Sei sulla pagina 1di 17

Advanced Drug Delivery Reviews 65 (2013) 104120

Contents lists available at SciVerse ScienceDirect

Advanced Drug Delivery Reviews


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

Advanced materials and processing for drug delivery: The past and the future
Ying Zhang, Hon Fai Chan, Kam W. Leong
Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA

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

Article history: Design and synthesis of efcient drug delivery systems are of vital importance for medicine and healthcare. Ma-
Accepted 16 October 2012 terials innovation and nanotechnology have synergistically fueled the advancement of drug delivery. Innovation
Available online 23 October 2012 in material chemistry allows the generation of biodegradable, biocompatible, environment-responsive, and
targeted delivery systems. Nanotechnology enables control over size, shape and multi-functionality of particulate
Keywords:
drug delivery systems. In this review, we focus on the materials innovation and processing of drug delivery
Commercialized drug delivery system
Nanoparticle
systems and how these advances have shaped the past and may inuence the future of drug delivery.
Polyplex 2012 Elsevier B.V. All rights reserved.
Natural polymers
Polymerdrug conjugate
Combinatorial chemistry
Microuidics
Particle replication in non-wetting template
Step-ash imprint lithography

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105
2. Advances in materials design . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106
2.1. Early FDA approved drug carriers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106
2.1.1. Biodegradable drug carrier: PLGA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
2.1.2. Other FDA approved biodegradable DDS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
2.2. Block copolymers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
2.3. Polymer drug conjugates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
2.4. Novel use of nature polymers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
2.5. Recombinant protein-based drug carriers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
2.6. Smart DDS. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
2.7. Combinatorial chemistry and polymer library . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
3. Conventional NP fabrication . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
3.1. One-step NP formation: nanoprecipitation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
3.2. Two-step NP formation: emulsication-based methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
3.2.1. Emulsication-solvent evaporation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
3.2.2. Emulsication-solvent diffusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111
3.2.3. Emulsication-salting out . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111

Abbreviations: CFL, continuous ow photolithography; DDS, drug delivery system; dex-HEMA, dextranhydroxyethyl methacrylate; EE, encapsulation efciency; ELPs, elastomer-like
proteins; EPR, enhanced permeability effect; GFP, green uorescence protein; HFR, heparinfolic acidretinoic acid; HPMA, N-(2-hydroxypropyl) methacrylamide; IPA, isopropyl alcohol;
LBL, layer-by-layer; NPs, nanoparticles; O/W, oil-in-water; PAA, poly(acrylic acid); PDMS, polydimethylsiloxane; PEG, polyethylene glycol; PEG-DA, poly(ethylene glycol) diacrylate; PEG
PLA, poly(ethylene glycol)-b-polylactide; PEI, poly(ethyleneimine); PEO-PPO-PEO, poly(propylene oxide)- poly(ethylene oxide)- poly(propylene oxide); PFPE, peruoropolyether; PGA,
poly(glycol acid)/ poly (L-glutamic acid); PLA, poly(lactic acid)/polylactide; PLGA, poly (lactide-co-glycolide); PMMA, polymethyl methacrylate; pNIPAAM, poly(N-isopropylacrylamide);
PRINT, particle replication in non-wetting template; PS, polystyrene; PTMC, poly(trimethylene carbonate); PVA, poly(vinyl alcohol); RAFT, reversible additionfragmentation chain transfer;
RES, reticuloendothelial system; S-FIL, step-ash imprint lithography; SDS, sodium dodecyl sulphate; SFL, stop ow lithography technique; SLP, silk-like proteins; W/O, water-in-oil; W/O/W,
water-in-oil-in-water.
This review is part of the Advanced Drug Delivery Reviews theme issue on 25th Anniversary issue Advanced Drug Delivery: Perspectives and Prospects.
Corresponding author. Tel.: +1 919 660 8466; fax: +1 919 660 0031.
E-mail address: kam.leong@duke.edu (K.W. Leong).

0169-409X/$ see front matter 2012 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.addr.2012.10.003
Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120 105

3.3. Layer-by-layer synthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111


4. Microuidic platforms for DDS fabrication . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111
4.1. Microuidics as versatile platform for DDS synthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112
4.1.1. Nano DDS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112
4.1.2. Microparticles/microspheres . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112
4.1.3. Core-shell structures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112
4.1.4. Smart drug delivery core-shell structures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112
4.1.5. Janus particles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
4.1.6. Multi-shells, chamber particles and microbers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
4.2. Microuidics offers precise control over drug carrier synthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
4.2.1. Control of particle size . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
4.2.2. Control of particle shape. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
4.2.3. Control of particle shell thickness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
4.2.4. Control of drug loading efciency . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
4.2.5. Control of drug release rate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
4.3. Future perspective: scale-up production and versatility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
5. Topdown NP fabrication . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116
5.1. Particle replication in non-wetting template (PRINT). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116
5.2. Step-ash imprint lithography (S-FIL). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116
5.3. Microuidics-based topdown approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116
6. Challenges and future perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
Acknowledgments. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117

1. Introduction lower than that for a new drug (approximately $500 million and over
10 years) [2]. Not surprisingly the US market for advanced DDS has
Drug delivery is a eld of vital importance to medicine and healthcare. grown from $75 million in 2001 to $121 billion in 2010 [3]; the annual
Controlled drug delivery improves bioavailability by preventing prema- worldwide market for polymer-based controlled release system alone
ture degradation and enhancing uptake, maintains drug concentration is estimated to be $60 billion in 2010 [4].
within the therapeutic window by controlling the drug release rate, Exciting advances in genomics and systems biology have continued to
and reduces side effects by targeting to disease site and target cells. identify new molecular targets. Future therapeutics will be increasingly
Since the rst FDA approval of drug delivery system (DDS), Liposomal nucleic acid (plasmid DNA, siRNA, mRNA, and aptamer) and peptidic
amphotericin B, in 1990, more than 10 DDS are now commercially avail- (small peptide and protein) in nature. They have to act intracellularly, al-
able to treat diverse diseases ranging from cancer to fungal infection and though the polar nature of nucleic acids and proteins hinders their cellu-
to muscular degeneration (Fig. 1, Table 1) [1]. In improving therapeutic lar entry. They are also typically more fragile than small molecules. Drug
efcacy, DDS has beneted tens of millions of patients by relieving suffer- delivery will hence play an increasingly important role in realizing the full
ing and prolonging life. They have also changed the economics of drug potential of the next generation of therapeutics.
development. Packaging an existing drug into controlled release formula- Innovations in materials chemistry have initially fueled the develop-
tions may not only improve its performance but also extend its patent life ment of DDS, creating carriers that are biodegradable, biocompatible,
as a new product. The average cost and time required to develop a new targeting, and stimulus-responsive. Nanotechnology has joined forces in
DDS (approximately $2050 million and 34 years) is signicantly the past decade. The realization that size and shape of nanoparticles

Fig. 1. Timeline showing FDA approved DDS in the market.


106 Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120

Table 1
Examples of FDA approved DDS in the market.

Product name and category Year of approval Technology Indication

Liposome & micelle


Doxil 1995 PEGylated liposomal doxorubicin Various types of cancer
Daunoxome 1996 Liposomal daunorubicin Advanced HIV-associated Kaposi's sarcoma
Ambisome 1997 Liposomal amphotericin B Fungal infections
Depocyt 1999 Liposomal cytarabine Lymphomatous meningitis
Visudyne 2000 Liposomal verteporn Age-related macular degeneration
Estrasorb 2003 Estradiol micellar nanoparticles Moderate to severe vasomotor symptoms of menopause
DepoDur 2004 Liposomal morphine sulfate Postoperative pain

Polymerdrug conjugate
Adagen 1990 PEGylated adenosine deaminase Adenosine deaminase deciency causing
severe combined immunodeciency disease
Oncaspar 1994 PEGylated L-asparaginase Acute lymphoblastic leukemia
PEG-intron 2001 PEGylated interferon alfa-2b Chronic hepatitis C
PEG-ASYS 2002 PEGylated interferon alfa-2a Chronic hepatitis C & B
Neulasta 2002 PEGylated granulocyte colony-stimulating Neutropenia
factor analog
Somavert 2003 PEGylated recombinant analogue of the Acromegaly
human growth hormone
Macugen 2004 Pegylated anti-VEGF aptamer Age-related macular degeneration
Mircera 2007 PEGylated erythropoetin receptor activators Anaemia associated with chronic kidney disease
Cimzia 2008 PEGylated tumor necrosis factor alpha inhibitor Crohn's disease
Krystexxa 2010 PEGylated urate oxidase Gout
Omontys 2012 PEGylated peginesatide Anemia caused by chronic kidney disease

Biodegradable materials
Zoladex 1989 PLGA/goserelin acetate Prostate and breast cancer
Lupron Depot 1989 PLGA/leuprolide acetate Prostate cancer and endometriosis
Gliadel 1996 Polifeprosan 20/carmustine High-grade & recurrent glioblastoma multiforme
Sandostatin LAR 1998 PLGA-glucose/octreotide acetate Acromegaly
Atridox 1998 PLA/doxycycline hyclate Periodontal disease
Nutropin depot 1999 PLGA/recombinant human growth hormone Growth hormone deciency
Trelstar 2000 PLGA/triptorelin pamoatea Advanced prostate cancer
Arestin 2001 PLGA/minocycline Adult periodontitis
Eligard 2002 PLGA/leuprolide acetate Advanced prostate cancer
Risperdal Consta 2003 PLGA/risperidone Schizophrenia & bipolar I Disorder
Vivitrol 2006 PLGA/naltrexone Alcohol dependence & opioid dependence
Somatuline 2007 PLGA/lanreotide Acromegaly
Ozurdex 2009 PLGA/dexamethasone Macular edema

Protein-based DDS
Zevalin 2002 Anti-CD20 monoclonal antibody/yttrium-90 Non-Hodgkin's lymphoma
Bexxar 2003 Anti-CD20 monoclonal antibody/iodine-131. non-Hodgkin's lymphoma
Abraxane 2005 Albumin/paclitaxel Breast cancer
Brentuximab Vedotin 2011 Anti-CD30 monoclonal antibody/monomethyl auristatin E Hodgkin lymphoma & systemic anaplastic
large cell lymphoma

Source: U S Food and Drug Administration website (http://www.accessdata.fda.gov/scripts/cder/drugsatfda/). Websites of various pharmaceutical companies supplying the drugs.

(NPs) can help navigate biological carriers has stimulated the application design the development of DDS will enter a new era of improving
of nanofabrication technologies, both topdown and bottomup, to healthcare.
develop more effective particulate DDS. For instance, the size of NPs de-
termines their biodistribution. Whereas particles smaller than 20 nm 2. Advances in materials design
will be cleared from circulation via reticuloendothelial system (RES)
within a few hours when injected intravenously, larger ones will be 2.1. Early FDA approved drug carriers
trapped in the liver and the spleen within minutes [5,6]. In a study of
accumulation of long-circulating polymeric micelles at tumor site, Design and synthesis of various biocompatible materials has driven
Kataoka and colleagues show only those smaller than 30 nm could the progress of DDS in the past few decades. Liposome has been the
effectively penetrate poorly permeable pancreatic tumors [7]. Fabri- most successful candidate in clinical applications. Most of the FDA ap-
cating techniques such as nanoprecipitation, emulsion-based phase proved DDS are liposome or lipid-based (Fig. 1, Table 1) [1]. Moreover,
inversion, microuidics-based self-assembly, layer-by-layer synthe- polymerdrug conjugates, especially those conjugated with polyethylene
sis, and nanoimprinting have been used to generate particulate DDS glycol (PEG) to reduce protein adsorption and aggregation, is now a stan-
to deliver a wide range of drugs. To fully realize the potential of a par- dard method to increase circulation and bioavailability of biomacro-
ticulate DDS with controlled size and shape, nanofabrication and molecules such as antibodies. FDA approval of paclitaxel and albumin
nanomanufacturing will play a more prominent role in the future. conjugates in replacement of the traditional Cremophor represents
In this article, we will rst briey review DDS development from a the rst successful clinical example of using biomacromolecules to facili-
materials perspective. This will be followed by a more detailed discus- tate drug delivery (Table 1). In a randomized, open-labeled trial of 460 pa-
sion on recent innovative fabrication techniques for DDS. It is our belief tients, the new formulation performed better than Taxol alone and
that when innovations in fabrication can catch up with those in materials showed signicantly reduced side effect even at a 50% higher dose [8].
Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120 107

2.1.1. Biodegradable drug carrier: PLGA This system is under phase II clinical investigation to deliver paclitaxel
Biodegradable polymers are often considered as alternatives to lipids (NK105) and doxorubicin (NK911). The other one, which is a poly(pro-
for their improved in vivo stability. The degradability could be tuned to pylene oxide)- poly(ethylene oxide)- poly(propylene oxide) (PEO-PPO-
control the rate of drug release. Among all, poly (lactic acid) (PLA), PEO) (Pluronic) triblock copolymer developed by Kabanov et al. to deliv-
poly(glycol acid) (PGA) and their copolymers, poly (lactide-co-glycolide) er doxorubicin, is under phase III clinical evaluation in Canada. Other than
(PLGA) are the most widely used for DDS development because of their amphiphilic block copolymers, PEG-polycation block copolymers are also
biodegradability, biocompatibility and ease of processing [9,10]. PLGA of great interest due to their ability to condense nucleic acids into
NPs are used to deliver various bioactive agents, such as small drugs nanosized polyplex with protective and biocompatible PEG shell. Block
[1114], peptides and proteins [1517], and recently plasmid DNA copolymers of PEG-PEI, PEG-polylysine have been investigated by
[18,19]. The size of PLGA particles can be adjusted [20,21] by changing Kataoka et al. in the 1990s [37].
the chemical composition as well as the fabrication method. Drug release A major drawback of polymeric micelles is their relative instability in
rate from PLGA NPs could be controlled by varying the molecular weight blood upon systematic administration, which leads to rapid dissociation
of PLA [2224], which determines the degradation rate of the vesicle. and burst release. To tackle this problem, various innovative approaches
PLGA NPs can be easily modied to accommodate targeting motif, have been used to engineering the micelle core with different chemistry:
PEGylation and environment-responsive elements. Incorporation of D en- a) Increase the hydrophobicity of the core by attaching pendant groups
antiomer in PLA segments leads to the formation of stereocomplexes to the backbone, such as fatty acid, benzyl groups, cholesterol [3840];
with improved mechanical property and slower degradation [25,26]. In b) Introduce hydrogen-bond interaction in the core. For example,
one study, stererocomplexes are used to form nanobers with paclitaxel Hedrick and Yang et al. have created an amphiphilic PEO-PCL based poly-
and provide a controlled release of the drug for more than 30 days [27]. mer containing urea functional group in the side chain that shows im-
All these support the notion that PLGA is a versatile system for drug deliv- proved kinetic stability of micelles in the presence of a destabilizing
ery. It has been a popular platform for new DDS development in the past agent [41]; c) Promote electrostatic interactions by introducing oppo-
and will continue to be so in the future because many PLGA-based DDS sitely charged groups in the micelle core, such as PEO with either poly
are already on the market (Fig. 1, Table 1). (L-lysine) or poly(aspartic acid) [37]; d) Crosslink the micelle core by
thermal or photo-induced polymerization. For instance, Kataoka et al.
2.1.2. Other FDA approved biodegradable DDS synthesized PEO-PLA block copolymers having polymerizable metha-
A vast array of biodegradable polymers, ranging from synthetic to cryloyl pendant groups at the PLA block [42,43]. Upon micelle formation,
natural and to hybrid, have been studied and developed for drug deliv- the methacryloyal groups are crosslinked thermally or photochemically.
ery, many of which are highlighted in this theme issue and in recent The nal micelle structure is stable at high temperature and in organic
reviews. Polyanhydride is another class of biodegradable polymers solvent. By incorporating reversible crosslinkers such as di-sulde
whose application as DDS has enjoyed clinical success. To overcome bonds, polymeric micelles can be made stable in the blood stream but
the undesirable bulk-degrading behavior of polyesters, polyanhydrides dissemble in reductive environment inside the cell [44]. Furthermore,
are designed to undergo surface-degradation. This is made possible by with a chemical structure that affords better miscibility with the encap-
the hydrolytically labile anhydride linkage, which combines with a hy- sulated cargo, higher loading level can be achieved [45,46].
drophobic backbone, restricts biodegradation at the surface since the The shell of polymeric micelles could also be modied according to
rate of hydrolysis at the surface is much faster than the rate of water pen- the application. For example, the micellar shell can be crosslinked to
etration into the core of the sample. This in principle can lead to a steadi- alter the permeability of the micelle and control drug release rate. A
er release kinetics if the release mechanism is controlled by the carrier poly(acrylic acid) shell could be crosslinked with diamino compounds
degradation. First approved in 1996, Gliadel, a polyanhydride wafer to form amide bonds in a condensation reaction. The hydrophilic shell
placed in the resection cavity after brain tumor surgery for the delivery could also be functionalized with various ligands (antibodies, small or-
of carmustine to patients with recurrent malignant glioma showed mod- ganic molecules, carbohydrates, peptides or polymers) to promote specif-
est improvement in survival [28]. In 2003, Gliadel received additional ic binding to cancer cells. The creation of polymeric micelles with cationic
FDA approval to treat patients with newly diagnosed malignant glioma surface allows co-delivery of nucleic acids and small organic drugs. Yang
as adjunct to surgery and radiation therapy (Table 1). Recent phase III tri- et al. has synthesized a cationic amphiphilic block copolymer, which
als further investigated its use as rst-line treatment when followed by comprises poly(N-methyldietheneamine sebacate) (PMDS) as the cat-
concomitant radiochemotherapy with temozolomide [29]. Although ionic block for siRNA and plasmid binding, and a cholesterol pendant
the treatment is still associated with signicant toxicity due to its aggres- group to increase the hydrophobicity of the core for efcient drug loading
sive nature, the benet of controlled drug delivery is implicated in these [47]. The cationic core-shell micelle structures could efciently deliver
trials. As diverse properties of drugs used for various therapies demand both cargos in vivo and exhibit synergistic effects to suppress tumor
carriers of different properties to achieve optimal delivery, many biode- growth upon intra-tumor injection in a breast cancer model. A triblock
gradable polymers have been proposed to satisfy the need [3034]. Chal- copolymer, poly (ethylene glycol)--poly (-caprolactone)--poly(2-
lenges to gain regulatory approval have slowed the progress but the aminoethyl ethylene phosphate) (mPEG--PCL--PPEEA) is made by
tremendous benets of controlled drug delivery should see the emer- Wang et al. to co-deliver Plk1 siRNA and paclitaxel [48]. Synergistic
gence of other new biodegradable DDS eventually. tumor suppression is observed in a dose-dependent manner following
systematic administration. More such co-delivery systems have been in-
2.2. Block copolymers vestigated by various groups [4953]. Though in vivo stability and the risk
of exposing nucleic acids on micellelar surface to enzymatic degradations
Block copolymers have captivated the imagination of researchers needs to be further addressed, these systems provide promising alterna-
since the early days of drug delivery because of their remarkable chemi- tives to liposomes for the co-delivery of hydrophilic macromolecule and
cal exibility [35]. Depending on the choice of building blocks, they could small lipophilic drugs.
assemble to nanostructures in the form of micelles, electrostatic com-
plexes, or polymersomes. The versatility in forming colloidally stable 2.3. Polymer drug conjugates
nanoparticles is appealing for passive tumor targeting utilizing the EPR
effect. In late 1980s and early 1990s, two groups independently reported In addition to encapsulation and non-covalent complexation, conju-
the successful development of micellar DDS [36]. One is from Kataoka et gation to a polymeric carrier via a liable linker presents another attractive
al., which is an A-B block copolymer comprised of PEG and poly(aspartic approach to alter and optimize the pharmacokinetics of therapeutic
acid) modied by 4-phenyl-1-butanol to enhance the hydrophobicity. agents. Many early polymerdrug conjugates have exclusively focused
108 Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120

on the delivery of commercialized anti-cancer drugs, such as paclitaxel, and conne, are smaller in size, easier and cheaper to synthesize and pu-
doxorubicin and camptothecin [54]. Since the size of polymerdrug rify, and less immunogenic. Furthermore, nucleic acid itself as a biomate-
conjugates could be controlled by adjusting the molecular weight of the rial and building block may be an interesting platform for delivery
polymer, it could be optimized to maximize the benets of enhanced per- applications [77]. There are no nanoscale interactions more elegant
meability effect (EPR) at leaky tumor vasculature. Moreover, conjugating than those governing nucleic acids by the WatsonCrick pairing rules.
drugs to a polymeric carrier can enhance solubility of hydrophobic drugs, The innite combinations of DNA/RNA base pairs and their remarkable
extend drug circulation in vivo and enhance uptake by addition of molecular recognition capability can give rise to interesting nanostruc-
targeting motifs to the polymer. With 16 such systems (excluding PEG tures that are only limited by our imagination. Creative assembly of
conjugates) entering clinical trials [55], the last decade has witnessed nucleic acids has fashioned a plethora of 2D and 3D nanostructures
the rapid development and clinical validation of polymerdrug conju- with precisely controlled size, shape, and spatial functionalization. Vari-
gates. Most of these clinical studies have adopted N-(2-hydroxypropyl) ous DNA-based nanostructures suitable for drug encapsulation have
methacrylamide (HPMA) or poly (L-glutamic acid) (PGA) as the carrier. been invented, such as DNA nanotubes [78,79] and DNA nanoboxes
Originally developed by Kopecek and colleagues [56,57], the HPMA [80]. The nanobox which is about 35 nm in size has been designed to
doxorubicin conjugate comprises doxorubicin linked to a 30 kD backbone have a lid that can be locked or opened with a molecular key. This inter-
via an enzyme degradable peptide in the sidechain, with about 8 wt.% esting design allows for controlled release of small chemicals. Other DNA
drug content [57]. After structural validation and promising preclinical nano-architectures have been used to deliver various bioagents such as
results, HPMAdoxorubicin entered into a Phase I clinical trial in 1990 doxorubicin [81], CpG [82,83], and siRNA [81,83]. The susceptibility of
and that marked the rst milestone of development of polymerdrug these DNA nanostructures to premature nuclease degradation may
conjugates. In contrast to HPMA, PGA contains a biodegradable backbone limit their utility, but modied nucleic acids have proved much more
and allows for higher drug content (37% for PGA-paclitaxel conjugate) stable [84,85]. In another case, modication of protein has led to a
[58,59]. Conjugates of PGA with paclitaxel and camptothecin are also novel carrier design. Liu et al. from Harvard mutated the green uores-
under clinical evaluation [60,61]. Regulatory approval of these systems cence protein (GFP) to change from a wild-type with a net of seven neg-
is however slow. Many polymerdrug conjugates show unexpected re- atively charged groups to a polypeptide with 36 positively charged
lease behavior and side effects in clinical testing except PGApaclitaxel groups. The mutations do not alter the GFP structure and uorescence
conjugates (Xyotax). Early interpretation of phase III clinical trial of intensity, and yet offer great chemical resistance to enzyme degradation
Xyotax concluded with no therapeutic activity [62,63]. However, subse- and denaturation. These supercharged GFP are used to deliver various
quent analysis did show that Xyotax could improve survival rate in proteins [86,87] and DNA/siRNA [87] into different cells. Not only serv-
women although not in men [54,64]. An estrogen-dependent cathepsin ing a delivery function, the intrinsic uorescence from GFP also allows
B activity, the enzyme responsible for degrading PGA backbone, is the for tracking of the delivered cargo. This example highlights the potential
key player for these gender-dependent responses. The nding indicates of polypeptides as drug carriers, which is being increasingly pursued by
the importance of using suitable biomarkers, which may be different drug delivery researchers using protein engineering.
from those for the naked drug, in the development of DDS.
In parallel with clinical validation of the rst-generation polymer 2.5. Recombinant protein-based drug carriers
drug conjugates, several innovative strategies of creating polymer
drug conjugates have been proposed recently. Cheng et al. used a Recombinant proteins bear the intrinsic advantages of natural bio-
drug-initiated, controlled polymerization protocol to form polylactide polymers for medical application as they are biodegradable and poten-
(PLA)-drug conjugates. Drugs including paclitaxel [65], doxorubicin [66], tially biocompatible if the articial sequence is not antigenic. Genetic
cyclosporine A [67] and camptothecin [68] were conjugated to PLA via a engineering allows precise control over structural and functional proper-
hydrolysable ester linker. High drug content (1030 wt.%) was obtained ties of recombinant proteins, such as their molecular weight, hydropho-
and controlled release was observed. Reversible additionfragmentation bicity, targeting motif, secondary structures, and drug conjugation sites.
chain transfer (RAFT) polymerization, a technique that can achieve excel- Two broadly studied recombinant protein systems are the elastomer-
lent control over polymer molecular weight and composition, has also like proteins (ELPs), and silk-like proteins (SLP).
been applied to produce HPMA-SN-38 (a camptothecin analog) conjugate ELP is a family of recombinant proteins derived from the elastomeric
[69]. The polymer-conjugate displays good aqueous solubility and similar domain of extracellular matrix protein elastin. This short hydrophobic
anticancer effect of free drug. These one-step synthetic approaches sim- domain, comprised of ve amino acids (GVGVP), is the basic building
plify the manufacturing process as compared to conventional post- block of ELPs [88]. Moreover, by substituting the second amino acid in
synthesis conjugation methods. However, they also limit the linker choice the pentapeptide, ELPs can undergo reversible and rapid phase transi-
between polymer and drugs, which potentially diminishes the control tion in response to temperature. Chilkoti and colleagues have designed
over drug release rate. More innovative chemistries to expand the linker a series of ELP with distinct transition temperatures as drug carriers
library will further increase the appeal of polymerdrug conjugates. [8992]. In one system, ELP-peptide fusion protein was conjugated to
doxorubicin [91], which formed micelles and aggregated in the tumor
2.4. Novel use of nature polymers microenvironment under hyperthermic, leading to increased accumula-
tion at tumor site. In another study, the effect of hyperthermia-induced
Natural polymers derived from biological systems including protein, micelle formation was exploited to present multivalent targeting motifs
DNA, and polysaccharides are biocompatible and biodegradable poly- to enhance cellular uptake [92]. Chaikof and colleagues have also devel-
mers. They possess low toxicity and potentially favorable pharmacoki- oped multiblock ELP for drug delivery in the conguration of NP, hydro-
netics in the circulation. Use of natural polymers as drug carriers has a gel or lm depending on the multiblock composition and processing
long history. The use of polysaccharides [70] such as heparin, chondroitin method [9396].
sulfate, and chitosan [70,71] as carriers, and coupled with the use of an- Silk protein is a native block copolymer with alternating large hy-
tibody [72,73] and transferrin [73,74] as targeting motif has all brought drophobic and hydrophilic blocks. The hydrophobic block is generally
signicant clinical benets. Nonetheless, the tremendous potential of a repetitive sequence conserved with short-chain amino acids, such as
natural polymers as drug carriers is still under-represented and deserves glycine and alanine. The hydrophilic block is less conserved and usually
more attention. An interesting direction has been the use of nucleic acid contains non-repetitive sequences rich in charged amino acids. The hy-
in drug delivery. For instance, there is a growing interest in the use of drophilic domain is often substituted with other peptide sequences to
DNA or RNA aptamer as new targeting motif in replacement of antibody achieve specic function for drug delivery. The length of the hydropho-
[75,76]. Aptamers, the conjugation chemistry of which is easier to control bic domain could also be tuned to yield protein NPs with reproducible
Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120 109

Fig. 2. Conventional nanoparticle fabrication methods. a) Nanoprecipitation. Polymer dissolved in organic solvent is added to an aqueous solution in a dropwise manner under con-
stant agitation. Nanoparticles containing drugs form instantaneously as the polymer diffuses to the aqueous phase. b) Layer-by-layer assembly. Solid form of drugs are used as the
core. A polymer layer is rst adsorbed onto the drug colloidal template by incubating in polymer solution and transferred to the oppositely charged polymer solution for additional
layering. This process is repeated until nanoparticles of desired sizes are formed. c) Emulsion-based two step methods. Emulsied oil-in-water droplets containing polymer and
drugs are formed in the rst step. In the second step, different methods are applied to remove the solvent and precipitate nanoparticles. Top panel: solvent evapoaration method.
Solvents are gradually evaporated under vacuum and high pressure. Middle panel: Solvent diffusion method. The solvent used to prepare emulsion drops is partially miscible with
water. When the emulsion droplets are diluted with water containing stabilizer, organic solvent rapidly diffuses out from the droplets, leading to condensation of the materials
within and formation of polymer nanoparticles. Bottom panel: salting out. The solvent used to prepare polymer and drug solution is totally miscible with water. Emulsication
is conducted with aqueous phase containing high concentration of salt. The saturated aqueous phase prevents solvent from mixing with water. The emulsied droplets are then
diluted in water. A sudden drop of salt concentration in continuous phase causes extraction of organic solvent and precipitation of polymer drug nanoparticles.

sizes for drug and gene delivery [97,98]. A recent study demonstrated and biological (enzymes, proteins) signals have been used as the trigger-
that a SLP recombinant protein endowed with a cell penetrating peptide ing stimuli [100]. Numerous monomers have been discovered to possess
could achieve transfection efciency 45 times higher than that of sensitivity to a specic stimulus. They can be assembled to a homo-
poly(ethyleneimine) (PEI) [99]. polymer with tunable sensitivity to one signal or to a co-polymer
responding to multiple stimuli. Smart polymer coupled with targeting
ligand can probably best minimize off-target effects and maximize pro-
2.6. Smart DDS grammability. Smart DDS has been covered in excellent reviews else-
where [101104].
Much of innovations in materials design for drug delivery manifest in
producing smart DDS that are able to release the therapeutic payload
on-demand. Stimuli-responsive polymers mimic the behavior of biolog- 2.7. Combinatorial chemistry and polymer library
ical molecules where external stimuli or changes in local environment
can trigger a change in property: conformation, solubility, shape, charge, Structurefunction relationship for material design in drug delivery
and size. Drug release can be regulated not only in a spatial manner via has generally been derived from empirical deduction of available sys-
targeting, but also in a temporal manner when the external stimuli are tems, and only with limited structural variations coming out from a sin-
applied. Various chemical (pH, ionic strength), physical (temperature, gle laboratory. A systematic approach is required. Systematic study is
light, electricity, magnetic eld, ultrasound, mechanical stress etc.), difcult for polymeric systems, not only because the synthesis is tedious,
110 Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120

but also due to the intrinsic polydispersity of most polymers. Rapid 3. Conventional NP fabrication
progress in combinatorial chemistry enables the ne-tuning of drug car-
rier characteristics at the molecular level. In early days, combinatorial 3.1. One-step NP formation: nanoprecipitation
chemistry is used to synthesize and screen biocompatible polymers for
coating of medical implants and devices. One approach was described Nanoprecipitation (Fig. 2a) is one of the most commonly used NP fab-
by Kohn and coworkers, who built a library of 112 polyacrylates from rication methods, which accounts for more than 50% of the NPs reported
14 tyrosine-derived diphenols and eight diacids with slight differences for drug delivery [106]. It is performed by adding an organic solution
in polymer bulkiness, exibility, hydrophobicity, and cellular response which contains the polymer and lipophilic drugs into an aqueous solution
that affected their potential utility in medical implant applications [105]. in a drop-wise manner under constant stirring. NPs containing drugs form
Recently, combinatorial chemistry was attempted to generate a library instantaneously as the polymer diffuses into the aqueous phase. The mis-
of potential gene carriers. Akinc et al. used a simple synthetic scheme cibility of the solvent with water is the most critical parameter governing
to produce a library of over 1200 lipid-like polymers by combinatorial the outcome [109]. The rate of polymer addition and stirring speed also
chemistry to delivery siRNA in vivo [106]. In this study, a collection of inuence the size and drug loading level. Particle size formed by this
alkyl acrylates or acrylamides was reacted with different diamines via method is usually around 200 nm, typically smaller than those produced
Michael's addition to produce thousands of siRNA delivery vectors by other processes. The method can be applied to a wide range of poly-
with diverse physiochemical properties. A follow-up study used im- mers [110], peptides [111] and amphiphilic cyclodextrins [112], although
proved chemistry to synthesize another library of lipid-like polymers scale up would be inefcient due to the nature of drop-wise addition.
to reduce dosage of siRNA required for in vivo study [107]. Though
there is a conict in the optimal alkyl length for siRNA delivery between 3.2. Two-step NP formation: emulsication-based methods
these two studies, they share some common conclusions such as reten-
tion of a secondary amine and possession of either two long amide tails Many fabrication methods for NP preparation adopt a two-step
or several smaller amide tails for high siRNA transfection efciency in process. In the rst step, the organic phase containing the polymers
Hela cells. In a separate study, Weissleder et al. generated a small library and drugs are vigorously agitated or sonicated in the aqueous phase
of 146 NPs each multivalently presenting unique small molecules on sur- to form emulsied droplets. Depending on the emulsied system
face [108]. These magnetouorescent NPs were coated with dextran and (from nanoemulsion to macroemulsions) used, the eventual particle
functionalized with primary amine for conjugating small molecules for size and drug loading varies. Double emulsions could also be used
targeting. With this library, the group was able to identify small molecules to prepare core-shell vesicular structures. Following emulsication,
that produced differential afnity for different cell lines in vitro and in various methods are used to precipitate the polymer to form dense
vivo, without knowing a priori the molecular receptors involved. drug-loaded particles (Fig. 2b).
Chemical composition greatly inuences physicochemical proper-
ties of NPs, which in turn governs the pharmacokinetics and bio- 3.2.1. Emulsication-solvent evaporation
distribution of particulate DDS. Nonetheless, fabrication or formulation Emulsication-solvent evaporation is the most common method
also plays an equally essential part for optimal delivery. As discussed reported to prepare NPs [109]. In this method, polymer is dissolved in
earlier, structurefunction relationship is essential for design, charac- a volatile solvent (such as dichloromethane, chloroform) and emulsi-
terization and validation of DDS development. However, if the formula- ed in an aqueous phase. Formation of NPs is achieved by the evapora-
tion is awed in producing inferior products, the structurefunction tion of the solvent under reduced pressure [113]. Nevertheless, this is a
relationship would be inaccurate and even misleading. For example, slow process compared to nanoprecipitation which happens in millisec-
the optimal N/P ratio of polyplexes for nonviral gene delivery deter- onds. For instance, it requires 80 min to evaporate 10 mL of ethyl ace-
mined by bulk mixing might be inaccurate if such formulation produces tate from a 50 mL aqueous emulsion. During the rst 40 min 90% of
highly heterogeneous and unstable polyplexes within the same mix- the solvent is removed but it takes another 40 min to completely get
ture. With the same materials chemistry, fabrication may also enable rid of the rest [114]. The size of NPs drops to minimal during the rst
generation of NPs with different characteristics such as sizes, shapes, 40 min, and increases in the second 40 min due to coalescence of emul-
and drug loading efciencies and release proles, which impacts the sion droplets. Thus, the coalescence determines the nal particle size
therapeutic outcome. Moreover, fabrication methods directly inuence which is largely dependent on the evaporation condition. This high-
the cost, ease of purication, reproducibility and scalability of a drug, lights the importance of optimizing processing conditions in determin-
which are also important considerations in pharmaceutical develop- ing quality of particulate DDS. Adjusting solvent evaporation condition
ment. As it is envisioned that NPs will become the dominant DDS for in- such as temperature and pressure would improve the quality. The use
tracellular delivery, and nanomedicine has captured the imagination of of a surfactant such as sodium dodecyl sulphate (SDS) or poly(vinyl
scientists and layman alike, herein we will discuss the fabrication of alcohol) (PVA) would also minimize the coalescence effect and produce
NPs, the development of which is fueled by nanotechnology innova- smaller NPs [115].
tions. Since conventional fabrication methods for particulate DDS are Emulsion-solvent evaporation is widely used to encapsulate lipophilic
less reviewed elsewhere, we will also spend some time describing drugs. However, the loading level for hydrophilic drugs, such as proteins
them before introducing the recent innovations in fabrication such as and peptides is generally poor due to diffusion of the hydrophilic drug
microuidics and imprinting technologies. into the aqueous phase before the polymer can solidify to entrap the

Fig. 3. Common types of microuidics design. (a) Flow focusing, (b) T-junction and (c) concentric capillaries.
Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120 111

Fig. 4. Schematic illustration of the microuidics preparation process of various drug carriers. (a) Nanocomplex. During hydrodynamic ow focusing, precursors self-assemble into
nanoparticles when precursor-solvent solution is mixed with buffer, in which the precursor is poorly soluble. The process occurs in three stages involving nucleation of
nanoparticles, growth through aggregation and stabilization. (b) Microparticle/microsphere. Disperse phase is broken up by the continuous phase to form droplets which in
turn give rise to microparticles upon solidication. (c) Core-shell structure. Sequential encapsulation generates double emulsion after which the shell is solidied to produce a
shell layer with a liquid core. (d) Janus particle. The disperse phase comprises two distinct inputs is broken up by the continuous phase to produce particle with two distinct phases.

drug [116]. To overcome this problem, water-in-oil-in-water (W/O/W) and precipitation of NPs. This method works exclusively for lipophilic
double emulsion can be used to reduce the loss and also to preserve the drugs. The choice of salting-out agent greatly impacts particle size and
bioactivity of delicate drugs such as proteins in the aqueous phase. Typi- drug encapsulation efciency, whereas mechanical mixing, stabilizer
cally, the primary emulsion is formed by ultrasound treatment of a concentration have less profound effect. Zhang et al. compared the
mixture of aqueous phase containing therapeutics and organic phase solvent-diffusion and salting-out methods to prepare poly(trimethylene
containing the polymer and an organic surfactant serving as the stabilizer carbonate) (PTMC) NPs containing a lipophilic drug. Particle size from
for water-in-oil (W/O) emulsion. The second emulsion is formed by son- salting-out is smaller but drug loading efciency is also lower [121].
icating a mixture of the organic phase containing dispersed W/O emul-
sions and aqueous phase containing a hydrophilic stabilizer. Sonication
3.3. Layer-by-layer synthesis
duration for the second step is more critical in determining the nal par-
ticle size as compared to that of the rst step and surfactant concentration
Layer-by-layer (LBL) NP preparation (Fig. 2c) makes use of the
[117]. Longer sonication time signicantly reduces the size of the particles
electrostatic interaction between oppositely charged polyelectrolytes,
and produces smaller polydispersity, but this has to be balanced against
such as polylysine, chitosan, gelatin B, or poly(ethyleneimine) (PEI)
the potential risk of damaging the drug.
complexing with sodium alginate, poly(acrylic acid) (PAA), dextran
sulfate, hyaluronic acid, chondroitin sulfate or heparin. Solid form of
3.2.2. Emulsication-solvent diffusion bioactive agents is often used as the core to grow the vesicular structure.
Emulsication-solvent diffusion is another widely used method to A polymer layer is rst adsorbed onto the colloidal template by incuba-
prepare drug-loaded NPs. In this method, the polymer is dissolved in a tion in the polymer solution, washed, and transferred to the oppositely
partially water-miscible solvent (such as benzyl alcohol and propylene charged polymer solution. Repeat of the cycle leads to a multi-layered
carbonate) which is pre-saturated with water [118]. A typical emulsi- coating that can control the release kinetics. Calcium phosphate nano-
cation method is then used to produce oil-in-water (O/W) emulsion crystal, which could be dissolved away by chloric acid, is frequently
droplets from the water-polymer saturated solvent. The dispersed used as the core. This method can be used to encapsulate various bioac-
droplets are then diluted by a large amount of water containing a stabi- tive agents, such as paclitaxel [122], tamoxifen [123], vitamins [124],
lizer. The diffusion of organic solvent out from the droplets leads to the insulin [124], antigenic peptides [125], and nucleic acids [126]. In the
condensation of the materials within the droplet and formation of NPs. latter case, oral delivery of siRNA against TNF- and suppression of
The solvent extraction process takes places within a few milliseconds, inammation was achieved by LBL assembly of PEI and siRNA onto a
causing a drop in particle size. In general, the diameter of particles pre- 1,3-d-glucan core extracted from yeast. In this method, particle size
pared by this method is around 150 nm. Due to the fast solvent extraction and drug release rate could be controlled by controlling the thickness
kinetics and well-dened solventwater interaction, physical properties of the shell.
of NPs prepared with this method is highly reproducible and the polydis-
persity is signicantly lower than NPs prepared by other conventional
4. Microuidic platforms for DDS fabrication
methods [119].
Conventional NP fabrication techniques are prone to polydispersity
3.2.3. Emulsication-salting out and batch-to-batch variations. For instance, the nal size of NPs generat-
Emulsication-salting out is a derivative of the emulsication-solvent ed by the emulsion-based techniques is directly determined by the size
diffusion method. The organic solvent used is totally miscible with water, of the emulsion droplets, which itself could be very heterogeneous in
such as acetone. The polymer-containing solvent is emulsied in an aque- bulk mixing. While heterogeneity remains an insurmountable obstacle
ous phase containing high concentration of salt (magnesium chloride, in bulk preparation of DDS, microuidics, the manipulation of uid in
calcium chloride) or sucrose [120]. The saturated aqueous solution pre- nano/picoliter scale channels, presents exciting opportunities to im-
vents acetone from mixing with water. Dilution of the emulsion droplets prove the fabrication and manufacturing of particulate DDS. The general
in a large amount of water results in an abrupt drop of salt concentra- benets of conducting reaction in microuidics include but not limited
tion of the continuous phase, leading to the extraction of organic solvent to rapid mixing of reagents, homogeneous reaction environment,
112 Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120

exibility for multi-step reaction design, enhanced processing accuracy poly(N-isopropylacrylamide) pNIPAAM [133,134,152,155], hydro-
and efciency, better heat transfer due to high surface-to-volume ratio, gelator [153], silk protein [135], pectin [175], hydrazide and
miniaturization, and cost savings from reduced consumption of reagents aldehyde-functionalized carbohydrates [158], dextranhydroxyethyl
[127]. In particular, the rapid mixing feature, exibility for multi-step re- methacrylate (dex-HEMA) [166] and silica [154]) materials have been
action and efcient processing, as we shall see later, contribute to the investigated for microparticle formation through generation of liquid
improved fabrication process of particulate DDS in three aspects. First, precursor droplets in microuidics prior to solidication by solvent ex-
microuidics is a versatile platform that can be used to synthesize vari- traction or induced polymerization (Fig. 4b). Furthermore, porous micro-
ous types of drug carriers. Second, microuidics-aided synthesis exhibits particles are also fabricated with the help of microuidics to facilitate
better controllability over the physical properties of drug carriers. Third, drug release. The use of poly(ethylene glycol)-b-polylactide (PEGPLA),
microuidic possesses are amenable to scale-up and on-line quality con- which is oil-soluble, as gel matrix in O/W emulsion leads to the formation
trol, factors important for translation and commercialization. of a reverse micelle structure with a small amount of water encapsulated
Most of the microuidic devices nowadays are fabricated in either within, creating pores upon freeze drying [151]. PEG has also been used
glass [128143] or polydimethylsiloxane (PDMS) [144167]. Silicon as porogens during the formation of porous pNIPAAM microparticles
[168170], peruoropolyether [171], polymethyl methacrylate (PMMA) [155].
[172,173], polyurethane [174], polycarbonate [175] and stainless steel
[176] microuidic devices are also reported. In the commonest approach,
4.1.3. Core-shell structures
two streams containing continuous and disperse phases are infused into
W/O/W is useful to prepare core-shell structures. However, conven-
two separate inlets, and the disperse phase is conned into isolated drop-
tional emulsication techniques used to prepare core-shell structure gen-
lets or narrow stream at T-junction, in ow-focusing and concentric cap-
erate droplets with great variation in sizes. The possibility of adopting
illaries. The three most common types of microuidics design is shown in
microuidics for production of core-shell structure has been explored.
Fig. 3. The droplet conguration would produce spherical particulate DDS
Utada et al. rst reported the formation of W/O/W double emulsion drop-
in a discrete manner whereas the focused-stream conguration would
lets from a concentric microcapillary device [131]. Such W/O/W droplets/
promote self-assembly at the interface of the two adjacent streams in a
capsules could be used to trap and release insulin [178] and antibiotics
continuous manner.
[130] by either diffusion of substances through the oily membrane or dis-
ruption of the droplet structure under shear stress [179]. Using this meth-
4.1. Microuidics as versatile platform for DDS synthesis
od, DDS such as microscale polymersomes could be fabricated with
precise control over their size [128]. In other studies, microcapsules
4.1.1. Nano DDS
with alginate, PLA or a polyelectrolyte shell comprising anionic platinum
Due to the intrinsic limitation of bulk synthesis, the adoption of
NPs and cationic diazoresin, are prepared by forming single emulsion
microuidics for synthesis of nano-sized DDS has become an attractive al-
followed by ionic crosslinking of the surface of the capsule, solvent evap-
ternative [169]. Self-assembly of liposome in microuidics system is
oration or coating with polyelectrolytes [129,140,180].
demonstrated [168170]. Lipids dissolved in isopropyl alcohol (IPA) in
the disperse phase is channeled through two streams of buffer solutions
of continuous phase in a ow-focusing device where dilution of IPA 4.1.4. Smart drug delivery core-shell structures
occurs through mixing of uids at the interface. Liposome particles Microuidics has also been used to produce smart capsules for drug
(50200 nm in diameter) highly homogenous in size are precipitated at delivery. Using pNIPAAM along with crosslinker (BIS) as the disperse
downstream of the channel (Fig. 4a). Using the same approach, monodis- phase and hexadecane containing photoinitiator as the continuous
perse PLGA-PEG NPs ranging from 20 to 100 nm have also been synthe- phase, thermosensitive pNIPAAM capsule could be generated in a single
sized [146,147]. Drugs such as docetaxel and platinum (IV) prodrug have emulsion system by applying UV light to photopolymerize pNIPAAM
been encapsulated as an example. By selecting two immiscible phases precursor droplets from outside after the photoinitiators have diffused
containing different precursor molecules, lipidpolymer hybrid DDS are into contact with the monomer [167]. Change of capsule volume is
prepared in ow focusing device by dissolving hydrophilic PEG or induced by subjecting the capsule to different temperatures, a mecha-
PNIPAM in aqueous continuous phase and hydrophobic lipids in the nism that could potentially be used to trigger the release of drugs.
organic phase. The formation of PLGA/lecithin/PEG NPs (35180 nm) Using the double emulsion approach, thermosensitive microcapsules
and lipid-PNIPAM (150300 nm) are reported [148,177]. Apart from pre- could be made by coating polyacrylamide microgel as the inner core
cipitation reaction, recently the synthesis of polymerdrug conjugates with (pNIPAAM) precursor followed by crosslinking to form microgel
using microuidics system has also been demonstrated. Heparinfolic capsule [181]. A stimulus-responsive colloidosome which exhibits 80%
acidretinoic acid (HFR) bioconjugates (100 nm) with high drug cou- decrease in volume when actuated is fabricated by assembling pNI
pling ratio and uniform size distribution could be produced efciently PAAM microgels at the interface of W/O emulsion [141]. Moreover, a
after thorough mixing of two reactants introduced into a microchannel pH-responsive capsule can be generated from an O/W/O emulsion tem-
[171]. Reduced reaction time and high drug conjugation efciency is plate comprising chitosan in the middle water phase and oil-soluble
attained in a single-step assembly reaction. crosslinker in the inner oil phase [142]. Chitosan is crosslinked at the
While microuidics facilitates the preparation of nano-sized DDS by inner O/W interface as the inner oil contacts with the middle chitosan
rapidly mixing the precursor solution with buffer, the fabrication of DNA layer during transition in the microuidics device. The chitosan capsules
nanocomplexes such as lipoplex or polyplex for nonviral gene delivery displays an acid-sensitive burst release of cargo from the inner core by
of DNA, siRNA could also benet from the same platform [132,144, decomposition of the shell layer. Monodisperse polymersomes are fabri-
145]. Polyplexes (250 nm) and lipoplexes (200 nm) formed in picoliter cated using the similar approach in W/O/W emulsion when PEG-b-PLA
droplets by using a ow-focusing microuidics device show smaller adsorbs at the oilwater interfaces. The encapsulation and triggered
size, narrower size distribution, lower cytotoxicity, and higher transfec- release upon osmotic shock of 4000 Da dextran molecules is described.
tion efciency compared to those formed by bulk mixing, demonstrating Finally, a smart magnetic eld-sensitive drug delivery device is con-
the appeal of self-assembly in a small volume [144,145]. structed with high molecular weight chitosan embedded with magne-
tite particles as the capsule layer [149]. The release of drug (aspirin in
4.1.2. Microparticles/microspheres this case) from the inner core is sparked by an AC magnetic eld that
The capability of microuidics to generate discrete droplets is lev- causes the compression and extension oscillation of the capsule, resem-
eraged to fabricate microparticulate DDS. A range of organic (PLA bling the pumping action of drug. The drug release behavior could be
[151,161] and PLGA [156,157]) and inorganic (chitosan [172,173], tuned by simply varying the applied magnetic eld.
Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120 113

Fig. 5. Potential platform for customized drug carrier synthesis and sorting. Mass production of microdroplets is achieved by incorporation of multiple droplet generators, with
valves installed at the inlets of continuous and disperse phases to precisely control the size of particles. A continuous sorting component is included to sort particles according
to their properties.

4.1.5. Janus particles with the drug to be encapsulated at different layers for combination
In addition to conventional drug-encapsulated microparticles, Janus drug therapy or theranostics. As an example, a multilayer gas liposphere
particles, which have an interesting anisotropic structure and are dif- is fabricated in a microuidics system [160]. The liposphere contains an
cult to prepare by conventional methods, could be readily fabricated oil layer incorporated with hydrophobic drugs wrapping around a gas
in microuidics. Janus particles are fabricated by co-injecting two im- core intended as a contrast agent for ultrasound imaging. The lipid-PEG
miscible streams in the disperse phase and solidifying droplets generat- layer in exterior can be functionalized for targeted delivery. Multi-shell
ed at T-junction (Fig. 4d) [174]. These particles are attractive since they particles could prove useful in cancer therapy where combination drug
can be used to co-deliver two drugs with distinct properties or a drug therapy is more effective to eradicate tumor cells and as theranostic
and an imaging marker. As a demonstration, water soluble (doxorubicin agents where the site of drug delivery can be pinpointed by imaging
hydrochloride) and water insoluble (paclitaxel) drugs are encapsulated [147]. A multi-chamber particle can serve similar functions by encapsu-
in a single Janus particle. Each drug is separately released from the par- lating more than one inner sphere within a particle. A thermosensitive
ticles [176]. hydrogel encapsulating multiple inner oil droplets is fabricated by
microuidics, and the release of both hydrophobic (carried by the oil
4.1.6. Multi-shells, chamber particles and microbers phase) and hydrophilic molecules (associated with the aqueous hydro-
Multi-shell and multi-chamber particles, which have been shown to gel phase) is demonstrated [161]. Furthermore, microuidics system
be efciently produced on microuidics platform, represent novel drug allows precise control of the composition of inner spheres [137].
delivery vehicles whose potential has yet to be fully explored [138]. A Another potential DDS that can be fabricated on a microuidics plat-
multi-shell particle allows multiple drugs or a diagnostic agent along form in a simple and cost-effective manner is drug-loaded microbers.

Fig. 6. Particle Replication In Non-wetting Template (PRINT). A non-wetting PFPE mold with cavities of predesigned patterns is pressed against a polymer solution deposited on
another non-wetting surface. The liquid polymer solution is then solidied by applying pressure or temperature. The solidied particles could be recovered from the mold by
using an adhesive lm.
114 Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120

Fig. 7. Step-Flash Imprint Lithography (S-FIL). A quartz mold with cavities of predesigned shapes is pressed against a photo-crosslinkable monomer solution on top of a silica wafer.
A PVA layer is put beneath the polymer solution for the release of imprinted particles. The monomers are cross linked by applying UV light. Residual layer from crosslinking reaction
is removed by oxygen plasma etching and particles are freed by dissolving away the PVA layer.

Such a brous structure can be useful in surgical reconstruction where precipitation or polymerization, respectively. Among them, alginate
diffusion of drug can improve wound healing and minimize scarring bres encapsulating silver NPs are successfully fabricated for application
[182]. The production of alginate [162,163], PLA [182], an amphiphilic co- as wound dressing to stimulate healing while inhibiting microorganism
polymer [159] and poly(ethylene glycol) diacrylate (PEG-DA) [164] bers growth [163]. The tensile strength of bers, drug loading efciency
have been reported by owing a precursor solution into microchannels and degradation properties are parameters that need to be optimized
under laminar ow conditions to induce ber formation by crosslinking, to present its appeal as a novel DDS.

Fig. 8. Continuous ow photolithography. A stream of photo-crosslinkable monomer solution continuously ows through the rectangular channel of microuidic device. A photo-
mask with dened patterns is placed underneath through which pulses of UV light are applied. Particles of dened shapes are formed via cross-linking reaction and are ushed out
for collection.
Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120 115

4.2. Microuidics offers precise control over drug carrier synthesis which would be undesirable if uncontrolled. In the double emulsion ap-
proach, the thickness of the capsule shell can be easily controlled via op-
In addition to its versatile nature, microuidics platform offers anoth- timizing the design of microuidics device or simply altering the ow rate
er advantage over existing processing methods by exhibiting high degree adopted in the encapsulation process. PLA microcapsules with a shell
of control of DDS preparation in ve aspects: particle size, shape, shell thickness as thin as 80 nm could be reproducibly and effectively pro-
thickness of capsule, drug loading efciency and release rate. The follow- duced on microuidics platform, which is otherwise not achievable in
ing sections illustrate how the precise control is achieved in each aspect. bulk preparation process [139]. The precise control of the capsule shell
thickness afforded by microuidics enables tuning of the drug release
4.2.1. Control of particle size prole of microcapsules for different controlled drug release applications.
The most important benet of using microuidics to fabricate partic-
ulate DDS is the ability to precisely control particle size. The NPs or micro- 4.2.4. Control of drug loading efciency
particles generated from microuidics platform typically have a narrower Compared with conventional fabrication methods for microspheres
size distribution than those acquired by conventional methods [146,157]. which resulted in a drug encapsulation efciency (EE) of 5090% [150],
By two ways can microuidics platform facilitate the fabrication of mono- several studies that looked into drug loading efciency associated with
disperse particles. a) Using a T-junction or ow-focusing device, the a microuidics approach consistently reported an EE of 95% or greater
breakup of the disperse phase by the continuous phase is periodic and [150,173]. This is a direct result of generating isolated emulsion droplets
predictable, leading to discrete and consistently sized droplets formed during microparticle or microcapsule synthesis where no drug is lost
at the junction. The size of droplets can be conveniently controlled via from the droplets. In the case of nano DDS synthesis, one study reported
altering the ow rates of continuous and disperse phases. These drop- an EE of 2145% for PLGA-PEG NPs fabricated in bulk (emulsication-
lets subsequently are solidied to give microspheres or undergo further solvent diffusion) while a microuidics approach achieved an EE of
encapsulation to generate uniform core-shell structure. In the case of 2851% [146]. This could be explained by the effect of different mixing
nanocomplex synthesis, it is hypothesized that the charge neutraliza- rates. When the mixing rate is slow, the time scale of NP assembly is
tion between the cationic gene carrier and the negatively charged smaller than the time scale of solvent diffusion. Thus in bulk mixing
nucleic acid would be more complete by conning the two components NP starts assembling when the solvent concentration is still high and
in picoliter droplet, yielding nanocomplexes more uniform and compact results in drugs (e.g. docetaxel) escaping the encapsulation process. In
while exhausting any unreacted cationic gene carrier that is often cyto- the microuidics approach the time scales of the two processes are com-
toxic. b) In hydrodynamic focusing, the uid stream to be mixed owing parable and hence more drugs can be encapsulated within the carrier.
along the central microuidics channel is conned into a narrow stream This advantage would be of particular importance when one deals
after encountering two adjacent streams at higher ow rates. The diblock with expensive drugs.
copolymers or lipids dissolved in solvent self assembles as solubility de-
creases via solvent dilution through mixing with buffer in adjacent 4.2.5. Control of drug release rate
streams, the process of which takes place in micron scale. In theory, While the successful demonstration of microuidics-aided fabrication
the efciency of mixing by diffusion is length dependent. In reactors of different types of delivery vehicles is prevalent, there has been little
with a characteristic length scale larger than 1 mm, mixing by diffusion effort devoted to systematically characterizing the drug release kinetics
is inefcient whereas mixing time drops signicantly as diffusion dis- prole of the particles generated from a microuidics versus the con-
tance falls below 100 m [127]. Microuidic channels, usually designed ventional approach. The study carried out by Q. Xu et al. compared
with width of 100 m or less, represent an efcient and readily accessi- the kinetics of drug release of PLGA microparticles generated from the
ble reactor for rapid mixing of uids. The three stages of carrier fabrica- two approaches [157]. The monodisperse particles prepared using
tion by self-assembly (nucleation, growth through aggregation and microuidics release drug more slowly and have a smaller initial burst
stabilization after a characteristic aggregation time scale) are well con- effect than those observed in conventional polydisperse particles. The
trolled in the microuidics channel, resulting in carriers with highly authors attributed the effect to more uniform drug distribution in the
reduced polydispersity. microuidics-produced particles; hence drug release by degradation
of the former would be more gradual. Similarly, Karnik et al. reported
4.2.2. Control of particle shape the microuidic-aided self-assembled PLGA-PEG NPs had slower and
In addition to particle size, the effect of particle shape on drug carrier smaller initial burst of drug release than those fabricated by conven-
performance has recently gained more attention because of its inuence tional approaches [146]. Again rapid mixing pertaining to microuidics
on tissue biodistribution and endocytic uptake [183,184]. Conventional approach may lead to a more uniform drug distribution inside the
soft lithography can be used to fabricate non-spherical particles but the particles so that drug release is steadier by uniform degradation of the
throughput is limited by the mold size. Using a combined microscope particles. These studies serve to justify the use of microuidics over con-
projection lithography and microuidics approach, however, the fabrica- ventional approaches in controlling the drug release rate.
tion process for non-spherical objects could be rendered continuous
which obviously appeals to clinical translation and product manufactur- 4.3. Future perspective: scale-up production and versatility
ing [165]. In this approach, a mask with desired features is inserted in the
microscope to generate a mask-dened UV light beam projection on the As we have seen, microuidics platform shows promise to revolu-
monomer stream owing in a PDMS microuidics device above. Particles tionize particulate DDS synthesis. However, research development is
with desired shape are polymerized and advect along the unpolymerized merely the rst milestone before the process can be translated into
monomer stream to make way for subsequent polymerization to occur. industry-scale manufacturing. In order for the technique to be widely
Particles with a variety of shapes can be efciently generated. These adopted, two requirements must be satised: scalability and versatility.
examples showcase another attractive feature of microuidics-aided Industrial application of microuidics platform for drug carrier synthesis
drug carrier synthesis, one that can be leveraged to mass produce drug requires a high throughput production system. Effort has been made to
carriers with various shapes to optimize their biological performance. construct devices that support generation of particles/capsules at high
rates [143,185,186]. Incorporation of parallel generators in a single chip
4.2.3. Control of particle shell thickness is one way to achieve mass production. Up to 128 droplet generators
In microcapsule, the shell represents a diffusion barrier to control the can be fabricated in a 4 cm X 4 cm chip to produce a throughput of
sustained release from the capsule. The drug release prole associated 0.3 kg/h of monodisperse acrylic microspheres [185]. As miniaturization
with microcapsules of varying shell thickness would be inhomogeneous, is a key feature of microuidics device, tens or hundreds of these chips,
116 Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120

occupying far less space than conventional synthesis equipment, can be Conventional NP synthesis typically relies on bottomup approaches.
made to operate simultaneously to manufacture DDS. As discussed earlier, the capacity to achieve large size differences and
The drug fabrication process has also to be versatile in such a way shape variation is greatly limited by the nature of the self-assembly pro-
that DDS with diverse properties can be efciently fabricated and sorted. cess. In consequence, topdown methods that can produce particulate
Current microuidics-based synthesis is largely passive, where the parti- DDS with well-controlled size and shape are attractive.
cle properties are tuned by changing the ow rates of two immiscible
phases. One of the limitations is that any change at input has a long re- 5.1. Particle replication in non-wetting template (PRINT)
sponse time; for instance, transition from dripping to jetting in droplet
production sets a limit on the frequency of generation [187]. Recently PRINT (Fig. 6), rst introduced in 2005, is a topdown technique to
an automated active droplet production system was proposed to address fabricate monodisperse particles with precise particle structures [196].
the issue by using valves to externally control the infusion of two phases A non-wetting peruoropolyether (PFPE) elastomeric mold containing
so that the droplet volume and volume of continuous phase separating wells or cavities of predened shape and size is used to fabricate the par-
each droplet can be precisely and sharply tuned [187]. This can potential- ticles. Polymer liquid solution containing the cargo is conned in the
ly increase versatility of the microuidics drug carrier synthesis platform cavities by pressure applied between the mold and the PFPE surface,
as carrier properties can be properly adjusted. Throughput will suffer but followed by crosslinking or solvent evaporation. The low surface energy
likely be addressable in the future. of PFPE prevents the overow of polymer solution to non-cavities region,
Following the synthesis of drug carriers varying in size or shell thick- leading to well-isolated NP formation. With this method, particles from
ness, steps need to be taken to separate them according to their proper- 80 nm to 20 m have been fabricated with PLA, PEG hydrogels, proteins
ties. One of the attractive features of microuidics is the exibility for and stimuli-responsive polymers with disulde [197] or silyl ether linkers
multi-step processing; hence functions such as particle sorting can be in- [198]. Particles with various structures, such as circular disc, cube, rod,
corporated into the system conveniently. In fact, continuous particle cone, and others have been made [196,197,199], with cargos ranging
sorting based on size has been carried out in microuidics using optical from chemotherapeutics [200] to imaging uorophores [201]. With the
lattice [188], sedimentation [189], hydrodynamic ltration [190], and PRINT technology, Gratton et al. generated a library of PEG-based particles
lateral displacement [191]. In these examples, mixture of particles of dif- with a wide array of size and shape to study cellular uptake [183]. They
ferent sizes is channeled through the device, and subjected to physical also demonstrated that rod-like particles were more readily to be
force the magnitude of which depends on their size. Subsequently, the engulfed by Hela cells compared to particles of other structures.
separate streams containing particles identical in size are collected in dif-
ferent reservoirs. Furthermore, the particle-sorting step can also be used 5.2. Step-ash imprint lithography (S-FIL)
as quality control, which ensures the process of synthesizing drugs on
microuidics platform is robust and reproducible by generating drug S-FIL (Fig. 7) is a commercially available nanoimprint technique
carriers with properties that are within an acceptable range. Even though that uses a quartz template with predesigned patterns for particle
properties including particle size, shell thickness, drug loading level are synthesis. Polymer solution containing cross-linkable PEG-DA is added
already tightly controlled in the microuidics synthesis process, addition- to the cavities of template and polymerized via UV light. A PVA layer
al monitoring and screening steps would increase the value of the entire is deposited beneath the polymer layer and on top of a silica wafer for
drug processing system. The integration of high throughput particle syn- the release of imprinted particles. Glangchai et al. used this method to
thesis and sorting on chip can be seen as one of the greatest potential of fabricate stimuli-responsive NPs of 50400 nm and various shapes
microuidics-aided drug carrier synthesis platform, and ultimately be [202]. By incorporating an enzymatic degradable peptide GFLGK be-
adopted to advance DDS development (Fig. 5). tween PEG and diacrylate, the particles release plasmid DNA upon en-
zyme treatment in vitro. S-FIL relied on oxygen plasma treatment to
release the NPs from PVA layer [202]. On one hand, it does not involve
5. Topdown NP fabrication any mechanical stretch and maximally preserve the structure of NPs.
On the other, oxygen plasma generates large quantity of reactive oxy-
The physical properties of DDS would affect drug delivery efciency. gen species and free radicals, which could damage biological materials
While the inuence of particle size on drug delivery efciency has been such as DNA and protein and induces polymer degradation [203]. Fur-
well characterized, it is not until recently that the role of particle shape thermore, this method is restricted to photo-crosslinkable polymers
on drug delivery has been revealed. Rod-like structures demonstrate and two dimensional shapes.
the highest cellular uptake efciency, followed by spheres, cylinders
and cubes [183,184]. Shape-specic inuence on particle circulation 5.3. Microuidics-based topdown approaches
has also been discussed. Filamentous micelles have been shown to cir-
culate 10 times longer than their spherical counterpart [192]. Nonethe- Dendukuri et al. used continuous ow photolithography (CFL)
less, the precise role of particle shape in drug delivery has not been (Fig. 8) to fabricate particles of complex shapes [165]. A stream of
elucidated due to the limited studies available. This is partly attributed PEGDA containing a photoinitiator is continuously channeled through a
to the lack of easy and robust methods in producing NPs with various microuidic device. A photomask with dened shapes is placed under-
shapes [193]. neath the channel while pulses of UV light are applied. Particles of dened
Early attempts to study the effect of particle shape on bioavailability shapes are formed via cross-linking and ushed to the outlet for collection
have stretched spherical NPs into ellipsoidal particles [194]. Embedded because of the continuous ow. This method eliminates the requirement
in a PVA lm, the spherical particles deform when the lm is heated, of further harvesting steps and the particles could be puried by centrifu-
stretched, and quenched. The heating temperature required to deform gation and re-suspension in water. However, due to the continuous ow
the particles is determined by the glass transition temperature of the nature, the nal shape is usually deformed from the designed structure,
polymeric carrier, which is around 90 C for PLGA and 140 C for poly- reducing geometrical resolution of the particles. To increase the resolu-
styrene (PS) [195]. The high temperature greatly limits the type of bio- tion, the same group has developed stop ow lithography technique
active agents that could be encapsulated in these deformed particles. (SFL) [204,205]. In SFL, a three-way solenoid valve is applied to control
One advantage of this method is that it produces non-spherical particles the ow inside the channels to stop briey during each exposure for
with exactly the same volume. Nonetheless, the process is highly the polymerization to complete. Complex shapes with high resolution
material-specic and has only been demonstrated for PLGA and PS par- are formed, including toroid, hexongonal and worm-like structures. The
ticles with very limited scalability [184]. high resolution and ease of harvesting and purication render this
Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120 117

method a promising tool for topdown particle synthesis. However, it is [11] H.S. Yoo, K.H. Lee, J.E. Oh, T.G. Park, In vitro and in vivo anti-tumor activities of
nanoparticles based on doxorubicin-PLGA conjugates, J. Control. Release 68
also limited to photo-crosslinkable systems. In addition, solvent vulnera- (2000) 419431.
bility of PDMS channels poses severe barrier for extension of the method [12] K. Avgoustakis, A. Beletsi, Z. Panagi, P. Klepetsanis, A.G. Karydas, D.S. Ithakissios,
to fabricate other particles. The dimensions of particles synthesized from PLGA-mPEG nanoparticles of cisplatin: in vitro nanoparticle degradation, in vitro
drug release and in vivo drug residence in blood properties, J. Control. Release 79
this method are in the micrometer scale, which is primarily restricted by (2002) 123135.
the resolution of current photolithography techniques. [13] G. Fontana, M. Licciardi, S. Mansueto, D. Schillaci, G. Giammona, Amoxicillin-loaded
polyethylcyanoacrylate nanoparticles: inuence of PEG coating on the particle size,
drug release rate and phagocytic uptake, Biomaterials 22 (2001) 28572865.
[14] H. Suh, B. Jeong, R. Rathi, S.W. Kim, Regulation of smooth muscle cell proliferation
6. Challenges and future perspectives
using paclitaxel-loaded poly(ethylene oxide)-poly(lactide/glycolide) nanospheres,
J. Biomed. Mater. Res. 42 (1998) 331338.
In summary, novel materials synthesis, introduction of [15] G.P. Carino, J.S. Jacob, E. Mathiowitz, Nanosphere based oral insulin delivery,
environmental-sensitive polymers, successful adoption of natural poly- J. Control. Release 65 (2000) 261269.
[16] J. Panyam, M.M. Dali, S.K. Sahoo, W. Ma, S.S. Chakravarthi, G.L. Amidon, R.J. Levy, V.
mers as carrier and improved understanding of the structurefunction Labhasetwar, Polymer degradation and in vitro release of a model protein from
relationship have together transformed DDS development. The parallel poly(D, L-lactide-co-glycolide) nano- and microparticles, J. Control. Release 92
advancement in bottomup and topdown nanofabrication enables pre- (2003) 173187.
[17] M. van de Weert, W.E. Hennink, W. Jiskoot, Protein instability in poly(lactic-
cise control over size and shape of particulate DDS. Moreover, the use of co-glycolic acid) microparticles, Pharm. Res. 17 (2000) 11591167.
microuidics in formulating DDS opens up the possibility for automation [18] J. Panyam, W.Z. Zhou, S. Prabha, S.K. Sahoo, V. Labhasetwar, Rapid endo-lysosomal
and scaling up, as well as on-chip particle screening and analysis. escape of poly(DL-lactide-co-glycolide) nanoparticles: implications for drug and
gene delivery, FASEB J. 16 (2002) 12171226.
However, many challenges remain. From the materials perspective, [19] S. Prabha, V. Labhasetwar, Critical determinants in PLGA/PLA nanoparticle-mediated
the majority of smart delivery systems works well in vitro but fails in gene expression, Pharm. Res. 21 (2004) 354364.
more complicated in vivo environment. Furthermore, the challenge to [20] P. Elamanchili, C.M. Lutsiak, S. Hamdy, M. Diwan, J. Samuel, Pathogen-mimicking
nanoparticles for vaccine delivery to dendritic cells, J. Immunother. 30 (2007)
integrate programmability, targetability, and environmental respon- 378395.
siveness remains formidable. From the fabrication perspective, conven- [21] J.M. Lu, X. Wang, C. Marin-Muller, H. Wang, P.H. Lin, Q. Yao, C. Chen, Current ad-
tional techniques have the advantage of easy scale-up, but lose accuracy vances in research and clinical applications of PLGA-based nanotechnology, Expert.
Rev. Mol. Diagn. 9 (2009) 325341.
in control over particle characteristics. Topdown approaches, in con-
[22] N. Faisant, J. Siepmann, J.P. Benoit, PLGA-based microparticles: elucidation of
trast, offer precise control over particle size and shape. However, up mechanisms and a new, simple mathematical model quantifying drug release,
till now they are applicable to only a few systems, and with the excep- Eur. J. Pharm. Sci. 15 (2002) 355366.
tion of PRINT are difcult to scale up. Although microuidics holds [23] G. Crotts, T.G. Park, Protein delivery from poly(lactic-co-glycolic acid) biode-
gradable microspheres: release kinetics and stability issues, J. Microencapsul.
promise in addressing these issues, current lithographic techniques 15 (1998) 699713.
limit the manipulation of uids in the micro-scale. In the context of [24] H. Kranz, N. Ubrich, P. Maincent, R. Bodmeier, Physicomechanical properties of
nanomedicine, nanouidics would be desirable for DDS fabrication. biodegradable poly(D, L-lactide) and poly(D, L-lactide-co-glycolide) lms in
the dry and wet states, J. Pharm. Sci. 89 (2000) 15581566.
When innovations in fabrication catch up with those of materials de- [25] H. Shirahama, A. Ichimaru, C. Tsutsumi, Y. Nakayama, H. Yasuda, Characteristics
sign, the goal for constructing and translating multifunctional, precisely of the biodegradability and physical properties of stereocomplexes between
controlled, and biocompatible DDS will be one step closer to reality. poly(L-lactide) and poly(D-lactide) copolymers, J. Polymer Sci., Part A: Polymer
Chem. 43 (2005) 438454.
[26] H. Tsuji, Poly(lactide) stereocomplexes: formation, structure, properties, degrada-
tion, and applications, Macromol. Biosci. 5 (2005) 569597.
Acknowledgments [27] J.P. Tan, S.H. Kim, F. Nederberg, E.A. Appel, R.M. Waymouth, Y. Zhang, J.L.
Hedrick, Y.Y. Yang, Hierarchical supermolecular structures for sustained drug
release, Small 5 (2009) 15041507.
This work is funded by NSF EEC-0425626, NIH HL089764 and
[28] H.C. Lawson, P. Sampath, E. Bohan, M.C. Park, N. Hussain, A. Olivi, J. Weingart, L.
HL109442. YZ and HFC are grateful for fellowship support from the Kleinberg, H. Brem, Interstitial chemotherapy for malignant gliomas: the Johns
Agency for Science, Technology and Research (Singapore) and the Hopkins experience, J. Neurooncol. 83 (2007) 6170.
[29] H.C. Bock, M.J. Puchner, F. Lohmann, M. Schutze, S. Koll, R. Ketter, R. Buchalla, N.
Sir Edward Youde Memorial Fund Council (Hong Kong), respectively.
Rainov, S.R. Kantelhardt, V. Rohde, A. Giese, First-line treatment of malignant
glioma with carmustine implants followed by concomitant radiochemotherapy:
a multicenter experience, Neurosurg. Rev. 33 (2010) 441449.
References [30] Z. Zhao, J. Wang, H.Q. Mao, K.W. Leong, Polyphosphoesters in drug and gene delivery,
Adv. Drug Deliv. Rev. 55 (2003) 483499.
[1] T.M. Allen, P.R. Cullis, Drug delivery systems: entering the mainstream, Science [31] Y.C. Wang, Y.Y. Yuan, J.Z. Du, X.Z. Yang, J. Wang, Recent progress in
303 (2004) 18181822. polyphosphoesters: from controlled synthesis to biomedical applications,
[2] R.K.G. Verma, Sanjay current status of drug delivery technologies and future direc- Macromol. Biosci. 9 (2009) 11541164.
tions, Pharm. Technol. On-Line 25 (2001) 4. [32] S.L. Bourke, J. Kohn, Polymers derived from the amino acid L-tyrosine:
[3] A.J. Almeida, E. Souto, Solid lipid nanoparticles as a drug delivery system for polycarbonates, polyarylates and copolymers with poly(ethylene glycol), Adv.
peptides and proteins, Adv. Drug Deliv. Rev. 59 (2007) 478490. Drug Deliv. Rev. 55 (2003) 447466.
[4] L. Zhang, D. Pornpattananangku, C.M. Hu, C.M. Huang, Development of nanoparticles [33] J.M. Karp, R. Langer, Development and therapeutic applications of advanced bio-
for antimicrobial drug delivery, Curr. Med. Chem. 17 (2010) 585594. materials, Curr. Opin. Biotechnol. 18 (2007) 454459.
[5] M.L. Schipper, G. Iyer, A.L. Koh, Z. Cheng, Y. Ebenstein, A. Aharoni, S. Keren, L.A. [34] R. Langer, D.A. Tirrell, Designing materials for biology and medicine, Nature 428
Bentolila, J. Li, J. Rao, X. Chen, U. Banin, A.M. Wu, R. Sinclair, S. Weiss, S.S. Gambhir, (2004) 487492.
Particle size, surface coating, and PEGylation inuence the biodistribution of quan- [35] A.S. Hoffman, The origins and evolution of controlled drug delivery systems,
tum dots in living mice, Small 5 (2009) 126134. J. Control. Release 132 (2008) 153163.
[6] H. Carrstensen, R.H. Muller, B.W. Muller, Particle size, surface hydrophobicity [36] J. Gong, M. Chen, Y. Zheng, S. Wang, Y. Wang, Polymeric micelles drug delivery
and interaction with serum of parenteral fat emulsions and model drug carriers system in oncology, J. Control. Release 159 (2012) 312323.
as parameters related to RES uptake, Clin. Nutr. 11 (1992) 289297. [37] A. Harada, K. Kataoka, Formation of polyion complex micelles in an aqueous milieu
[7] H. Cabral, Y. Matsumoto, K. Mizuno, Q. Chen, M. Murakami, M. Kimura, Y. from a pair of oppositely-charged block-copolymers with poly(ethylene glycol)
Terada, M.R. Kano, K. Miyazono, M. Uesaka, N. Nishiyama, K. Kataoka, Accumu- segments, Macromolecules 28 (1995) 52945299.
lation of sub-100 nm polymeric micelles in poorly permeable tumours depends [38] A. Lavasanifar, J. Samuel, S. Sattari, G.S. Kwon, Block copolymer micelles for the
on size, Nat. Nanotechnol. 6 (2011) 815823. encapsulation and delivery of amphotericin B, Pharm. Res. 19 (2002) 418422.
[8] H.K. Sajja, M.P. East, H. Mao, Y.A. Wang, S. Nie, L. Yang, Development of multi- [39] A. Falamarzian, A. Lavasanifar, Chemical modication of hydrophobic block in
functional nanoparticles for targeted drug delivery and noninvasive imaging of poly(ethylene oxide) poly(caprolactone) based nanocarriers: effect on the solu-
therapeutic effect, Curr. Drug Discov. Technol. 6 (2009) 4351. bilization and hemolytic activity of amphotericin B, Macromol. Biosci. 10 (2010)
[9] R. Jain, N.H. Shah, A.W. Malick, C.T. Rhodes, Controlled drug delivery by biode- 648656.
gradable poly(ester) devices: different preparative approaches, Drug Dev. Ind. [40] A. Falamarzian, A. Lavasanifar, Optimization of the hydrophobic domain in
Pharm. 24 (1998) 703727. poly(ethylene oxide)-poly(varepsilon-caprolactone) based nano-carriers for
[10] R.A. Jain, The manufacturing techniques of various drug loaded biodegradable the solubilization and delivery of Amphotericin B, Colloids Surf. B Biointerfaces
poly(lactide-co-glycolide) (PLGA) devices, Biomaterials 21 (2000) 24752490. 81 (2010) 313320.
118 Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120

[41] S.H. Kim, J.P. Tan, F. Nederberg, K. Fukushima, J. Colson, C. Yang, A. Nelson, Y.Y. [67] J. Azzi, L. Tang, R. Moore, R. Tong, N. El Haddad, T. Akiyoshi, B. Mfarrej, S.M. Yang, M.
Yang, J.L. Hedrick, Hydrogen bonding-enhanced micelle assemblies for drug de- Jurewicz, T. Ichimura, N. Lindeman, J.J. Cheng, R. Abdi, Polylactide-cyclosporin A
livery, Biomaterials 31 (2010) 80638071. nanoparticles for targeted immunosuppression, FASEB J. 24 (2010) 39273938.
[42] M. Iijima, Y. Nagasaki, T. Okada, M. Kato, K. Kataoka, Core-polymerized reactive [68] K.J. Chen, L. Tang, M.A. Garcia, H. Wang, H. Lu, W.Y. Lin, S. Hou, Q. Yin, C.K.F. Shen, J.J.
micelles from heterotelechelic amphiphilic block copolymers, Macromolecules Cheng, H.R. Tseng, The therapeutic efcacy of camptothecin-encapsulated supra-
32 (1999) 11401146. molecular nanoparticles, Biomaterials 33 (2012) 11621169.
[43] J.H. Kim, K. Emoto, M. Iijima, Y. Nagasaki, T. Aoyagi, T. Okano, Y. Sakurai, K. [69] C.C. Williams, S.H. Thang, T. Hantke, U. Vogel, P.H. Seeberger, J. Tsanaktsidis, B.
Kataoka, Core-stabilized polymeric micelle as potential drug carrier: Increased Lepenies, RAFT-derived polymerdrug conjugates: poly(hydroxypropyl methacry-
solubilization of taxol, Polym. Adv. Technol. 10 (1999) 647654. lamide) (HPMA)-7-ethyl-10-hydroxycamptothecin (SN-38) conjugates, ChemMed-
[44] Y. Kakizawa, A. Harada, K. Kataoka, Environment-sensitive stabilization of Chem 7 (2012) 281291.
core-shell structured polyion complex micelle by reversible cross-linking of [70] Z. Liu, Y. Jiao, Y. Wang, C. Zhou, Z. Zhang, Polysaccharides-based nanoparticles as
the core through disulde bond, J. Am. Chem. Soc. 121 (1999) 1124711248. drug delivery systems, Adv. Drug Deliv. Rev. 60 (2008) 16501662.
[45] R. Nagarajan, M. Barry, Unusual Selectivity in Solubilization by Block Copolymer [71] O. Felt, P. Buri, R. Gurny, Chitosan: a unique polysaccharide for drug delivery,
Micelles, in: Abstr Pap Am Chem S, 191, 1986, p. 287-COLL. Drug Dev. Ind. Pharm. 24 (1998) 979993.
[46] X. Shuai, H. Ai, N. Nasongkla, S. Kim, J. Gao, Micellar carriers based on block co- [72] P.M. Friden, L.R. Walus, G.F. Musso, M.A. Taylor, B. Malfroy, R.M. Starzyk,
polymers of poly(epsilon-caprolactone) and poly(ethylene glycol) for doxorubi- Anti-transferrin receptor antibody and antibodydrug conjugates cross the
cin delivery, J. Control. Release 98 (2004) 415426. bloodbrain barrier, Proc. Natl. Acad. Sci. U. S. A. 88 (1991) 47714775.
[47] Y. Wang, S. Gao, W.H. Ye, H.S. Yoon, Y.Y. Yang, Co-delivery of drugs and DNA [73] H. Li, Z.M. Qian, Transferrin/transferrin receptor-mediated drug delivery, Med.
from cationic core-shell nanoparticles self-assembled from a biodegradable co- Res. Rev. 22 (2002) 225250.
polymer, Nat. Mater. 5 (2006) 791796. [74] Z.M. Qian, H. Li, H. Sun, K. Ho, Targeted drug delivery via the transferrin
[48] T.M. Sun, J.Z. Du, Y.D. Yao, C.Q. Mao, S. Dou, S.Y. Huang, P.Z. Zhang, K.W. Leong, receptor-mediated endocytosis pathway, Pharmacol. Rev. 54 (2002) 561587.
E.W. Song, J. Wang, Simultaneous delivery of siRNA and paclitaxel via a [75] Y.F. Huang, D. Shangguan, H. Liu, J.A. Phillips, X. Zhang, Y. Chen, W. Tan, Molec-
two-in-one micelleplex promotes synergistic tumor suppression, ACS Nano 5 ular assembly of an aptamerdrug conjugate for targeted drug delivery to tumor
(2011) 14831494. cells, ChemBioChem 10 (2009) 862868.
[49] T.L. Kaneshiro, Z.R. Lu, Targeted intracellular codelivery of chemotherapeutics [76] V. Bagalkot, O.C. Farokhzad, R. Langer, S. Jon, An aptamerdoxorubicin physical
and nucleic acid with a well-dened dendrimer-based nanoglobular carrier, Bio- conjugate as a novel targeted drug-delivery platform, Angew. Chem. Int. Ed.
materials 30 (2009) 56605666. 45 (2006) 81498152.
[50] C. Zhu, S. Jung, S. Luo, F. Meng, X. Zhu, T.G. Park, Z. Zhong, Co-delivery of siRNA and [77] T.H.L.H. Li, K.W. Leong, Nucleic acid-based nanoengineering: novel structures for
paclitaxel into cancer cells by biodegradable cationic micelles based on PDMAEMA- biomedical applications, Interface Focus 1 (2011) 702724.
PCL-PDMAEMA triblock copolymers, Biomaterials 31 (2010) 24082416. [78] S. Ko, H. Liu, Y. Chen, C. Mao, DNA nanotubes as combinatorial vehicles for cel-
[51] X.B. Xiong, A. Lavasanifar, Traceable multifunctional micellar nanocarriers for lular delivery, Biomacromolecules 9 (2008) 30393043.
cancer-targeted co-delivery of MDR-1 siRNA and doxorubicin, ACS Nano 5 [79] O.I. Wilner, R. Orbach, A. Henning, C. Teller, O. Yehezkeli, M. Mertig, D. Harries, I.
(2011) 52025213. Willner, Self-assembly of DNA nanotubes with controllable diameters, Nat.
[52] L. Fan, F. Li, H. Zhang, Y. Wang, C. Cheng, X. Li, C.H. Gu, Q. Yang, H. Wu, S. Zhang, Commun. 2 (2011) 540.
Co-delivery of PDTC and doxorubicin by multifunctional micellar nanoparticles [80] E.S. Andersen, M. Dong, M.M. Nielsen, K. Jahn, R. Subramani, W. Mamdouh, M.M.
to achieve active targeted drug delivery and overcome multidrug resistance, Golas, B. Sander, H. Stark, C.L. Oliveira, J.S. Pedersen, V. Birkedal, F. Besenbacher,
Biomaterials 31 (2010) 56345642. K.V. Gothelf, J. Kjems, Self-assembly of a nanoscale DNA box with a controllable
[53] N. Wiradharma, Y. Zhang, S. Venkataraman, J.L. Hedrick, Y.Y. Yang, Self-assembled lid, Nature 459 (2009) 7376.
polymer nanostructures for delivery of anticancer therapeutics, Nano Today 4 [81] Y.H. Roh, J.B. Lee, P. Kiatwuthinon, M.R. Hartman, J.J. Cha, S.H. Um, D.A. Muller, D.
(2009) 302317. Luo, DNAsomes: multifunctional DNA-based nanocarriers, Small 7 (2011)
[54] R. Duncan, Polymer conjugates as anticancer nanomedicines, Nat. Rev. Cancer 6 7478.
(2006) 688701. [82] J. Li, H. Pei, B. Zhu, L. Liang, M. Wei, Y. He, N. Chen, D. Li, Q. Huang, C. Fan,
[55] M.J. Vicent, H. Ringsdorf, R. Duncan, Polymer therapeutics: clinical applications Self-assembled multivalent DNA nanostructures for noninvasive intracellular
and challenges for development Preface, Adv. Drug Deliv. Rev. 61 (2009) delivery of immunostimulatory CpG oligonucleotides, ACS Nano 5 (2011)
11171120. 87838789.
[56] J. Kopecek, H. Bazilova, Poly[N-(2-hydroxypropyl)methacrylamide].1. Radical [83] K.A. Afonin, W.W. Grabow, F.M. Walker, E. Bindewald, M.A. Dobrovolskaia, B.A.
polymerization and copolymerization, Eur. Polym. J. 9 (1973) 714. Shapiro, L. Jaeger, Design and self-assembly of siRNA-functionalized RNA nano-
[57] P.A. Vasey, S.B. Kaye, R. Morrison, C. Twelves, P. Wilson, R. Duncan, A.H. particles for use in automated nanomedicine, Nat. Protoc. 6 (2011) 20222034.
Thomson, L.S. Murray, T.E. Hilditch, T. Murray, S. Burtles, D. Fraier, E. Frigerio, [84] S.K. Alahari, N.M. Dean, M.H. Fisher, R. Delong, M. Manoharan, K.L. Tivel, R.L. Juliano,
J. Cassidy, C.R.C.P.I.I. Comm, Phase I clinical and pharmacokinetic study of PK1 Inhibition of expression of the multidrug resistance-associated P-glycoprotein of
[N-(2-hydroxypropyl)methacrylamide copolymer doxorubicin]: First member by phosphorothioate and 5 cholesterol-conjugated phosphorothioate antisense
of a new class of chemotherapeutic agents drugpolymer conjugates, Clin. oligonucleotides, Mol. Pharmacol. 50 (1996) 808819.
Cancer Res. 5 (1999) 8394. [85] J.H. Jeong, H. Mok, Y.K. Oh, T.G. Park, siRNA conjugate delivery systems,
[58] C. Li, D.F. Yu, R.A. Newman, F. Cabral, L.C. Stephens, N. Hunter, L. Milas, S. Wallace, Bioconjug. Chem. 20 (2009) 514.
Complete regression of well-established tumors using a novel water-soluble [86] J.J. Cronican, D.B. Thompson, K.T. Beier, B.R. McNaughton, C.L. Cepko, D.R. Liu,
poly(L-glutamic acid) paclitaxel conjugate, Cancer Res. 58 (1998) 24042409. Potent delivery of functional proteins into mammalian cells in vitro and in vivo
[59] J.W. Singer, B. Baker, P. De Vries, A. Kumar, S. Shaffer, E. Vawter, M. Bolton, P. using a supercharged protein, ACS Chem. Biol. 5 (2010) 747752.
Garzone, Poly-(L)-glutamic acid-paclitaxel (CT-2103) [XYOTAX (TM)], a biode- [87] B.R. McNaughton, J.J. Cronican, D.B. Thompson, D.R. Liu, Mammalian cell penetra-
gradable polymeric drug conjugate characterization, preclinical pharmacolo- tion, siRNA transfection, and DNA transfection by supercharged proteins, Proc.
gy, and preliminary clinical data, Polym. Drugs Clin. Stage Advant. Prospects Natl. Acad. Sci. U. S. A. 106 (2009) 61116116.
519 (2003) 8199. [88] J.L. Frandsen, H. Ghandehari, Recombinant protein-based polymers for advanced
[60] R. Bhatt, P. de Vries, J. Tulinsky, G. Bellamy, B. Baker, J.W. Singer, P. Klein, Synthe- drug delivery, Chem. Soc. Rev. 41 (2012) 26962706.
sis and in vivo antitumor activity of poly(l-glutamic acid) conjugates of [89] J.A. MacKay, M. Chen, J.R. McDaniel, W. Liu, A.J. Simnick, A. Chilkoti, Self-assembling
20S-camptothecin, J. Med. Chem. 46 (2003) 190193. chimeric polypeptide-doxorubicin conjugate nanoparticles that abolish tumours
[61] G.M. Springett, C. Takimoto, M. McNamara, J.H. Doroshow, S. Syed, E. Eastham, D. after a single injection, Nat. Mater. 8 (2009) 993999.
Spriggs, S. Pezzulli, G. Michelson, J. Dupont, Phase I study of CT-2106 (polyglutamate [90] D.E. Meyer, B.C. Shin, G.A. Kong, M.W. Dewhirst, A. Chilkoti, Drug targeting using
camptothecin) in patients with advanced malignancies, in: 2004 ASCO Annu. Meet. thermally responsive polymers and local hyperthermia, J. Control. Release 74
Proc (Post-Meeting Edition), J. Clin. Oncol., 22, 2004, p. 3127. (2001) 213224.
[62] C.J. Langer, K.J. O'Byrne, M.A. Socinski, S.M. Mikhailov, K. Lesniewski-Kmak, M. [91] M.R. Dreher, W. Liu, C.R. Michelich, M.W. Dewhirst, A. Chilkoti, Thermal cycling en-
Smakal, T.E. Ciuleanu, S.V. Orlov, M. Dediu, D. Heigener, A.J. Eisenfeld, L. Sandalic, hances the accumulation of a temperature-sensitive biopolymer in solid tumors,
F.B. Oldham, J.W. Singer, H.J. Ross, Phase III trial comparing paclitaxel poliglumex Cancer Res. 67 (2007) 44184424.
(CT-2103, PPX) in combination with carboplatin versus standard paclitaxel and [92] M.R. Dreher, A.J. Simnick, K. Fischer, R.J. Smith, A. Patel, M. Schmidt, A. Chilkoti,
carboplatin in the treatment of PS 2 patients with chemotherapy-naive advanced Temperature triggered self-assembly of polypeptides into multivalent spherical
non-small cell lung cancer, J. Thorac. Oncol. 3 (2008) 623630. micelles, J. Am. Chem. Soc. 130 (2008) 687694.
[63] L. Paz-Ares, H. Ross, M. O'Brien, A. Riviere, U. Gatzemeier, J. Von Pawel, E. Kaukel, L. [93] R.E. Sallach, W. Cui, J. Wen, A. Martinez, V.P. Conticello, E.L. Chaikof,
Freitag, W. Digel, H. Bischoff, R. Garcia-Campelo, N. Iannotti, P. Reiterer, I. Bover, J. Elastin-mimetic protein polymers capable of physical and chemical crosslinking,
Prendiville, A.J. Eisenfeld, F.B. Oldham, B. Bandstra, J.W. Singer, P. Bonomi, Phase Biomaterials 30 (2009) 409422.
III trial comparing paclitaxel poliglumex vs docetaxel in the second-line treatment [94] W. Kim, E.L. Chaikof, Recombinant elastin-mimetic biomaterials: emerging ap-
of non-small-cell lung cancer, Br. J. Cancer 98 (2008) 16081613. plications in medicine, Adv. Drug Deliv. Rev. 62 (2010) 14681478.
[64] F. Canal, J. Sanchis, M.J. Vicent, Polymerdrug conjugates as nano-sized medi- [95] S.W. Jordan, C.A. Haller, R.E. Sallach, R.P. Apkarian, S.R. Hanson, E.L. Chaikof, The
cines, Curr. Opin. Biotechnol. 22 (2011) 894900. effect of a recombinant elastin-mimetic coating of an ePTFE prosthesis on acute
[65] R. Tong, J.J. Cheng, Paclitaxel-initiated, controlled polymerization of lactide for thrombogenicity in a baboon arteriovenous shunt, Biomaterials 28 (2007)
the formulation of polymeric nanoparticulate delivery vehicles, Angew. Chem. 11911197.
Int. Ed. Engl. 47 (2008) 48304834. [96] X. Wu, R.E. Sallach, J.M. Caves, V.P. Conticello, E.L. Chaikof, Deformation responses
[66] R. Tong, J.J. Cheng, Ring-opening polymerization-mediated controlled formulation of a physically cross-linked high molecular weight elastin-like protein polymer,
of polylactide-drug nanoparticles, J. Am. Chem. Soc. 131 (2009) 47444754. Biomacromolecules 9 (2008) 17871794.
Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120 119

[97] K. Numata, B. Subramanian, H.A. Currie, D.L. Kaplan, Bioengineered silk protein-based [128] W.J. Duncanson, T. Lin, A.R. Abate, S. Seiffert, R.K. Shah, D.A. Weitz, Microuidic
gene delivery systems, Biomaterials 30 (2009) 57755784. synthesis of advanced microparticles for encapsulation and controlled release,
[98] K. Numata, D.L. Kaplan, Silk-based delivery systems of bioactive molecules, Adv. Lab Chip 12 (2012) 21352145.
Drug Deliv. Rev. 62 (2010) 14971508. [129] D. Lensen, K. van Breukelen, D.M. Vriezema, J.C. van Hest, Preparation of biode-
[99] K. Numata, D.L. Kaplan, Silk-based gene carriers with cell membrane destabilizing gradable liquid core PLLA microcapsules and hollow PLLA microcapsules using
peptides, Biomacromolecules 11 (2010) 31893195. microuidics, Macromol. Biosci. 10 (2010) 475480.
[100] S. Kim, J.H. Kim, O. Jeon, I.C. Kwon, K. Park, Engineered polymers for advanced [130] H. Okochi, M. Nakano, Preparation and evaluation of w/o/w type emulsions
drug delivery, Eur. J. Pharm. Biopharm. 71 (2009) 420430. containing vancomycin, Adv. Drug Deliv. Rev. 45 (2000) 526.
[101] J. He, X. Qi, Y. Miao, H.L. Wu, N. He, J.J. Zhu, Application of smart nanostructures [131] A.S. Utada, E. Lorenceau, D.R. Link, P.D. Kaplan, H.A. Stone, D.A. Weitz, Monodis-
in medicine, Nanomedicine (Lond.) 5 (2010) 11291138. perse double emulsions generated from a microcapillary device, Science 308
[102] Y.S. Tarahovsky, Smart liposomal nanocontainers in biology and medicine, (2005) 537541.
Biochemistry (Mosc) 75 (2010) 811824. [132] T. Endres, M. Zheng, M. Beck-Broichsitter, O. Samsonova, H. Debus, T. Kissel,
[103] S.R. MacEwan, D.J. Callahan, A. Chilkoti, Stimulus-responsive macromolecules Optimising the self-assembly of siRNA loaded PEG-PCL-lPEI nano-carriers employing
and nanoparticles for cancer drug delivery, Nanomedicine (Lond.) 5 (2010) different preparation techniques, J. Control. Release 160 (2012) 583591.
793806. [133] J.W. Kim, A.S. Utada, A. Fernandez-Nieves, Z. Hu, D.A. Weitz, Fabrication of mono-
[104] D. Schmaljohann, Thermo- and pH-responsive polymers in drug delivery, Adv. disperse gel shells and functional microgels in microuidic devices, Angew.
Drug Deliv. Rev. 58 (2006) 16551670. Chem. Int. Ed. Engl. 46 (2007) 18191822.
[105] S. Brocchini, K. James, V. Tangpasuthadol, J. Kohn, A combinatorial approach for [134] R.K. Shah, J.W.K., J.J. Agresti, D.A. Weitz, L.Y. Chu, Fabrication of monodisperse
polymer design, J. Am. Chem. Soc. 119 (1997) 45534554. thermosensitive microgels and gel capsules in microuidic devices, Soft Matter
[106] C.E. Mora-Huertas, H. Fessi, A. Elaissari, Polymer-based nanocapsules for drug 4 (2008) 23032309.
delivery, Int. J. Pharm. 385 (2010) 113142. [135] D.N. Breslauer, S.J. Muller, L.P. Lee, Generation of monodisperse silk micro-
[107] K.T. Love, K.P. Mahon, C.G. Levins, K.A. Whitehead, W. Querbes, J.R. Dorkin, J. Qin, spheres prepared with microuidics, Biomacromolecules 11 (2010) 643647.
W. Cantley, L.L. Qin, T. Racie, M. Frank-Kamenetsky, K.N. Yip, R. Alvarez, D.W. [136] L.Y. Chu, J.Y.K., R.K. Shah, D.A. Weitz, Monodisperse thermoresponsive microgels with
Sah, A. de Fougerolles, K. Fitzgerald, V. Koteliansky, A. Akinc, R. Langer, D.G. tunable volume-phase transition kinetics, Adv. Funct. Mater. 17 (2007) 34993504.
Anderson, Lipid-like materials for low-dose, in vivo gene silencing, Proc. Natl. [137] W. Wang, R. Xie, X.J. Ju, T. Luo, L. Liu, D.A. Weitz, L.Y. Chu, Controllable
Acad. Sci. U. S. A. 107 (2010) 18641869. microuidic production of multicomponent multiple emulsions, Lab Chip 11
[108] R. Weissleder, K. Kelly, E.Y. Sun, T. Shtatland, L. Josephson, Cell-specic targeting (2011) 15871592.
of nanoparticles by multivalent attachment of small molecules, Nat. Biotechnol. [138] L.Y. Chu, A.S. Utada, R.K. Shah, J.W. Kim, D.A. Weitz, Controllable monodisperse
23 (2005) 14181423. multiple emulsions, Angew. Chem. Int. Ed. Engl. 46 (2007) 89708974.
[109] C. Vauthier, K. Bouchemal, Methods for the preparation and manufacture of [139] S.H. Kim, J.W. Kim, J.C. Cho, D.A. Weitz, Double-emulsion drops with ultra-thin
polymeric nanoparticles, Pharm. Res. 26 (2009) 10251058. shells for capsule templates, Lab Chip 11 (2011) 31623166.
[110] F. Ganachaud, J.L. Katz, Nanoparticles and nanocapsules created using the Ouzo [140] P.W. Ren, X.J. Ju, R. Xie, L.Y. Chu, Monodisperse alginate microcapsules with oil
effect: spontaneous emulsication as an alternative to ultrasonic and high-shear core generated from a microuidic device, J. Colloid Interface Sci. 343 (2010)
devices, ChemPhysChem 6 (2005) 209216. 392395.
[111] C. Duclairoir, E. Nakache, H. Marchais, A.M. Orecchioni, Formation of gliadin [141] R.K. Shah, J.W. Kim, D.A. Weitz, Monodisperse stimuli-responsive colloido-
nanoparticles: inuence of the solubility parameter of the protein solvent, Col- somes by self-assembly of microgels in droplets, Langmuir 26 (2010) 1561
loid Polym. Sci. 276 (1998) 321327. 1565.
[112] M. Skiba, D. Wouessidjewe, F. Puisieux, D. Duchene, A. Gulik, Characterization [142] L. Liu, J.P. Yang, X.J. Ju, R. Xie, Y.M. Liu, W. Wang, J.J. Zhang, C.H. Niu, L.Y. Chu,
of amphiphilic beta-cyclodextrin nanospheres, Int. J. Pharm. 142 (1996) 121 Monodisperse core-shell chitosan microcapsules for pH-responsive burst re-
124. lease of hydrophobic drugs, Soft Matter 7 (2011) 48214827.
[113] J.W. McGinity, P.B. O'Donnell, Preparation of microspheres by the solvent evapora- [143] M.B. Romanowsky, A.R. Abate, A. Rotem, C. Holtze, D.A. Weitz, High throughput
tion technique, Adv. Drug Deliv. Rev. 28 (1997) 2542. production of single core double emulsions in a parallelized microuidic device,
[114] S. Desgouilles, C. Vauthier, D. Bazile, J. Vacus, J.-L. Grossiord, M. Veillard, P. Lab Chip 12 (2012) 802807.
Couvreur, The design of nanoparticles obtained by solvent evaporation: a com- [144] A.T. Hsieh, N. Hori, R. Massoudi, P.J. Pan, H. Sasaki, Y.A. Lin, A.P. Lee, Nonviral gene
prehensive study, Langmuir 19 (2003) 95049510. vector formation in monodispersed picolitre incubator for consistent gene delivery,
[115] D. Lemoine, V. Preat, Polymeric nanoparticles as delivery system for inuenza Lab Chip 9 (2009) 26382643.
virus glycoproteins, J. Control. Release 54 (1998) 1527. [145] Y.P. Ho, C.L. Grigsby, F. Zhao, K.W. Leong, Tuning physical properties of
[116] Y.Y. Yang, T.S. Chung, N.P. Ng, Morphology, drug distribution, and in vitro release nanocomplexes through microuidics-assisted connement, Nano Lett. 11
proles of biodegradable polymeric microspheres containing protein fabricated (2011) 21782182.
by double-emulsion solvent extraction/evaporation method, Biomaterials 22 [146] R. Karnik, F. Gu, P. Basto, C. Cannizzaro, L. Dean, W. Kyei-Manu, R. Langer, O.C.
(2001) 231241. Farokhzad, Microuidic platform for controlled synthesis of polymeric nanoparticles,
[117] U. Bilati, E. Allemann, E. Doelker, Sonication parameters for the preparation of Nano Lett. 8 (2008) 29062912.
biodegradable nanocapsules of controlled size by the double emulsion method, [147] N. Kolishetti, S. Dhar, P.M. Valencia, L.Q. Lin, R. Karnik, S.J. Lippard, R. Langer, O.C.
Pharm. Dev. Technol. 8 (2003) 19. Farokhzad, Engineering of self-assembled nanoparticle platform for precisely
[118] J.C. Leroux, E. Allemann, E. Doelker, R. Gurny, New approach for the preparation of controlled combination drug therapy, Proc. Natl. Acad. Sci. U. S. A. 107 (2010)
nanoparticles by an emulsicationdiffusion method, Eur. J. Pharm. Biopharm. 41 1793917944.
(1995) 1418. [148] P.M. Valencia, P.A. Basto, L. Zhang, M. Rhee, R. Langer, O.C. Farokhzad, R. Karnik,
[119] D. Moinard-Checot, Y. Chevalier, S. Briancon, H. Fessi, S. Guinebretiere, Nano- Single-step assembly of homogenous lipid-polymeric and lipid-quantum dot
particles for drug delivery: review of the formulation and process difculties nanoparticles enabled by microuidic rapid mixing, ACS Nano 4 (2010)
illustrated by the emulsion-diffusion process, J. Nanosci. Nanotechnol. 6 (2006) 16711679.
26642681. [149] X. Gong, S. Peng, W. Wen, P. Sheng, W. Li, Design and Fabrication of Magnetically
[120] E. Allmann, R. Gurny, E. Doelker, Preparation of aqueous polymeric nanodispersions Functionalized Core/Shell Microspheres for Smart Drug Delivery, Adv. Funct.
by a reversible salting-out process: inuence of process parameters on particle size, Mater. 18 (2008) 16.
Int. J. Pharm. 87 (1992) 247253. [150] T. He, Q.L., K. Zhang, X. Mu, T. Luo, Y. Wang, G. Luo, A modied microuidic chip
[121] Z. Zhang, D.W. Grijpma, J. Feijen, Poly(trimethylene carbonate) and monomethoxy for fabrication of paclitaxel-loaded poly(L-lactic acid) microspheres, Microuid.
poly(ethylene glycol)-block-poly(trimethylene carbonate) nanoparticles for the Nanouid. 10 (2011) 12891298.
controlled release of dexamethasone, J. Control. Release 111 (2006) 263270. [151] T. Watanabe, T.O., Y. Kimura, Continuous fabrication of monodisperse polylactide
[122] S. Krol, A. Diaspro, O. Cavalleri, D. Cavanna, P. Ballario, B. Grimaldi, P. Filetici, P. microspheres by droplet particle technology using microuidic emulsication and
Ornaghi, A. Gliozzi, Nanocapsule A Novel Tool for Medicine and Science, in: E. emulsionsolvent diffusion, Soft Matter 7 (2011) 98949897.
Buzaneva, P. Scharff (Eds.), Frontiers of multifunctional integrated nanosytems, [152] S. Seiffert, D.A.W., Controlled fabrication of polymer microgels by polymer-
Academic Publishers, the Netherlands, 2004, pp. 439446. analogous gelation in droplet microuidics, Soft Matter 6 (2010) 31843190.
[123] A. Agarwal, Y. Lvov, R. Sawant, V. Torchilin, Stable nanocolloids of poorly soluble [153] W. Chen, Y. Yang, C. Rinadi, D. Zhou, A.Q. Shen, Formation of supramolecular hy-
drugs with high drug content prepared using the combination of sonication and drogel microspheres via microuidics, Lab Chip 9 (2009) 29472951.
layer-by-layer technology, J. Control. Release 128 (2008) 255260. [154] I. Lee, Y.Y., Z. Cheng, H.K. Jeong, Generation of monodisperse mesoporous silica
[124] Z. Dai, A. Heilig, H. Zastrow, E. Donath, H. Mohwald, Novel formulations of vita- microspheres with controllable size and surface morphology in a microuidic
mins and insulin by nanoengineering of polyelectrolyte multilayers around mi- device, Adv. Funct. Mater. 18 (2008) 40144021.
crocrystals, Chemistry 10 (2004) 63696374. [155] S. Huang, B. Lin, J. Qin, Microuidic synthesis of tunable poly-(N-isopro-
[125] R. Palankar, A.G. Skirtach, O. Kreft, M. Bedard, M. Garstka, K. Gould, H. Mohwald, pylacrylamide) microparticles via PEG adjustment, Electrophoresis 32 (2011)
G.B. Sukhorukov, M. Winterhalter, S. Springer, Controlled intracellular release of 33643370.
peptides from microcapsules enhances antigen presentation on MHC class I [156] L.H. Hung, S.Y. Teh, J. Jester, A.P. Lee, PLGA micro/nanosphere synthesis by droplet
molecules, Small 5 (2009) 21682176. microuidic solvent evaporation and extraction approaches, Lab Chip 10 (2010)
[126] M. Aouadi, G.J. Tesz, S.M. Nicoloro, M. Wang, M. Chouinard, E. Soto, G.R. Ostroff, 18201825.
M.P. Czech, Orally delivered siRNA targeting macrophage Map4k4 suppresses [157] Q. Xu, M. Hashimoto, T.T. Dang, T. Hoare, D.S. Kohane, G.M. Whitesides, R. Langer,
systemic inammation, Nature 458 (2009) 11801184. D.G. Anderson, Preparation of monodisperse biodegradable polymer microparticles
[127] A.J. DeMello, Control and detection of chemical reactions in microuidic systems, using a microuidic ow-focusing device for controlled drug delivery, Small 5
Nature 442 (2006) 394402. (2009) 15751581.
120 Y. Zhang et al. / Advanced Drug Delivery Reviews 65 (2013) 104120

[158] L.R. Kesselman, S. Shinwary, P.R. Selvaganapathy, T. Hoare, Synthesis of monodis- [182] D.M. Lavin, R.M. Stefani, L. Zhang, S. Furtado, R.A. Hopkins, E. Mathiowitz,
perse, covalently cross-linked, degradable smart microgels using microuidics, Multifunctional polymeric microbers with prolonged drug delivery and struc-
Small 8 (2012) 10921098. tural support capabilities, Acta Biomater. 8 (2012) 18911900.
[159] M. Marimuthu, S.K., J. An, Amphiphilic triblock copolymer and a microuidic de- [183] S.E. Gratton, P.A. Ropp, P.D. Pohlhaus, J.C. Luft, V.J. Madden, M.E. Napier, J.M.
vice for porous microber fabrication, Soft Matter 6 (2010) 22002207. DeSimone, The effect of particle design on cellular internalization pathways,
[160] K. Hettiarachchi, S. Zhang, S. Feingold, A.P. Lee, P.A. Dayton, Controllable microuidic Proc. Natl. Acad. Sci. U. S. A. 105 (2008) 1161311618.
synthesis of multiphase drug-carrying lipospheres for site-targeted therapy, [184] J.A. Champion, Y.K. Katare, S. Mitragotri, Particle shape: a new design parameter
Biotechnol. Prog. 25 (2009) 938945. for micro- and nanoscale drug delivery carriers, J. Control. Release 121 (2007)
[161] D. Jagadeesan, I. Nasimova, I. Gourevich, S. Starodubtsev, E. Kumacheva, 39.
Microgels for the encapsulation and stimulus-responsive release of molecules [185] T. Nisisako, T. Torii, Microuidic large-scale integration on a chip for mass pro-
with distinct polarities, Macromol. Biosci. 11 (2011) 889896. duction of monodisperse droplets and particles, Lab Chip 8 (2008) 287293.
[162] S.J. Shin, J.Y. Park, J.Y. Lee, H. Park, Y.D. Park, K.B. Lee, C.M. Whang, S.H. Lee, On [186] I. Kobayashi, Y.W., K. Uemura, M. Nakajima, Microchannel emulsication for
the y continuous generation of alginate bers using a microuidic device, mass production of uniform ne droplets: integration of microchannel arrays
Langmuir 23 (2007) 91049108. on a chip, Microuid. Nanouid. 8 (2010) 255262.
[163] J. Su, Y. Zheng, H. Wu, Generation of alginate microbers with a roller-assisted [187] J. Guzowski, P.M. Korczyk, S. Jakiela, P. Garstecki, Automated high-throughput
microuidic system, Lab Chip 9 (2009) 9961001. generation of droplets, Lab Chip 11 (2011) 35933595.
[164] C.H. Choi, H. Yi, S. Hwang, D.A. Weitz, C.S. Lee, Microuidic fabrication of [188] M.P. MacDonald, G.C. Spalding, K. Dholakia, Microuidic sorting in an optical lat-
complex-shaped microbers by liquid template-aided multiphase microow, tice, Nature 426 (2003) 421424.
Lab Chip 11 (2011) 14771483. [189] D. Huh, J.H. Bahng, Y. Ling, H.H. Wei, O.D. Kripfgans, J.B. Fowlkes, J.B. Grotberg, S.
[165] D. Dendukuri, D.C. Pregibon, J. Collins, T.A. Hatton, P.S. Doyle, Continuous-ow Takayama, Gravity-driven microuidic particle sorting device with hydrody-
lithography for high-throughput microparticle synthesis, Nat. Mater. 5 (2006) namic separation amplication, Anal. Chem. 79 (2007) 13691376.
365369. [190] M. Yamada, M. Seki, Hydrodynamic ltration for on-chip particle concentration
[166] B.G. De Geest, J.P. Urbanski, T. Thorsen, J. Demeester, S.C. De Smedt, Synthesis of and classication utilizing microuidics, Lab Chip 5 (2005) 12331239.
monodisperse biodegradable microgels in microuidic devices, Langmuir 21 [191] L.R. Huang, E.C. Cox, R.H. Austin, J.C. Sturm, Continuous particle separation
(2005) 1027510279. through deterministic lateral displacement, Science 304 (2004) 987990.
[167] C.H. Choi, J.H. Jung, D.W. Kim, Y.M. Chung, C.S. Lee, Novel one-pot route to [192] Y. Geng, P. Dalhaimer, S. Cai, R. Tsai, M. Tewari, T. Minko, D.E. Discher, Shape ef-
monodisperse thermosensitive hollow microcapsules in a microuidic system, fects of laments versus spherical particles in ow and drug delivery, Nat.
Lab Chip 8 (2008) 15441551. Nanotechnol. 2 (2007) 249255.
[168] A. Jahn, W.N. Vreeland, M. Gaitan, L.E. Locascio, Controlled vesicle self-assembly [193] T.J. Merkel, K.P. Herlihy, J. Nunes, R.M. Orgel, J.P. Rolland, J.M. DeSimone, Scal-
in microuidic channels with hydrodynamic focusing, J. Am. Chem. Soc. 126 able, shape-specic, topdown fabrication methods for the synthesis of
(2004) 26742675. engineered colloidal particles, Langmuir 26 (2010) 1308613096.
[169] A. Jahn, W.N. Vreeland, D.L. DeVoe, L.E. Locascio, M. Gaitan, Microuidic directed [194] C.C. Ho, A. Keller, J.A. Odell, R.H. Ottewill, Preparation of monodisperse ellipsoi-
formation of liposomes of controlled size, Langmuir 23 (2007) 62896293. dal polystyrene particles, Colloid Polym. Sci. 271 (1993) 469479.
[170] A. Jahn, S.M. Stavis, J.S. Hong, W.N. Vreeland, D.L. DeVoe, M. Gaitan, Microuidic [195] J.A. Champion, Y.K. Katare, S. Mitragotri, Making polymeric micro- and
mixing and the formation of nanoscale lipid vesicles, ACS Nano 4 (2010) nanoparticles of complex shapes, Proc. Natl. Acad. Sci. U. S. A. 104 (2007)
20772087. 1190111904.
[171] T.H. Tran, C.T. Nguyen, D.P. Kim, Y.K. Lee, K.M. Huh, Microuidic approach for [196] J.P. Rolland, B.W. Maynor, L.E. Euliss, A.E. Exner, G.M. Denison, J.M. DeSimone, Di-
highly efcient synthesis of heparin-based bioconjugates for drug delivery, Lab rect fabrication and harvesting of monodisperse, shape-specic nanobiomaterials,
Chip 12 (2012) 589594. J. Am. Chem. Soc. 127 (2005) 1009610100.
[172] J.H. Xu, H.Z., W.J. Lan, G.S. Luo, A novel microuidic approach for monodispersed [197] R.A. Petros, P.A. Ropp, J.M. DeSimone, Reductively labile PRINT particles for the
chitosan microspheres with controllable structures, Adv. Healthc. Mater. 1 delivery of doxorubicin to HeLa cells, J. Am. Chem. Soc. 130 (2008) 50085009.
(2012) 106111. [198] M.C. Parrott, J.C. Luft, J.D. Byrne, J.H. Fain, M.E. Napier, J.M. Desimone, Tunable bi-
[173] J.H. Xu, S.W. Li, C. Tostado, W.J. Lan, G.S. Luo, Preparation of monodispersed chi- functional silyl ether cross-linkers for the design of acid-sensitive biomaterials,
tosan microspheres and in situ encapsulation of BSA in a co-axial microuidic J. Am. Chem. Soc. 132 (2010) 1792817932.
device, Biomed. Microdevices 11 (2009) 243249. [199] L.E. Euliss, J.A. DuPont, S. Gratton, J. DeSimone, Imparting size, shape, and com-
[174] Z. Nie, W. Li, M. Seo, S. Xu, E. Kumacheva, Janus and ternary particles generated position control of materials for nanomedicine, Chem. Soc. Rev. 35 (2006)
by microuidic synthesis: design, synthesis, and self-assembly, J. Am. Chem. Soc. 10951104.
128 (2006) 94089412. [200] J. Nunes, K.P. Herlihy, L. Mair, R. Superne, J.M. DeSimone, Multifunctional shape
[175] D. Ogonczyk, M. Siek, P. Garstecki, Microuidic formulation of pectin microbeads and size specic magneto-polymer composite particles, Nano Lett. 10 (2010)
for encapsulation and controlled release of nanoparticles, Biomicrouidics 5 11131119.
(2011) 13405. [201] D.A. Canelas, K.P. Herlihy, J.M. DeSimone, Topdown particle fabrication: control
[176] H. Xie, Z.G. She, S. Wang, G. Sharma, J.W. Smith, One-step fabrication of poly- of size and shape for diagnostic imaging and drug delivery, Wiley Interdiscip.
meric Janus nanoparticles for drug delivery, Langmuir 28 (2012) 44594463. Rev. Nanomed. Nanobiotechnol. 1 (2009) 391404.
[177] J.S. Hong, S.M. Stavis, S.H. DePaoli Lacerda, L.E. Locascio, S.R. Raghavan, M. Gaitan, [202] L.C. Glangchai, M. Caldorera-Moore, L. Shi, K. Roy, Nanoimprint lithography based
Microuidic directed self-assembly of liposome-hydrogel hybrid nanoparticles, fabrication of shape-specic, enzymatically-triggered smart nanoparticles, J. Control.
Langmuir 26 (2010) 1158111588. Release 125 (2008) 263272.
[178] R.H. Engel, S.J. Riggi, M.J. Fahrenbach, Insulin: intestinal absorption as water- [203] R. Mogul, A.A. Bol'shakov, S.L. Chan, R.M. Stevens, B.N. Khare, M. Meyyappan, J.D.
in-oil-in-water emulsions, Nature 219 (1968) 856857. Trent, Impact of low-temperature plasmas on deinococcusradiodurans and bio-
[179] V. Muguet, M. Seiller, G. Barratt, O. Ozer, J.P. Marty, J.L. Grossiord, Formulation of molecules, Biotechnol. Prog. 19 (2003) 776783.
shear rate sensitive multiple emulsions, J. Control. Release 70 (2001) 3749. [204] P. Panda, S. Ali, E. Lo, B.G. Chung, T.A. Hatton, A. Khademhosseini, P.S. Doyle,
[180] M.T. Gokmen, B.G. De Geest, W.E. Hennink, F.E. Du Prez, Giant hollow multilayer Stop-ow lithography to generate cell-laden microgel particles, Lab Chip 8 (2008)
capsules by microuidic templating, ACS Appl. Mater. Interfaces 1 (2009) 11961202. 10561061.
[181] S. Seiffert, J. Thiele, A.R. Abate, D.A. Weitz, Smart microgel capsules from macro- [205] D. Dendukuri, S.S. Gu, D.C. Pregibon, T.A. Hatton, P.S. Doyle, Stop-ow lithography
molecular precursors, J. Am. Chem. Soc. 132 (2010) 66066609. in a microuidic device, Lab Chip 7 (2007) 818828.

Potrebbero piacerti anche