Вы находитесь на странице: 1из 7

Materials Science & Engineering C 106 (2020) 110302

Contents lists available at ScienceDirect

Materials Science & Engineering C


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

Grand challenges in nanomedicine T


a,∗,1 b,1 c
Lin-Ping Wu , Danyang Wang , Zibiao Li
a
Drug Discovery Pipeline, Hefei Institute of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences,
Guangzhou, 510530, China
b
Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen,
Denmark
c
Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore

A R T I C LE I N FO A B S T R A C T

Keywords: Nanotherapeutics and nanopharmaceuticals could achieve and facilitate earlier and more precise individual
Nanomedicine diagnosis, improve targeted therapies, reduce side effects, and enhance therapeutic monitoring. These ad-
Nanoparticles vantages will improve quality of life, support a healthier and more independent aging population, and be in-
Drug delivery strumental in maximizing the cost-effectiveness of health care. However, the field of nanomedicine is at its early
Translation medicine
stage, most of the research still stays in the laboratory phase, and few success stories are translated into clinical
Diagnostics
trials and medical practice. This review will demonstrate the numerous challenges that are encountered during
the development of commercial nanoparticle-based therapeutics and the possible solutions.

1. Introduction and reproducible scale-up and manufacturing process to achieve a


consistent product with stable physicochemical characteristics, biolo-
In recent years, nanotechnology has been increasingly applied to the gical behaviors, and pharmacological profiles. The safety and reg-
area of medicine, which is defined as nanomedicine, as a new in- ulatory issues of nanomedicine need additional considerations com-
dependent field of life sciences [1,2]. Nanomedicine has the potential pared with conventional medicines. This review seeks to summary
advantages to overcome biological barriers, effectively deliver hydro- challenges and limitations during the development and commerciali-
phobic drugs and biologics, and preferentially target sites of disease zation of nanomedicine products and discusses potential solutions to
[3,4]. However, only a relatively small number of nanoparticle-based accelerate the growth of this important field.
medicines have been approved for clinical use because of numerous
challenges and hurdles at different stages of development [5,6]. The 2. Challenges and current limitations
first FDA-approved nano-drug is Doxil® in 1995 (Fig. 1), which can
prolong drug circulation time and is avoidance of the RES due to the use Successfully translating nanomedicine from pre-clinical proof of
of PEGylated nano-liposomes. What's more important, doxorubicin is concept to demonstration of therapeutic value in the clinic is still
highly and stably remote loaded in Doxil® using a transmembrane challenging, several obstacles have been identified as top scientific
ammonium sulfate gradient method, which also allows for drug release hurdles in bringing nanoengineered products to patients (Fig. 2).
at the tumor [7]. The promising nanomedicine approved by FDA or in
the clinical trials has been summarized by lots of reviews [8–11]. From 2.1. Challenges of biological barriers and specific targeting
1995 to 2017, 50 nano-pharmaceuticals have received FDA approval
and are currently available for clinical use [10]. Also as of August 2019, Before the drugs successfully reach the disease sites, the nano-for-
64 clinical trials including the term “nano” were listed as “recruiting” mulation has to help them to cross multiple biological barriers (Fig. 3)
or “active” on ClinicalTrials.gov. The attractiveness of nanomedicine [6,12]. For example, oral nanoparticles need to have high stability in
applications lies in the unique characteristics of three-dimensional the gastrointestinal tract, the ability to penetrate intestinal epithelium
constructs with multiple components in nanoscale. At the same time, and the ability to keep the high systemic bioavailability of drugs after
due to its complexity, nanomedicine product requires careful design crossing several barriers [12,13].
and engineering, strict characterization of physicochemical properties, Compared with most of the small molecules delivered orally, large


Corresponding author.
E-mail address: wu_linping@gibh.ac.cn (L.-P. Wu).
1
Contributed equally.

https://doi.org/10.1016/j.msec.2019.110302
Received 25 August 2018; Received in revised form 10 October 2019; Accepted 10 October 2019
Available online 12 October 2019
0928-4931/ © 2019 Elsevier B.V. All rights reserved.
L.-P. Wu, et al. Materials Science & Engineering C 106 (2020) 110302

Fig. 1. The first FDA-approved nano-drug Doxil®.


(A) Doxil® vial as sold by Sequus Pharmaceuticals
(starting 1996), (B) cryo-TEMs of commercial
Doxil, which is doxorubicin sulfate remote loaded
pegylated nano-liposomes, (C) a sterically stabi-
lized (PEGylated) liposome, shows lack of inser-
tion of plasma opsonin into its membrane (Reprint
from Ref. [7]).

Fig. 2. The challenges of safe translation nanomedicine products for diagnostics and treatment in clinic.

Fig. 3. Typical extremely effective biological


barriers largely prevent nanoparticles from
reaching their intended destinations. The barriers
include the reticuloendothelial system, en-
dothelial/epithelial membranes, complex net-
works of blood vessels, abnormal flow of blood,
and interstitial pressure gradients. Inset: A mag-
nified region of a tumor cell offers additional
biological challenges: cellular/nuclear membranes
and ionic/molecular pumps that can expel drugs
from the cancerous cells creating drug resistance
(Reprint from Ref. [6]).

molecular drugs, such as peptides, proteins, and polynucleotides, nanoparticles move across the BBB, as shown in Fig. 4 [15]. Another
usually adopt intravenous administration (IV) which remains the most major challenge of nanomedicine is to deliver therapeutic reagents into
direct and efficient route to deliver therapeutic reagents [14]. After IV solid tumors [18,19]. Although tumor vasculature is high hetero-
injection, drugs in circulation still need to overcome several biological geneous in distribution and more permeable in some places, large areas
barriers to reach their targets. For the treatment of central nervous of tumors with high cancer cell density and dense tumor stroma is still
system related disease, the blood-brain barrier (BBB) restricts the dif- poor perfused which further hamper the distribution of drugs in tumors
fusion of 99% large or hydrophilic molecules into the cerebrospinal [20]. Increased interstitial fluid pressure (IFP) caused by impaired
fluid and is a major obstacle for brain targeting delivery [15–17]. There lymphatic drainage in tumors [21,22] adds another barrier to delivery,
are various possible transport pathways by which solute molecules and which is one of the main factors contributing to reduce extravasation

2
L.-P. Wu, et al. Materials Science & Engineering C 106 (2020) 110302

Fig. 4. Transport pathways across BBB (Reprint from Ref. [15]).

and transvascular transport of drugs despite the leaky tumor micro- (EGFR), decorate the surface of nanoparticles to increase cellular in-
vasculature, and restrain the transport of molecules into interstitial ternalization of the reagents through endocytosis and improve the ef-
space of tumor [23]. Facing these challenges, multiple nanoparticle- ficacy of systemic anticancer therapy [33,34]. In addition, nano-
based strategies from biomaterials, including liposome, polymer mi- particles incorporating with protein transduction domain (PTD) and
celle, and peptidic or protein nanoparticles, are investigated to deliver cell penetrating peptides (CPPs), a class of small cationic peptides
drugs [24–26]. Because of the high permeability of tumor vasculature (< 20 amino acid), such as oligoarginine and TAT, enable the uniform
and lack of proper lymphatic drainage, the enhanced permeability and transport of large, biologically functional molecules that otherwise
retention effect (EPR) enhance the passive accumulation of nano- cannot effectively enter cancer cells [35,36]. Moreover, another pos-
particles and macromolecules in the tumor microenvironment and im- sible target of nanomedicine is tumor endothelial cells. For example,
prove tumor drug delivery [22]. First generation nanomedicine on the the cyclic and linear derivatives of oligopeptides RGD (Arg-Gly-Asp)
market is based on passive targeting, such as pegylated liposomal which bind to the integrins α2β3, αvb3, and α5β1, provide a tumor-
doxorubicin (Doxil®/Caelyx®) [7,27] and nab-paclitaxel (Abraxane®) penetrating function to polymers, liposomes and other nanoparticles.
[28]. The key issue of passively targeted first generation nanomedicine With RGD modification, nanoparticles deliver cytotoxic reagents to
is how to control the pharmacokinetics and biodistribution of nano- tumor tissue and achieve significant antitumor effects [37–39]. The
particles by modulating its physicochemical properties [29]. active targeting depends on the affinity and efficacy of a target and its
Even though there are some advantages of the passive process, the specific ligand. Besides, it is very important to optimize the density of
utilization of an active biological transport process can increase effi- targeting ligands per nanocarrier to achieve not only high targeting
cacy, decrease side effects, and reduce systemic drug exposures, which efficiency but also to ensure an optimal internalization [40].
further improve drug delivery [30]. The organized structures of active
targeting nanoparticles allow for the incorporation with various tar-
2.2. Analysis and characterization of nano-formulations
geting moieties including small molecular ligands for receptors, pep-
tides, proteins, antibodies, aptamers, and oligonucleotides. It can en-
Compared with traditional pharmaceuticals, nanomedicine is a
hance drug delivery to the target sites, reduce off-target organ
three-dimensional complex composed of several components and each
toxicities, and facilitate cellular uptake of therapeutic agents [31,32].
component serves to a specific function [41]. Therefore, it requires
Usually the ligands or monoclonal antibodies targeting to the surface
more sophisticated and appropriate analytical tests to fully detect,
receptors overexpressed by cancer cells, such as transferrin receptor
characterize and quantify each component and also evaluate the re-
(TfR), folate receptor (FR) and epidermal growth factor receptor
lationships and interactions between these components including both

3
L.-P. Wu, et al. Materials Science & Engineering C 106 (2020) 110302

physicochemical properties and biological behaviors [28]. But it is one dimensional complex products with specific arrangement of multi-
of the challenges in the development of nanomedicine not only the component, which make the chemistry, manufacturing, and control
technical aspect but also the regulatory perspective. In general, the process very demanding. And traditional pharmaceutical manu-
most important physicochemical features of nanomedicine are compo- facturing does not have rich experience to create nanoscale three-di-
sition, structure, size, surface properties, porosity, charge, and stability mensional multicomponent systems. All of these make a series of blocks
[42,43]. It is difficult to characterize nanomedicine products because of for the scale-up and manufacturing of nanomedicine. An example is in
their variability properties. Taking polydispersity (PD) as an example, it the production of nanomedical therapeutics Doxil®, the first FDA-ap-
is a key parameter that measures the heterogeneity of particles with proved nano-drug (1995) [7,62,63], which had to be suspended in
regard to size, shape, or mass. If the nanomedicine product with the November 2011 because of its manufacturing and sterility issues. The
same average size but different PD, it may cause dramatic changes in shortages of Doxil® were overcome until 2014, and an alternate man-
secondary properties such as targeting properties, drug release rate, ufacturing approach for Doxil® was adopted resulting in increased
biocompatibility, toxicity, and in vivo behaviors [44–47]. Another medication costs and delayed therapies for patients [28].
challenge is the characterization of stability and storage of nanomedi- A well understanding and identifying the components and their
cine. Because of the excellent features of biodegradable materials, such interactions in the early development help define the key characteristics
as lipid and polymer, they have been widely used in the development of greatly, such as the critical steps and analytical criteria, to ensure re-
nanomedicine products. But this biodegradable property of nanoma- producibility of the product during the larger-scale manufacturing. In
terials would also alter the properties of nanomedicine products during general, there are two categories of nanoparticle preparation methods,
the storage either in the solutions or in a lyophilized powder form “top-down” and “bottom-up” approaches [64]. Top-down methods
[48,49]. So it is substantial to improve quality assessment of the bio- manufacture nano-entities from larger ones, such as grinding of parti-
degradable materials by well-defined and reproducible standards. An- cles using the milling technique. In contrast, bottom-up methods ar-
other issue is detecting and visualizing the in vivo biodistribution of range smaller components into functional super-assemblies, such as
nanocarriers over time. After administration, nanocarriers or their de- polymerization of monomers or molecular self-assembly to arrange
gradable soft components would reach biological fluids and may in- single/multi-molecule components into a useful nanostructure by nat-
teract with these biological fluids (e.g. blood serum) or biomolecules ural driving [65]. During the nanomedical formulation process, the
(e.g. proteins), which could significantly change their physicochemical high-speed homogenization, sonication, milling, emulsification, cross-
properties as well, such as size, drug loading, release profile and ag- linking, evaporation of organic solvents, centrifugation, filtration, and
gregation or agglomeration, and alter the function of nanomedicine in lyophilization are always employed. It is very important to consider
biological systems [50–53]. It is essential to fully characterize the na- which approaches and parameters are suitable to scale up at the lab- or
nomedicine products under clinically relevant conditions using in vitro small scale stage and these crucial process conditions will benefit later
and in vivo models [54]. development process. These parameters may involve the ratio of na-
Although pictorial biodistribution of nanoparticle accumulation in nomaterials, drugs and targeting moieties, the type of organic solvent
vivo can be obtained by fluorescence or radiolabelling method [55–57], and emulsifier/stabilizer/crosslinker, the oil-to-water phase ratio,
it is still very hard to quantify mass-balance information which is temperature, pressure, and pH [65]. Choosing the wrong conditions
paramountly important to account the full administered dose and en- could lead to altering the chemical structure of the therapeutic re-
sure safety [58]. Usually the fluorescence and radiolabeling emitters agents, other accessories, and unpredictable impurities. Particularly, it
chemically conjugated to one composition of nanoparticles, which is is very easy to change chemical structure and conformation of macro-
unstable and easily degraded in the internal environment. Therefore, it molecules (protein, peptide) by denaturation, crosslinking, coagulation,
may lead to unreliable results by tracing the degraded moiety of and degradation. Thus, the manufacturing of nanomedicine products is
fluoresce or radio molecule instead of the whole nanoparticle. Ad- not a simple addition and mixture of individual components, while
ditionally, the distribution of fluorescent or radio-modified nano- well-defined production steps, accurate analysis and strict quality
particles may have different properties in comparison with the same control are required [66].
nanoparticles without fluorescence or radio-modification. Last but not The high costs of the raw materials and the need for a cockamamie
least, nanomedicine with more than one composition and the ability to multistep production process are also obstacles for the scale-up and
carry and deliver multiple therapeutic and imaging agents may require manufacturing of nanomedicine, which makes the production of nano-
individual tracking [59]. therapeutics very expensive. This may hinder pharmaceutical compa-
Consequently, multiple standard characterization methods in- nies from carrying out the large-scale production of nanocarriers. For
cluding the quantification of active and inactive ingredients and im- instance, the expense of manufacturing commercial nano-formulation
purities, visualization of nanoparticles by microscopy, measurement of drugs such as Abraxane® and Doxil® is far more than the cost of their
particle size and size distribution with light scattering, and new ad- free drug paclitaxel and doxorubicin, receptively [67]. Therefore, in
vanced techniques specifically for the characterization of nanoparticle order to compensate for the high costs on the development and man-
in vivo behaviors are necessary to ensure that the nanomedicine pro- ufacture of nanomedicine products [68], the clinical therapeutic effect
ducts have all the desired properties for the intended therapeutic pur- of nanomedicine drugs has to be much more advanced than conven-
pose on a batch-to-batch basis with fewer side-effects [28]. tional therapeutics.

2.3. Scale-up and manufacturing 2.4. Pharmacology and safety challenges

The key issue for successful scale-up and manufacturing of nano- Physicochemical characteristics of nanomedicine notably influence
medicine is controlling and keeping constant stability of physico- the pharmacological and safety profiles. Even subtle changes in com-
chemical properties on a batch-to-batch basis [60], which is the most position and small deviations in the final manufactured products could
challenging problem in pharmaceutical development. result in significant changes in pharmacology and toxicity of nanome-
Usually, preclinical and early clinical studies just need a small dicine [28,66].
amount of nanomedicine products. Small-scale processes may achieve One of the requirements of a successful nanomedicine is to achieve
reproducibility with well-characterized nanoparticles. However, in the desired pharmacological profile, for example pharmacokinetic
large-scale production, the polydispersity of the nanomaterial is diffi- parameter (PK) [57]. However, most of the researcher and pharma-
cult to control, which could cause batch-to-batch variations of physical ceutical companies apply the standard criteria of small molecule to the
and chemical properties [61]. Since most nanoparticles are three- assessment of nanomedicine. Usually, small molecular drugs alone

4
L.-P. Wu, et al. Materials Science & Engineering C 106 (2020) 110302

diffuse more readily through biological barriers and the drug con- materials with at least one dimension between 1 and 100 nm. Also, the
centration in blood is somewhat in equilibrium and related to achiev- FDA and European Technology Platform on Nanomedicine (ETPN) in-
able target tissue levels [28,66]. Thus, the measurement of drug con- tends to collaborate with the Nanotechnology Characterization La-
centration in the blood or plasma used as the standard approach to boratory (NCL) and European Nano-Characterization Laboratory (EU-
determine PK is reasonable. But if apply this methodology to nano- NCL) respectively to promote the regulatory review and in-depth
medicine, it cannot be assumed to be correct and could be inherently characterization of nanomedicine products [83]. The urgent demand
flawed. Accordingly, different PK approaches with different indications for regulatory agencies in nanomedicine therapeutics is to refine and
are required for different nanomedicine products. For example, instead standardize requirements for the approval of safe nanomedicine pro-
of the standard test of the behavior in plasma, the pharmacological ducts. Also, more advanced and multifunctional tools need to be de-
parameters at the specific target site could be more relevant to evaluate veloped, even though the complexity of the approval process could be
the therapeutic activity of nanomedicine and the reproducibility. Fur- increased.
thermore, the bioequivalency of nanotechnology-based products might
use these methods to evaluate as well.
In recent years, nanoparticles have been wide-spread used in med- 3. Solution and conclusions
icine, which makes it necessary to address toxicity issues for human
health [69,70]. The nanoscale dimension of nanomedicine products can In summary, nanoparticles have made major contributions to clin-
simulate the intracellular organelles or biomolecules such as protein, ical medicine in the areas of therapeutics and diagnosis, and several
polysaccharide, and enzyme involved in cell signaling, which may lead innovations for example stealth and long circulating liposome are by
to detrimental biological interactions. Now the nanotoxicology is an now well-established and transfer into clinical applications, newer
independent field of research [45,71]. And more and more data re- technologies continue to emerge following the same basic concepts of
garding the toxicity of different nanoparticles are becoming available. design. But from the therapeutic angle, nanomedicine is still in infancy
Nonetheless, it remains difficult to evaluate the toxicity of nano- and need to overcome several hindrances before it is approved for
materials, especially in vivo and long-term toxicity. At present, the as- marketing [6]. Firstly, it needs to design and engineer nanostructures
sessment of nanoparticle toxicity using classical drug toxicity assays with appropriate physicochemical properties and biological behaviors,
may be inadequate. And there is no standard list of required tests. Thus, optimize reproducible manufacturing processes, select reliable analysis
the development of advanced complementary assays and the standard methods for adequate characterization. For example in the size esti-
criterion for toxicity evaluation of nanomedicine products become mation, dynamic light scattering (DLS) is widely used for the size
emergent [72,73]. There are multiple properties, such as size, shape, analysis of nanoparticles in liquid solution [84]. The average particle
surface area, surface charge, porosity, or hydrophobicity, which affect size from DLS analysis is heavily biased towards small numbers of large
the behavior and performance of nanomedicine drugs at the nano-bio particles because the intensity of light scattered from a nanoparticle
interface. Hence, each nanomedicine product may have its own case by varies as a function of radius to the power of six for a Rayleigh particle
case issues and require particular evaluations. Usually, the toxicity in [84]. NTA (nanoparticle tracking analysis) is an alternative modality to
pharmaceutical industry refers to acute toxicity and chronic toxicity. DLS, which is gaining popularity [85]. Unlike DLS, NTA does not use
Acute toxicity generally includes hemolysis, oxidative stress, in- the intensity of light scattered as a measure, but measures the particle
flammation, impaired mitochondrial function [74], or complement size from the changing position of scattering diffusing particles through
activation [75–77]. Investigation of chronic toxicity is time-consuming, image analysis. This process overcomes inherent problems with DLS
and analyzing chronic toxicity data is more demanding. and can be used for monitoring polydisperse samples as well as ag-
Due to the lack of suitable animal model, immunotoxicity cannot gregates [85]. Fig. 5 shows a representative example of NTA analysis,
readily carry over from preclinical testing to humans. Nanoparticles demonstrating population changes that have occurred on coating a
themselves and the biologics such as proteins, peptides, antibody polymeric nanoparticle suspension with polyethylenimine [26]. Also,
fragments, and nucleic acids in nanoparticles can serve as antigens the nanomedicine products should have favorable pharmacology and
sources that can elicit the immune response. The immunogenicity of toxicity profile, and demonstrate safe and efficiency in clinical trials. To
nanoparticles can be affected by their physicochemical properties, such make this happen and get the attraction for pharma development,
as size, surface characteristics, charge, hydrophobicity, and solubility. collaboration is a key factor to the future success of nanomedicines. The
development of nanomedicine is a multidisciplinary endeavor, and it is
2.5. Regulatory challenges necessitates highly varied expertise to develop translatable therapeutics
because of the diversity and complexity. It is essential to the successful
Although the FDA and European Medicines Agency (EMA) approved cooperation from the experts of biology, chemistry, nanotechnology,
a number of nanomedicine products for cancer therapy, there are not imaging, and medicine. It is important to foster partnerships between
specifically implemented guidelines for drug products containing soft large pharma, smaller companies, and academia early in preclinical
materials by FDA, EMA, and other regulatory bodies yet. The lack of development to capitalise on the unique strengths of each partner. Ef-
guidance in the examination of nanomedicine, nanomedicine ther- fective integration of resources from academia, industry, consortia, and
apeutics can only make regulatory decisions based on individual as- hospitals will be essential to help the link of physico-chemical proper-
sessment of benefits and risks [66]. In this way, the regulatory process ties of nanomedicine systems with biological implications. Finally,
is time-consuming and requires the high-level expert in innovative health authorities should develop and consummate definitions, quality
technologies, which may result in regulatory delays. And regulatory standards, guidelines and regulatory issues of nanomedicine. Although
issues are important for the development of cutting-edge technologies any new drug has to face similar hurdles conceptually, the particular
to characterize and monitor the quality of nanomedicine products be- complexity and multicomponent nature of nanomedicine lead to a large
side clinical trials and the approval process. Especially there is an ur- number of additional variables that may substantially increase more
gent need for detailed guidelines for characterization and quality con- considerations in controlling processes and monitoring of behavior in a
trol of nanomedicine products [78]. The good thing is that the biological system. Through rational designed and systematic ap-
definitions, guidelines, and cooperation in this area are gradually es- proaches to meet these challenges and solve the problems, nanomedi-
tablished and improved [79–81]. For example, the guidance for in- cine will forward to a new step and provide realistic and significant
dustry entitled “Considering Whether an FDA-Regulated Product In- value to human medicine and healthcare.
volves the Application of Nanotechnology” is released by the FDA in
June 2014 [82], where the definition of nanomaterials is engineered

5
L.-P. Wu, et al. Materials Science & Engineering C 106 (2020) 110302

Fig. 5. Characteristics of engineered PHBHHx nanoparticles before and after PEI coating. Typical size distribution profile of PHBHHx nanoparticles before (A) and
after PEI coating (B) as a function of particle concentration determined by nanoparticle tracking analysis. The insets represent an example of the real-time capture
video frame of point scatters. (C, D) 2D plots of relative light scattering intensity of particles versus the estimate of the particle size (Reprint from Ref. [26]).

Declaration of competing interestCOI 85–98.


[9] F. Dilnawaz, S. Acharya, S.K. Sahoo, Recent trends of nanomedicinal approaches in
clinics, Int. J. Pharm. 538 (2018) 263–278.
The authors declare that they have no conflict of interest. [10] C.L. Ventola, Progress in nanomedicine: approved and investigational nanodrugs, P
& T: Peer-Reviewed J. Formul. Manag. 42 (2017) 742.
Acknowledgement [11] S.Y. Yu, H.B. You, Perspectives on the past, present, and future of cancer nano-
medicine, Adv. Drug Deliv. Rev. 130 (2018) 3–11.
[12] M.J. Alonso, Nanomedicines for overcoming biological barriers, Biomed.
L-P.Wu. acknowledges financial support from National Science and Pharmacother. 58 (2004) 168–172.
Technology Major Projects for Stem Cell and Translation (reference [13] A. des Rieux, V. Fievez, M. Garinot, Y.-J. Schneider, V. Préat, Nanoparticles as
potential oral delivery systems of proteins and vaccines: a mechanistic approach, J.
2019YFA0110500), National Science and Technology Major Projects Control. Release 116 (2006) 1–27.
for New Drug Development (reference 2018ZX09733-006), Guangdong [14] Z. Xu, D. Wang, Y. Cheng, M. Yang, L.-P. Wu, Polyester based nanovehicles for
Pearl River Talents Program (reference 2017GC010411), International siRNA delivery, Mater. Sci. Eng. C 92 (2018) 1006–1015.
[15] D. Wang, L.P. Wu, Nanomaterials for delivery of nucleic acid to the central nervous
Science and Technology Cooperation Project of Guangzhou Economic
system (CNS), Mater. Sci. Eng. C 70 (2017) 1039–1046.
and Technological Development District (reference 2018GH15) and [16] L. Wu, S.M. Moghimi, A nanoengineered peptidic delivery system with specificity
Drug Discovery Pipeline of Guangzhou Institutes of Biomedicine and for human brain capillary endothelial cells, Nanomedicine: NBM (NMR Biomed.) 12
Health (reference 201508020131). (2016) 474–475.
[17] A. Kabanov, H.E. Gendelman, Nanomedicine in the diagnosis and therapy of neu-
rodegenerative disorders, Prog. Polym. Sci. 32 (2007) 1054–1082.
References [18] R.K. Jain, T. Stylianopoulos, Delivering nanomedicine to solid tumors, Nat. Rev.
Clin. Oncol. 7 (2010) 653–664.
[19] S. Taurin, H. Nehoff, K. Greish, Anticancer nanomedicine and tumor vascular per-
[1] V. Wagner, A. Dullaart, A.-K. Bock, A. Zweck, The emerging nanomedicine land- meability; where is the missing link? J. Control. Release 164 (2012) 265–275.
scape, Nat. Biotechnol. 24 (2006) 1211–1217. [20] S.H. Jang, M.G. Wientjes, D. Lu, J.L.-S. Au, Drug delivery and transport to solid
[2] S.M. Moghimi, A.C. Hunter, J.C. Murray, Nanomedicine: current status and future tumors, Pharm. Res. 20 (2003) 1337–1350.
prospects, FASEB J. 19 (2005) 311–330. [21] A.I. Minchinton, I.F. Tannock, Drug penetration in solid tumours, Nat. Rev. Cancer
[3] K.A. Howard, Nanomedicine: Working Towards Defining The Field, Nanomedicine, 6 (2006) 583–592.
Springer, 2016, pp. 1–12. [22] H. Maeda, T. Sawa, T. Konno, Mechanism of tumor-targeted delivery of macro-
[4] S.M. Moghimi, A.C. Hunter, J.C. Murray, Long-circulating and target-specific na- molecular drugs, including the EPR effect in solid tumor and clinical overview of
noparticles: theory to practice, Pharmacol. Rev. 53 (2001) 283–318. the prototype polymeric drug SMANCS, J. Control. Release 74 (2001) 47–61.
[5] R. Gaspar, R. Duncan, Polymeric carriers: preclinical safety and the regulatory [23] R.K. Jain, Transport of molecules across tumor vasculature, Cancer Metastasis Rev.
implications for design and development of polymer therapeutics, Adv. Drug Deliv. 6 (1987) 559–593.
Rev. 61 (2009) 1220–1231. [24] K. Cho, X. Wang, S. Nie, D.M. Shin, Therapeutic nanoparticles for drug delivery in
[6] W.R. Sanhai, J.H. Sakamoto, R. Canady, M. Ferrari, Seven challenges for nanome- cancer, Clin. Cancer Res. 14 (2008) 1310–1316.
dicine, Nat. Nanotechnol. 3 (2008) 242–244. [25] L.-P. Wu, M. Ficker, J.B. Christensen, P.N. Trohopoulos, S.M. Moghimi, Dendrimers
[7] Y.C. Barenholz, Doxil®—the first FDA-approved nano-drug: lessons learned, J. in medicine: therapeutic concepts and pharmaceutical challenges, Bioconjug.
Control. Release 160 (2012) 117–134. Chem. 26 (2015) 1198–1211.
[8] J. Wolfram, M. Ferrari, Clinical cancer nanomedicine, Nano Today 25 (2019)

6
L.-P. Wu, et al. Materials Science & Engineering C 106 (2020) 110302

[26] L.P. Wu, D. Wang, L. Parhamifar, A. Hall, G.Q. Chen, S.M. Moghimi, Poly (3-hy- Prog. Lipid Res. 42 (2003) 463–478.
droxybutyrate-co-R-3-hydroxyhexanoate) nanoparticles with polyethylenimine coat [55] Z. Liu, C. Davis, W. Cai, L. He, X. Chen, H. Dai, Circulation and long-term fate of
as simple, safe, and versatile vehicles for cell targeting: population characteristics, functionalized, biocompatible single-walled carbon nanotubes in mice probed by
cell uptake, and intracellular trafficking, Adv. Healthc. Mater. 3 (2014) 817–824. Raman spectroscopy, Proc. Natl. Acad. Sci. U.S.A. 105 (2008) 1410–1415.
[27] M. Harrison, D. Tomlinson, S. Stewart, Liposomal-entrapped doxorubicin: an active [56] O. Rabin, J.M. Perez, J. Grimm, G. Wojtkiewicz, R. Weissleder, An X-ray computed
agent in AIDS-related Kaposi's sarcoma, J. Clin. Oncol. 13 (1995) 914–920. tomography imaging agent based on long-circulating bismuth sulphide nano-
[28] A. Wicki, D. Witzigmann, V. Balasubramanian, J. Huwyler, Nanomedicine in cancer particles, Nat. Mater. 5 (2006) 118–122.
therapy: challenges, opportunities, and clinical applications, J. Control. Release 200 [57] S.-D. Li, L. Huang, Pharmacokinetics and biodistribution of nanoparticles, Mol.
(2015) 138–157. Pharm. 5 (2008) 496–504.
[29] P.L. Golden, J. Huwyler, W.M. Pardridge, Treatment of large solid tumors in mice [58] K. Brindle, New approaches for imaging tumour responses to treatment, Nat. Rev.
with daunomycin-loaded sterically stabilized liposomes, Drug Deliv. 5 (1998) Cancer 8 (2008) 94–107.
207–212. [59] E. Tasciotti, X. Liu, R. Bhavane, K. Plant, A.D. Leonard, B.K. Price, M.M.-C. Cheng,
[30] V.P. Torchilin, Passive and Active Drug Targeting: Drug Delivery to Tumors as an P. Decuzzi, J.M. Tour, F. Robertson, Mesoporous silicon particles as a multistage
Example, Drug Deliv., Springer2010, pp. 3-53. delivery system for imaging and therapeutic applications, Nat. Nanotechnol. 3
[31] T. Lammers, W. Hennink, G. Storm, Tumour-targeted nanomedicines: principles (2008) 151–157.
and practice, Br. J. Canc. 99 (2008) 392–397. [60] K. Langer, M.G. Anhorn, I. Steinhauser, S. Dreis, D. Celebi, N. Schrickel, S. Faust,
[32] L.-P. Wu, M. Ficker, S.L. Mejlsøe, A. Hall, V. Paolucci, J.B. Christensen, V. Vogel, Human serum albumin (HSA) nanoparticles: reproducibility of prepara-
P.N. Trohopoulos, S.M. Moghimi, Poly-(amidoamine) dendrimers with a precisely tion process and kinetics of enzymatic degradation, Int. J. Pharm. 347 (2008)
core positioned sulforhodamine B molecule for comparative biological tracing and 109–117.
profiling, J. Control. Release 246 (2017) 88–97. [61] W.C. Zamboni, V. Torchilin, A.K. Patri, J. Hrkach, S. Stern, R. Lee, A. Nel,
[33] T.M. Allen, Ligand-targeted therapeutics in anticancer therapy, Nat. Rev. Cancer 2 N.J. Panaro, P. Grodzinski, Best practices in cancer nanotechnology: perspective
(2002) 750–763. from NCI nanotechnology alliance, Am. Assoc. Cancer Res. 18 (2012) 3229–3241.
[34] R. Duncan, Polymer conjugates as anticancer nanomedicines, Nat. Rev. Cancer 6 [62] A. McBride, L.M. Holle, C. Westendorf, M. Sidebottom, N. Griffith, R.J. Muller,
(2006) 688–701. J.M. Hoffman, National survey on the effect of oncology drug shortages on cancer
[35] B. Gupta, T.S. Levchenko, V.P. Torchilin, Intracellular delivery of large molecules care, Am. J. Health Syst. Pharm. 70 (2013) 609–617.
and small particles by cell-penetrating proteins and peptides, Adv. Drug Deliv. Rev. [63] J.L. Berger, A. Smith, K.K. Zorn, P. Sukumvanich, A.B. Olawaiye, J. Kelley,
57 (2005) 637–651. T.C. Krivak, Outcomes analysis of an alternative formulation of PEGylated lipo-
[36] T. Skotland, T.G. Iversen, M.L. Torgersen, K. Sandvig, Cell-penetrating peptides: somal doxorubicin in recurrent epithelial ovarian carcinoma during the drug
possibilities and challenges for drug delivery in vitro and in vivo, Molecules 20 shortage era, OncoTargets Ther. 7 (2014).
(2015) 13313–13323. [64] L.E. Euliss, J.A. DuPont, S. Gratton, J. DeSimone, Imparting size, shape, and com-
[37] P. Hölig, M. Bach, T. Völkel, T. Nahde, S. Hoffmann, R. Müller, R.E. Kontermann, position control of materials for nanomedicine, Chem. Soc. Rev. 35 (2006)
Novel RGD lipopeptides for the targeting of liposomes to integrin-expressing en- 1095–1104.
dothelial and melanoma cells, Protein Eng. Des. Sel. 17 (2004) 433–441. [65] S.-S. Feng, L. Mu, K.Y. Win, G. Huang, Nanoparticles of biodegradable polymers for
[38] R.M. Schiffelers, G.A. Koning, T.L. ten Hagen, M.H. Fens, A.J. Schraa, A.P. Janssen, clinical administration of paclitaxel, Curr. Med. Chem. 11 (2004) 413–424.
R.J. Kok, G. Molema, G. Storm, Anti-tumor efficacy of tumor vasculature-targeted [66] N. Desai, Challenges in development of nanoparticle-based therapeutics, AAPS J. 14
liposomal doxorubicin, J. Control. Release 91 (2003) 115–122. (2012) 282–295.
[39] F. Danhier, B. Vroman, N. Lecouturier, N. Crokart, V. Pourcelle, H. Freichels, [67] D.B. Resnik, S.S. Tinkle, Ethics in Nanomedicine, (2007).
C. Jérôme, J. Marchand-Brynaert, O. Feron, V. Préat, Targeting of tumor en- [68] T.M. Allen, P.R. Cullis, Liposomal drug delivery systems: from concept to clinical
dothelium by RGD-grafted PLGA-nanoparticles loaded with paclitaxel, J. Control. applications, Adv. Drug Deliv. Rev. 65 (2013) 36–48.
Release 140 (2009) 166–173. [69] T. Vines, T.A. Faunce, Assessing the Safety and Cost-Effectiveness of Early
[40] X. Xu, W. Ho, X. Zhang, N. Bertrand, O. Farokhzad, Cancer nanomedicine: from Nanodrugs, (2009).
targeted delivery to combination therapy, Trends Mol. Med. 21 (2015) 223–232. [70] B.S. Zolnik, A. Gonzalez-Fernandez, N. Sadrieh, M.A. Dobrovolskaia, Minireview:
[41] T.L. Doane, C. Burda, The unique role of nanoparticles in nanomedicine: imaging, nanoparticles and the immune system, Endocrinology 151 (2010) 458–465.
drug delivery and therapy, Chem. Soc. Rev. 41 (2012) 2885–2911. [71] N. Lewinski, V. Colvin, R. Drezek, Cytotoxicity of nanoparticles, Small 4 (2008)
[42] H. Kettiger, A. Schipanski, P. Wick, J. Huwyler, Engineered nanomaterial uptake 26–49.
and tissue distribution: from cell to organism, Int. J. Nanomed. 8 (2013) [72] V. Stone, H. Johnston, R.P. Schins, Development of in vitro systems for nanotox-
3255–3269. icology: methodological considerations, Crit. Rev. Toxicol. 39 (2009) 613–626.
[43] B. Fubini, M. Ghiazza, I. Fenoglio, Physico-chemical features of engineered nano- [73] C.M. Sayes, K.L. Reed, D.B. Warheit, Assessing toxicity of fine and nanoparticles:
particles relevant to their toxicity, Nanotoxicology 4 (2010) 347–363. comparing in vitro measurements to in vivo pulmonary toxicity profiles, Toxicol.
[44] K.L. Aillon, Y. Xie, N. El-Gendy, C.J. Berkland, M.L. Forrest, Effects of nanomaterial Sci. 97 (2007) 163–180.
physicochemical properties on in vivo toxicity, Adv. Drug Deliv. Rev. 61 (2009) [74] A. Hall, A.K. Larsen, L. Parhamifar, K.D. Meyle, L.-P. Wu, S.M. Moghimi, High re-
457–466. solution respirometry analysis of polyethylenimine-mediated mitochondrial energy
[45] M.A. Dobrovolskaia, S.E. McNeil, Immunological properties of engineered nano- crisis and cellular stress: mitochondrial proton leak and inhibition of the electron
materials, Nat. Nanotechnol. 2 (2007) 469–478. transport system, Biochim. Biophys. Acta Bioenerg. 1827 (2013) 1213–1225.
[46] A.E. Nel, L. Mädler, D. Velegol, T. Xia, E.M. Hoek, P. Somasundaran, F. Klaessig, [75] A. Mayer, M. Vadon, B. Rinner, A. Novak, R. Wintersteiger, E. Fröhlich, The role of
V. Castranova, M. Thompson, Understanding biophysicochemical interactions at nanoparticle size in hemocompatibility, Toxicology 258 (2009) 139–147.
the nano–bio interface, Nat. Mater. 8 (2009) 543–557. [76] J. Szebeni, F. Muggia, A. Gabizon, Y. Barenholz, Activation of complement by
[47] G.F. Paciotti, L. Myer, D. Weinreich, D. Goia, N. Pavel, R.E. McLaughlin, therapeutic liposomes and other lipid excipient-based therapeutic products: pre-
L. Tamarkin, Colloidal gold: a novel nanoparticle vector for tumor directed drug diction and prevention, Adv. Drug Deliv. Rev. 63 (2011) 1020–1030.
delivery, Drug Deliv. 11 (2004) 169–183. [77] S.M. Moghimi, Z.S. Farhangrazi, Nanomedicine and the complement paradigm,
[48] J.K. Vasir, V. Labhasetwar, Biodegradable nanoparticles for cytosolic delivery of Nanomedicine: NBM (NMR Biomed.) 9 (2013) 458–460.
therapeutics, Adv. Drug Deliv. Rev. 59 (2007) 718–728. [78] S. Tinkle, S.E. McNeil, S. Mühlebach, R. Bawa, G. Borchard, Y.C. Barenholz,
[49] D.C. Drummond, O. Meyer, K. Hong, D.B. Kirpotin, D. Papahadjopoulos, Optimizing L. Tamarkin, N. Desai, Nanomedicines: addressing the scientific and regulatory gap,
liposomes for delivery of chemotherapeutic agents to solid tumors, Pharmacol. Rev. Ann. N. Y. Acad. Sci. 1313 (2014) 35–56.
51 (1999) 691–744. [79] U. Food, D. Administration, Nanotechnology Task Force Report, (2007).
[50] I. Lynch, T. Cedervall, M. Lundqvist, C. Cabaleiro-Lago, S. Linse, K.A. Dawson, The [80] F. Ehmann, K. Sakai-Kato, R. Duncan, D.H.P. De La Ossa, R. Pita, J.-M. Vidal,
nanoparticle–protein complex as a biological entity; a complex fluids and surface A. Kohli, L. Tothfalusi, A. Sanh, S. Tinton, Next-generation nanomedicines and
science challenge for the 21st century, Adv. Colloid Interface Sci. 134 (2007) nanosimilars: EU regulators' initiatives relating to the development and evaluation
167–174. of nanomedicines, Nanomedicine 8 (2013) 849–856.
[51] I. Lynch, K.A. Dawson, Protein-nanoparticle interactions, Nano Today 3 (2008) [81] U. Food, D. Administration, Draft Guidance, Considering whether an FDA-
40–47. Regulated Product Involves the Application of Nanotechnology, Guidance for
[52] M.P. Monopoli, D. Walczyk, A. Campbell, G. Elia, I. Lynch, F. Baldelli Bombelli, Industry, 2011.
K.A. Dawson, Physical− chemical aspects of protein corona: relevance to in vitro [82] U. Food, D. Administration, Considering whether an FDA-Regulated Product
and in vivo biological impacts of nanoparticles, J. Am. Chem. Soc. 133 (2011) Involves the Application of Nanotechnology, (2011).
2525–2534. [83] A. Hafner, J. Lovrić, G.P. Lakoš, I. Pepić, Nanotherapeutics in the EU: an overview
[53] P. Aggarwal, J.B. Hall, C.B. McLeland, M.A. Dobrovolskaia, S.E. McNeil, on current state and future directions, Int. J. Nanomed. 9 (2014) 1005–1023.
Nanoparticle interaction with plasma proteins as it relates to particle biodistribu- [84] P.V.D. Meeren, M. Kasinos, H. Saveyn, Relevance of two-dimensional Brownian
tion, biocompatibility and therapeutic efficacy, Adv. Drug Deliv. Rev. 61 (2009) motion dynamics in applying nanoparticle tracking analysis, Methods Mol. Biol.
428–437. 906 (2012) 525.
[54] S.M. Moghimi, J. Szebeni, Stealth liposomes and long circulating nanoparticles: [85] M. Wright, Nanoparticle tracking analysis for the multiparameter characterization
critical issues in pharmacokinetics, opsonization and protein-binding properties, and counting of nanoparticle suspensions, Methods Mol. Biol. 906 (2012) 511–524.

Вам также может понравиться