolo- Biotechnol. 24 (6) (2013) 1159–1166.
gy, Nanomedicine 11 (8) (2015) 1893–1907. [65] C. Alexiou, W. Arnold, R.J. Klein, F.G. Parak, P. Hulin, C. Bergemann, W. Erhardt, S.
[33] Y. Matsumura, M. Gotoh, K. Muro, Y. Yamada, K. Shirao, Y. Shimada, M. Wagenpfeil, A.S. Lubbe, Locoregional cancer treatment with magnetic drug
Okuwa, S. Matsumoto, Y. Miyata, H. Ohkura, K. Chin, S. Baba, T. Yamao, A. targeting, Cancer Res. 60 (23) (2000) 6641–6648.
Kannami, Y. Takamatsu, K. Ito, K. Takahashi, Phase I and pharmacokinetic [66] S. Laurent, A.A. Saei, S. Behzadi, A. Panahifar, M. Mahmoudi, Superparamagnetic
study of MCC-465, a doxorubicin (DXR) encapsulated in PEG iron oxide nanoparticles for delivery of therapeutic agents: opportunities and chal-
immunoliposome, in patients with metastatic stomach cancer, Ann. Oncol. lenges, Expert Opin. Drug Deliv. 11 (9) (2014) 1449–1470.
15 (3) (2004) 517–525. [67] B. Polyak, I. Fishbein, M. Chorny, I. Alferiev, D. Williams, B. Yellen, G. Friedman, R.J. Levy,
[34] J.E. Zuckerman, I. Gritli, A. Tolcher, J.D. Heidel, D. Lim, R. Morgan, B. Chmielowski, A. High field gradient targeting of magnetic nanoparticle-loaded endothelial cells to the
Ribas, M.E. Davis, Y. Yen, Correlating animal and human phase Ia/Ib clinical data with surfaces of steel stents, Proc. Natl. Acad. Sci. U. S. A. 105 (2) (2008) 698–703.
CALAA-01, a targeted, polymer-based nanoparticle containing siRNA, Proc. Natl. Acad. [68] N. Schleich, C. Po, D. Jacobs, B. Ucakar, B. Gallez, F. Danhier, V. Préat, Comparison of
Sci. U. S. A. 111 (31) (2014) 11449–11454. active, passive and magnetic targeting to tumors of multifunctional paclitaxel/
[35] T. Flynn, C. Wei, The pathway to commercialization for nanomedicine, Nanomedicine 1 SPIO-loaded nanoparticles for tumor imaging and therapy, J. Control. Release 194
(1) (2005) 47–51. (2014) 82–91.
[36] J.W. Nichols, Y.H. Bae, Odyssey of a cancer nanoparticle: from injection site to site [69] M.I. Koukourakis, S. Koukouraki, A. Giatromanolaki, S. Kakolyris, V. Georgoulias, A.
of action, Nano Today 7 (6) (2012) 606–618. Velidaki, S. Archimandritis, N.N. Karkavitsas, High intratumoral accumulation of
[37] R.J. Gillies, P.A. Schornack, T.W. Secomb, N. Raghunand, Causes and effects of het- stealth liposomal doxorubicin in sarcomas–rationale for combination with radio-
erogeneous perfusion in tumors, Neoplasia 1 (3) (1999) 197–207. therapy, Acta Oncol. 39 (2) (2000) 207–211.
[38] R.K. Jain, Transport of molecules in the tumor interstitium: a review, Cancer Res. 47 [70] V.P. Chauhan, Z. Popovic, O. Chen, J. Cui, D. Fukumura, M.G. Bawendi, R.K. Jain, Fluo-
(12) (1987) 3039–3051. rescent nanorods and nanospheres for real-time in vivo probing of nanoparticle
[39] A.W. Nichols, Sports medicine and the Americans with Disabilities Act, Clin. J. Sport shape-dependent tumor penetration, Angew. Chem. Int. Ed. Engl. 50 (48) (2011)
Med. 6 (3) (1996) 190–195. 11417–11420.
[40] J.I. Hare, T. Lammers, M.B. Ashford, S. Puri, G. Storm, S.T. Barry, Challenges and [71] Y. Geng, P. Dalhaimer, S. Cai, R. Tsai, M. Tewari, T. Minko, D.E. Discher, Shape effects
strategies in anti-cancer nanomedicine development: an industry perspective, of filaments versus spherical particles in flow and drug delivery, Nat. Nanotechnol.
Adv. Drug Deliv. Rev. (2016). 2 (4) (2007) 249–255.
[41] T. Lammers, P. Peschke, R. Kuhnlein, V. Subr, K. Ulbrich, J. Debus, P. Huber, W. Hennink, [72] H. Maeda, J. Wu, T. Okamoto, K. Maruo, T. Akaike, Kallikrein-kinin in infection and
G. Storm, Effect of radiotherapy and hyperthermia on the tumor accumulation of cancer, Immunopharmacology 43 (2–3) (1999) 115–128.
F. Danhier / Journal of Controlled Release 244 (2016) 108–121 121

[73] H. Maeda, Y. Noguchi, K. Sato, T. Akaike, Enhanced vascular permeability in solid interstitial fluid pressure and improves oxygenation in breast cancers in patients
tumor is mediated by nitric oxide and inhibited by both new nitric oxide scavenger treated with neoadjuvant chemotherapy: clinical implications, J. Clin. Oncol. 23
and nitric oxide synthase inhibitor, Jpn. J. Cancer Res. 85 (4) (1994) 331–334. (9) (2005) 1951–1961.
[74] A.F. Chambers, L.M. Matrisian, Changing views of the role of matrix metallopro- [92] R. Ansiaux, C. Baudelet, B.F. Jordan, N. Beghein, P. Sonveaux, W.J. De, P. Martinive, V.
teinases in metastasis, J. Natl. Cancer Inst. 89 (17) (1997) 1260–1270. Gregoire, O. Feron, B. Gallez, Thalidomide radiosensitizes tumors through early
[75] H.R. Reichman, C.L. Farrell, R.F. Del Maestro, Effects of steroids and nonsteroid anti- changes in the tumor microenvironment, Clin. Cancer Res. 11 (2 Pt 1) (2005)
inflammatory agents on vascular permeability in a rat glioma model, J. Neurosurg. 743–750.
65 (2) (1986) 233–237. [93] J. Segers, V. F. Di, R. Ansiaux, P. Martinive, O. Feron, P. Wallemacq, B. Gallez, Poten-
[76] H. Maeda, J. Wu, T. Sawa, Y. Matsumura, K. Hori, Tumor vascular permeability and tiation of cyclophosphamide chemotherapy using the anti-angiogenic drug thalid-
the EPR effect in macromolecular therapeutics: a review, J. Control. Release 65 (1– omide: importance of optimal scheduling to exploit the ‘normalization’ window of
2) (2000) 271–284. the tumor vasculature, Cancer Lett. 244 (1) (2006) 129–135.
[77] F. Danhier, A. Le Breton, V. Préat, RGD-based strategies to target alpha(v) beta(3) [94] R.T. Tong, Y. Boucher, S.V. Kozin, F. Winkler, D.J. Hicklin, R.K. Jain, Vascular normal-
integrin in cancer therapy and diagnosis, Mol. Pharm. 9 (11) (2012) 2961–2973. ization by vascular endothelial growth factor receptor 2 blockade induces a pres-
[78] F. Nussenbaum, I.M. Herman, Tumor angiogenesis: insights and innovations, J. sure gradient across the vasculature and improves drug penetration in tumors,
Oncol. 2010 (2010) 132641. Cancer Res. 64 (11) (2004) 3731–3736.
[79] M.L. Salgaller, Technology evaluation: bevacizumab, Genentech/Roche, Curr. Opin. [95] A. Pluen, Y. Boucher, S. Ramanujan, T.D. McKee, T. Gohongi, E. di T., E.B. Brown, Y.
Mol. Ther. 5 (6) (2003) 657–667. Izumi, R.B. Campbell, D.A. Berk, R.K. Jain, Role of tumor-host interactions in inter-
[80] R.K. Jain, Normalization of tumor vasculature: an emerging concept in stitial diffusion of macromolecules: cranial vs. subcutaneous tumors, Proc. Natl.
antiangiogenic therapy, Science 307 (5706) (2005) 58–62. Acad. Sci. U. S. A. 98 (8) (2001) 4628–4633.
[81] F. Danhier, P. Danhier, C.J. De Saedeleer, A.C. Fruytier, N. Schleich, R.A. des, P. [96] N. Kuriyama, H. Kuriyama, C.M. Julin, K.R. Lamborn, M.A. Israel, Protease pretreat-
Sonveaux, B. Gallez, V. Préat, Paclitaxel-loaded micelles enhance transvascular per- ment increases the efficacy of adenovirus-mediated gene therapy for the treat-
meability and retention of nanomedicines in tumors, Int. J. Pharm. 479 (2) (2015) ment of an experimental glioblastoma model, Cancer Res. 61 (5) (2001)
399–407. 1805–1809.
[82] C.G. Lee, M. Heijn, T.E. di, G. Griffon-Etienne, M. Ancukiewicz, C. Koike, K.R. Park, N. [97] G. Alexandrakis, E.B. Brown, R.T. Tong, T.D. McKee, R.B. Campbell, Y. Boucher, R.K.
Ferrara, R.K. Jain, H.D. Suit, Y. Boucher, Anti-vascular endothelial growth factor Jain, Two-photon fluorescence correlation microscopy reveals the two-phase na-
treatment augments tumor radiation response under normoxic or hypoxic condi- ture of transport in tumors, Nat. Med. 10 (2) (2004) 203–207.
tions, Cancer Res. 60 (19) (2000) 5565–5570. [98] T.T. Goodman, P.L. Olive, S.H. Pun, Increased nanoparticle penetration in collage-
[83] A.V. Salnikov, V.V. Iversen, M. Koisti, C. Sundberg, L. Johansson, L.B. Stuhr, M. nase-treated multicellular spheroids, Int. J. Nanomedicine 2 (2) (2007) 265–274.
Sjoquist, H. Ahlstrom, R.K. Reed, K. Rubin, Lowering of tumor interstitial fluid pres- [99] L. Eikenes, M. Tari, I. Tufto, O.S. Bruland, D.C. de Lange, Hyaluronidase induces a
sure specifically augments efficacy of chemotherapy, FASEB J. 17 (12) (2003) transcapillary pressure gradient and improves the distribution and uptake of lipo-
1756–1758. somal doxorubicin (Caelyx) in human osteosarcoma xenografts, Br. J. Cancer 93
[84] C. Bouzin, O. Feron, Targeting tumor stroma and exploiting mature tumor vascula- (1) (2005) 81–88.
ture to improve anti-cancer drug delivery, Drug Resist. Updat. 10 (3) (2007) [100] R. Masterson, T.D. Hewitson, K. Kelynack, M. Martic, L. Parry, R. Bathgate, I. Darby,
109–120. G. Becker, Relaxin down-regulates renal fibroblast function and promotes matrix
[85] J.M. Yingling, K.L. Blanchard, J.S. Sawyer, Development of TGF-beta signalling inhib- remodelling in vitro, Nephrol. Dial. Transplant. 19 (3) (2004) 544–552.
itors for cancer therapy, Nat. Rev. Drug Discov. 3 (12) (2004) 1011–1022. [101] T. Stylianopoulos, C. Wong, M.G. Bawendi, R.K. Jain, D. Fukumura, Multistage nano-
[86] G. Griffon-Etienne, Y. Boucher, C. Brekken, H.D. Suit, R.K. Jain, Taxane-induced ap- particles for improved delivery into tumor tissue, Methods Enzymol. 508 (2012)
optosis decompresses blood vessels and lowers interstitial fluid pressure in solid 109–130.
tumors: clinical implications, Cancer Res. 59 (15) (1999) 3776–3782. [102] L. Zhu, P. Kate, V.P. Torchilin, Matrix metalloprotease 2-responsive multifunctional
[87] S.H. Jang, M.G. Wientjes, J.L. Au, Enhancement of paclitaxel delivery to solid tumors liposomal nanocarrier for enhanced tumor targeting, ACS Nano 6 (4) (2012)
by apoptosis-inducing pretreatment: effect of treatment schedule, J. Pharmacol. 3491–3498.
Exp. Ther. 296 (3) (2001) 1035–1042. [103] K.N. Sugahara, T. Teesalu, P.P. Karmali, V.R. Kotamraju, L. Agemy, O.M. Girard, D.
[88] J. Wang, Z. Lu, Y. Gao, M.G. Wientjes, J.L. Au, Improving delivery and efficacy of Hanahan, R.F. Mattrey, E. Ruoslahti, Tissue-penetrating delivery of compounds
nanomedicines in solid tumors: role of tumor priming, Nanomedicine (London) and nanoparticles into tumors, Cancer Cell 16 (6) (2009) 510–520.
6 (9) (2011) 1605–1620. [104] K.N. Sugahara, T. Teesalu, P.P. Karmali, V.R. Kotamraju, L. Agemy, D.R. Greenwald, E.
[89] F. Danhier, P. Danhier, N. Magotteaux, P.G. De, B. Ucakar, O. Karroum, B. Jordan, B. Ruoslahti, Coadministration of a tumor-penetrating peptide enhances the efficacy
Gallez, V. Préat, Electron paramagnetic resonance highlights that the oxygen effect of cancer drugs, Science 328 (5981) (2010) 1031–1035.
contributes to the radiosensitizing effect of paclitaxel, PLoS One 7 (7) (2012), [105] R. Tong, R. Langer, Nanomedicines targeting the tumor microenvironment, Cancer
e40772. J. 21 (4) (2015) 314–321.
[90] F. Danhier, N. Magotteaux, B. Ucakar, N. Lecouturier, M. Brewster, V. Préat, Novel [106] D. Liu, F. Yang, F. Xiong, N. Gu, The smart drug delivery system and its clinical po-
self-assembling PEG-p-(CL-co-TMC) polymeric micelles as safe and effective deliv- tential, Theranostics 6 (9) (2016) 1306–1323.
ery system for paclitaxel, Eur. J. Pharm. Biopharm. 73 (2) (2009) 230–238. [107] S. Mura, J. Nicolas, P. Couvreur, Stimuli-responsive nanocarriers for drug delivery,
[91] A.G. Taghian, R. Abi-Raad, S.I. Assaad, A. Casty, M. Ancukiewicz, E. Yeh, P. Molokhia, Nat. Mater. 12 (11) (2013) 991–1003.
K. Attia, T. Sullivan, I. Kuter, Y. Boucher, S.N. Powell, Paclitaxel decreases the

You might also like