Silk fibroin nanoparticle as a novel drug delivery system Fatemeh Mottaghitalab, Mehdi Farokhi, Mohammad Ali Shokrgozar, Fatemeh Atyabi, Hossein Hosseinkhani PII: DOI: Reference:

S0168-3659(15)00186-8 doi: 10.1016/j.jconrel.2015.03.020 COREL 7605

To appear in:

Journal of Controlled Release

Received date: Revised date: Accepted date:

11 January 2015 17 March 2015 18 March 2015

Please cite this article as: Fatemeh Mottaghitalab, Mehdi Farokhi, Mohammad Ali Shokrgozar, Fatemeh Atyabi, Hossein Hosseinkhani, Silk fibroin nanoparticle as a novel drug delivery system, Journal of Controlled Release (2015), doi: 10.1016/j.jconrel.2015.03.020

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Silk fibroin nanoparticle as a novel drug delivery system

Nanotechnology Research Center, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran

Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14174, Iran 4

Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei 10607, Taiwan

MA

Center of Excellence in Nanomedicine, National Taiwan University of Science and Technology, Taipei 10607, Taiwan

AC CE P

TE

D

5

NU

3

National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran

SC

2

RI

1

PT

Fatemeh Mottaghitalab1, Mehdi Farokhi2,*, Mohammad Ali Shokrgozar2, Fatemeh Atyabi3, Hossein Hosseinkhani4,5

*Corresponding author: Dr. [email protected]

Mehdi

Farokhi,

1

Tel/Fax:

+982166492595,

E-mail:

ACCEPTED MANUSCRIPT Abstract

SC

RI

PT

The design and synthesis of efficient drug delivery systems are of vital importance for medicine and healthcare. Nanocarrier-based drug delivery systems, in particular nanoparticles, have generated great excitement in the field of drug delivery since they provide new opportunities to overcome the limitations of conventional delivery methods with regards to the drugs. Silk fibroin (SF) is a naturally occurring protein polymer with several unique properties that make it a suitable material for incorporation into a variety of drug delivery vehicles capable of delivering a range of therapeutic agents. SF matrices have been shown to successfully deliver anticancer drugs, small molecules, and biomolecules. This review will provide an in-depth discussion of the development of SF nanoparticle-based drug delivery systems.

AC CE P

TE

D

MA

NU

Keywords: Drug delivery, Silk fibroin, Nanoparticles.

2

ACCEPTED MANUSCRIPT Contents 1. Introduction

PT

2. Drug delivery systems 2.1 Drug delivery systems based on nanotechnology vs. conventional carriers

RI

2.2 Important parameters for nanoparticle-based drug delivery

SC

2.3 Polymeric nanosystems for drug delivery 3. Characteristics of silk protein

NU

3.1 Silkworms 3.2 Molecular properties of silk protein

3.4 Biodegradation rate of silk protein

MA

3.3 Crystallinity of silk protein

D

3.5 Particulate silk protein preparation techniques

TE

3.6 Potential characteristics of silk fibroin for drug delivery applications 3.7 Silk fibroin nanoparticles vs. other silk fibroin based carriers

AC CE P

4. Silk protein as a drug carrier

4.1 Silk fibroin nanoparticulate for protein delivery 4.2 Silk fibroin nanoparticulate for small molecules delivery 4.3 Silk fibroin nanoparticulate for anticancer delivery 5. Conclusions References

3

ACCEPTED MANUSCRIPT 1. Introduction

AC CE P

TE

D

MA

NU

SC

RI

PT

In recent years, in order to optimize the efficacy of therapeutics, many drug delivery systems have been designed to administer multiple drugs and release them in a controlled manner [1-4]. The use of these systems offers many advantages such as enhancing the bioavailability of drugs by reducing their degradation rate, improving cellular uptake, allowing targeting and control of drug release, and reducing side effects [5]. To date, both synthetic and natural polymers have been used for drug delivery applications. Among wide range of applied synthetic polymers, polyesters, polyorthoesters, polyanhydrides, polyphosphazenes, and polyphosphoesters have found extensive application [6-8]. However, despite the wide range of available materials, the majority of licensed drug delivery systems are based on the FDA (U.S. Food and Drug Administration)- approved polymer, poly(lactic-co-glycolic acid) (PLGA), because of properties such as suitable pharmacokinetics and controllable degradation rate [9, 10]. However, the usefulness of PLGA is limited in applications such as protein therapeutics due to some of its intrinsic properties and processing requirements [11-14]. Therefore, natural polymers (e.g., alginates, chitosan, collagen, dextran, pullulan and gelatin) represent an attractive alternative with higher biocompatibility and biodegradability than PLGA [15-24]. In addition to their composition, the structure of drug delivery systems also needs serious consideration. To date, many systems have been designed with different morphologies and structures, including films, gels, foams, microparticles, and nanoparticles [25]. In the 1960s, liposomal carriers were the first nano-systems to be approved for the delivery of proteins and drugs [26]. Additionally, many studies have reported the high capacity of nanoparticles for therapeutic molecules [27-29]. In most cases, the use of particulate carriers reduces the rate of delivery of solubilized drugs by introducing a second limiting step [30, 31]. Furthermore, nanoparticles have many features, which are useful for drug delivery such as a high surface to volume ratio [32], an ability to act as modifiable platforms [33], and a tunable size [34]. Therefore, applying the principals of nanotechnology to the design of drug delivery systems will not only improve their therapeutic efficacy but could also preserve the properties of bioactive molecules [35]. It is necessary to consider the properties of biomaterials in terms of composition, structure, mechanical properties, and function to fabricate particulate drug delivery carriers. Silk proteins are FDA-approved polymers that have been used successfully as both sutures and drug delivery systems. These proteins have excellent mechanical properties, a flexible preparation process, and high biocompatibility [25]. So far, many review papers have been published concerning the use of silk proteins in the field of tissue engineering. While there are many original articles about the application of silk nanoparticles as drug delivery vehicles, to the best of our knowledge, no comprehensive review on their use for drug delivery has yet been published. Therefore, in this review, we provide an extensive overview on recent efforts in constructing silk protein nanostructures for drug delivery. Firstly, we introduce the different applications of nanotechnology-based systems. Secondly, the properties of silk proteins are discussed clearly, and finally, nanoparticulate silk proteins are considered in detail. 2. Drug delivery systems 2.1 Drug delivery systems based on nanotechnology vs. conventional carriers Historically, drug delivery systems were usually based on orally administered or injectable drugs. However, these have been found to be inappropriate for novel therapeutics such as 4

ACCEPTED MANUSCRIPT

TE

D

MA

NU

SC

RI

PT

proteins and nucleic acids. Novel technologies are required for delivery of new drug molecules in order to reduce their side effects, optimize their efficacy, and enhance patient compliance. Recently, the use of nanotechnology has led to the development of many novel carriers capable of controlled release and targeted delivery of a wide range of small molecules, proteins, peptides, and genes [36-41]. These devices can have many different structures, including liposomes, micelles, quantum dots, dendrimers, fullerenes, ferritin, and nanoparticles [42-45]. Among them, nanoparticles based on biodegradable and biocompatible polymers have potential applications in cancer therapy and as sustained drug delivery vehicles. These carriers can also be designed as low toxicity systems with suitable physical and chemical structures and specific targeting properties [46]. It was reported that particle size is the most important factor when designing drug delivery systems. Therefore, it is crucial to use nanoparticles for the delivery and targeting therapeutic molecules [47, 48]. These systems also have other advantages, including prolonged drug half-life, improved solubility of hydrophobic drugs, reduced immunogenicity, and reduced administration frequency [49]. As mentioned earlier, the possibility of targeted drug delivery is one of the main advantages of nanoparticles. It is strongly believed that the conjugation of different ligands to nanoparticles could improve the targeting efficacy of them as compared to conventional therapeutics [50]. The small size of nanoparticles also affects the targeting efficacy. Generally, nanosized particles experience efficient uptake, selective drug accumulation in the targeted site, and are able to penetrate into the endothelium at inflammatory sites, epithelium (e.g., intestinal tract and liver), tumors, or microcapillaries [51, 52]. The ability to co-deliver multiple drugs is another advantage of nano-based drug delivery systems in comparison to conventional systems [53]. Co-delivery of drugs offers several benefits such as the possibility of synergistic effects [54], suppressed drug resistance [55] and the ability to adjust the dosage of drugs to the level of a single nanoparticle carrier.

AC CE P

2.2 Important parameters for nanoparticle-based drug delivery Understanding the interactions between nanomaterials and cells/lipid bilayers is important in the fields of phototherapy, imaging, and drug/gene delivery. The reason for this is the small size of nanoparticles as compared to microparticles. In order to address this issue, Desai et al. have shown that the cellular uptake of 100-nm nanoparticles was 2.5 and six times higher than 1-µm and 10-µm microparticles, respectively [51]. It was also reported in a similar study that the cellular uptake of 100-nm nanoparticles was 15–250 times higher than 1- and 10-µm microparticles [56]. Chithrani et al. have claimed that efficient uptake of nanoparticles depends on their size. To this end, they have shown that 50-nm gold particles undergo the most effective uptake [57]. They have also stated that spherical nanoparticles experience five times greater uptake than rod-shaped particles. Therefore, it appears that size and shape are the two important factors that affect the cellular uptake of particles [57]. It is also known that, in addition to influencing the cellular uptake, particle size can also influence drug loading, drug release, and the stability of nanoparticles [58]. Along with size and shape, the nanoparticle’s surface properties are also important. Surface characteristics such as hydrophobicity and hydrophilicity determine the level of absorbance of blood components such as opsonins [59, 60]. However, it has been shown in in vitro studies that there is a connection between the extent of opsonization and the surface charge of nanoparticles and that less opsonization occurs in neutrally charged particles in comparison to charged particles [61]. For this reason, the use of shielding groups that is capable of blocking the electrostatic and hydrophobic interactions result in the binding of

5

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

opsonin to the surface of nanoparticles. A brief description of the effects of particle size and the surface properties of nanoparticles is summarized in diagram 1.

Diagram 1. Properties that define the behavior of silk nanoparticles (SFNPs).

2.3 Polymeric nanosystems for drug delivery Polymeric nanoparticles have been of great interest to many researchers for decades. Since the discovery of dendrimeric ―starburst‖ polymers in the 1980s, numerous other drug delivery strategies such as self-assembled micelles and encapsulated drug molecules have been developed [62, 63]. Biodegradable polymeric nanocarriers are predominantly used for their ability to control the release rate of drugs, and for this purpose, many synthetic and natural polymers have been used [36]. Synthetic polymers have greater structural integrity and higher purity, which makes the preparation of nanoparticles more reproducible, than natural polymers [36]. However, only synthetic polymers with an appropriate biodegradability and low cytotoxicity are suitable for drug delivery applications [64]. Additionally, the ability of synthetic polymers to control the release rate of drugs is much higher than natural polymers. Synthetic polymers are capable of controlling the release rate of drugs over a longer duration than natural polymers, which have relatively short durations of release. However, the harsher reaction conditions and use of organic solvents, which are necessary to prepare synthetic polymers, have limited their application. Poly(lactic acid) (PLA), poly(glycolic acid) (PGA), PLGA, and poly(alkylcyano acrylates) (PCA) are the most promising synthetic polymers for the development of drug delivery platforms for clinical applications [65] which are listed in Table 1. 6

ACCEPTED MANUSCRIPT Table 1. Potential synthetic nanoparticles for drug delivery applications. Processing method

Loaded Molecule

size (nm)

Key findings

Ref.

PLA1

Emulsificationsolvent evaporation

Aureusidine

231-376

Improving aureusidin's water solubility and light sensitivity

[66]

Effective cytotoxic T-lymphocyte responses and tumor antigen-specific cytotoxicity

[67]

Significant uptake of nanoparticles in the estrogen receptor positive MCF-7 cell line

[68]

[69]

150- 200

Highly negative zeta potential and good biocompatibility of nanoparticles

675

Prolonged BSA release to 30 h

[70]

Enhanced encapsulation efficacy, high uptake and retention by macrophages, increased drug efficacy against M. tuberculosis residing in macrophages

[71]

Involvement of different pathways depending on the surface properties of nanoparticle in the interaction between nanoparticles and the blood–brain barrier

[72]

Long-circulating properties of nanoparticles, reduced liver accumulation

[73]

86

RI

Double emulsion

PBCA7

PBCA8

Poly (hexadecylcy anoacrylate)

MA

250 – 300

Precipitation and dialysis method

Ovalbumin

Water-in-oil emulsion technology plus cyclic freezingthawing process

D

PVA5

Tamoxifen

BSA6

TE

g-PGA4

Solvent displacement

AC CE P

PCL3

NU

SC

PLGA2

Anti-OX40 monoclonal antibody

Anionic polymerisation

Moxifloxacin

Anionic polymerisation

Doxorubicin

Nanoprecipitation/

-

418

202-246

150

solvent diffusion

PT

Material

1

Poly(lactic acid), 2Poly(DL-lactide-co-glycolide), 3Poly(ε-caprolactone), 4Poly(g-glutamicacid), 5Poly(vinyl alcohol),6Bovine serum albumin, 7Poly(butyl cyanoacrylate), 8Poly(butyl cyanoacrylate)

In contrast, natural polymers have variable purity, lower stability, and require further modification and crosslinking prior to use [74]. Many studies have been fabricated nanoparticles based on natural polymer which are listed in Table 2.

7

ACCEPTED MANUSCRIPT Table 2. Potential natural nanoparticles for drug delivery applications. Processing method

Loaded molecule

Size (nm)

HSA1

Desolvation

Cetuximab

200-250

BSA2

Emulsificationdispersion

Tacrolimus

189

Collagen

Alkaline hydrolysis

17β-estradiolhemihydrate

Gelatin

Desolvation

Paclitaxel

Protamine

Self-assembly

CpGoligodeoxynucle otide

Gliadin

Desolvation

Targeting colon carcinoma cells

RI

Proteins

Key findings

PT

Material

Ref.

[75]

[76]

Prolonged estradiol release and absorption

[77]

600-1000

Rapid drug release, targeting human bladder cancer cells

[78]

215

Suppress T-helper 2 immune response, suitable for immunotherapy of allergy

[79]

Al-trans-retinoic acid

500

Good drug loading efficiency, biphasic releasing pattern

[80]

Coacervation

Folic acid

150

Folic acid encapsulation, release prevention in an acid environment, enhanced oral bioavailability

[81]

pH-coacervation

Methylene blue

250

Increased release with decrease in glutaraldehyde/lysine ratio

[82]

Thermoresponsive self-assembly

BMP3-2 and BMP-14

237

High encapsulation efficacy of BMPs with sustained release pattern, maintenance of growth factors activity

[83]

Chitosan

Ionic gelation

Cyclosporin A

293

High corneal-conjunctival intact, enhanced external ocular delivery compared to inner ocular

[84]

Alginate

Water-in-oil microemulsion

GFP-encoding plasmids

55-100

Suitable for gene therapy

[85]

SC

Enhanced accumulation in blood rather than other organs e.g. kidney

Casein

Legumin

Elastin

AC CE P

TE

D

MA

NU

123

Polysaccharides

8

ACCEPTED MANUSCRIPT Ionotropic gelation

Methotrexate

390

Sustained drug release, enhanced drug delivery

[86]

Starch

Reactive extrusion

-

160-300

160 nm starch particles preparation by appropriate crosslinkers addition

[87]

Dextran

Nanoprecipitation

lidocaine

86–256

High drug encapsulation efficacy

[88]

Pullulan

Dialysis

Adriamycin

156

High drug loading, pHsensitive in vitro drug release, enhanced cellular uptake

[89]

Hyaluronic acid

Self-assembly

-

Active targeting by strong receptor-binding affinity of nanoparticles to CD44

[90]

237–424

MA

1

NU

SC

RI

PT

Pectin

Human serum albumin, 2 Bovine serum albumin, 3Bone morphogenetic protein

D

3. Characteristics of silk protein

AC CE P

TE

Successful polymer-based delivery systems need to be biocompatible, biodegradable, have low toxicity, appropriate mechanical properties, require ambient processing conditions, and provide sustained release. Silk is a natural polymeric biomaterial that can address these requirements because of its unique structural properties, self-assembling ability, mechanical strength, processing flexibility, biodegradability, and biocompatibility [91]. 3.1 Silkworms

Wide varieties of silkworms produce natural silk worldwide. In general, the silk proteins produced by different silkworms differ in their structure and properties, and not all of the species are commercially viable. For instance, the Bombycoidea family contains eight subspecies, and only two members of this family – Bombycidae (Mulberry) and Saturniidae (non-mulberry) – are commercially important [92]. Bombyx mori is the commercial source of mulberry silk that is produced by the Bombycidae family. Mulberry silkworms are entirely domesticated and need human care for their growth and reproduction, which does not occur naturally [92]. The nonmulberry/mulberry silkworm classification originates from the feeding habits of silk producing insects, which belong to the Saturniidae and Lasiocampidae families [93]. Non-mulberry silkworms are comprised of the following species: the tropical (Antheraea mylitta) and temperate (A. pernyi, A. roylei, A. proylei, and A. frithi) tasar silkworms, eri silkworms (Philosamia ricini/Samia ricini), muga silkworms (A. assamensis), fagaria silkworms (Attacus atlas), and shashe silkworms (Gonometa postica) [94]. These species live principally in the wild and have been found in polymorphic forms in a variety of host plants in different geographical regions. For this reason, the silk produced by the species listed above varies in luster, color, and tensile strength [95]. 3.2 Molecular properties of silk protein

9

ACCEPTED MANUSCRIPT

NU

SC

RI

PT

The silk fibroin (SF) fibers from B mori fibers have a diameter of about 10-25 mm with three structural protein subunits, including a light chain (~26 kDa), a heavy chain (~390 kDa), and a small glycoprotein named P25 (of ca. 30 kDa). A disulfide bond links the light and heavy chains together, and P25 is attached to the fiber via non-covalent hydrophobic bond [95-97]. The heavy chain of SF has an amphiphilic nature and contains both hydrophobic and hydrophilic blocks. The hydrophobic blocks have a repeating sequence of Gly-Ala-Gly-Ala-Gly-Ser, which is responsible for generating the crystalline structure of SF by folding into β-sheets. However, the hydrophilic region is a short and non-repetitive segment in comparison to the hydrophobic region [96, 98]. Fibroin could be therefore be considered a hydrophobic glycoprotein; hence, it is insoluble in water [99]. Another component of B. mori SF fibers is sericin (20 kDa to 310 kDa). Sericin contains two subunits α-sericin and β-sericin, which are found in the external and inner layers of the cocoon, respectively. Sericin [98] can be isolated as a hydrophilic protein from fibroin via a thermochemical procedure called degumming [100]. Sericin has an amorphous and glue-like structure that is responsible for binding two fibroin fibers together, and thus it maintains the structural integrity of the cocoons [101].

MA

3.3 Crystallinity of silk protein

AC CE P

TE

D

As mentioned previously, SF is a semi-crystalline biopolymer and consists of crystalline and amorphous regions. The crystalline part has two prominent structures, i.e., silk I and silk II [102]. Unstable and water-soluble state silk I is obtained from spinning dope [103, 104]. Silk I can be made more stable after transforming it to silk II during the spinning process. The crystalline structure of B. mori fibers consists principally of silk II structure as a result of its β-sheet conformation, while the amorphous part of fibroin is in random coil conformation [105-107]. It should be mentioned that the β-sheet has an asymmetrical structure featuring hydrogen side chains from glycine on one side and methyl side chains from alanine on the other, creating hydrophobic domains. Strong hydrogen bonds and van der Waals forces between the methyl and hydrogen groups on opposing sides allow inter-sheet stacking in the crystals and thus, generates a thermodynamically stable structure [96]. Most of the characteristic physical and chemical properties of SF such as high strength and resistance to chemicals and micro-organisms and low elasticity and extensibility are derived from its crystalline structure [108]. There are many methods to enrich SF with β-sheet structure, and they are listed in Table 3.

10

ACCEPTED MANUSCRIPT Table 3. Methods capable of enriching SF in β-sheet structure.

50% (v/v) methanol solution

Silk concentration

Treatment time

20–25% (w/v)

β-sheet induction

PT

Casting

Treatment

2 days

Yes

Laminar jet break-up

Water vapor

3% and 9%

Film

Casting

High temperatures

1–2%

24 hour

Yes

Not reported

Yes

Capillary shape solidified sample

Membrane

Freeze drying

Freezing temperature

AC CE P

Particle

TE

D

MA

Sphere

NU

SC

Film

Method

RI

Structure

Solidification

Casting

Shear force

Ion concentration

2% (v/v)

24 hour

39 wt.%

Not reported

3% (w/v)

Not reported

11

To somewhat

Yes

Yes

FTIR or Raman finding Amide I shift to 1665 cm1, appearance of new maxima at 1262, 1236 cm1 (amide III), and 1084 cm1 Shifts of 1658 cm-1 (amide I) and at 1540 cm-1 (amide II) bands to 1629 cm-1 (amide I) and 1517 cm-1 (amide II) Not reported

Ref.

[109]

[110]

[111]

Appearance of absorption band at 1265 cm-1 (amide III) by increasing the freezing temperature -60 to -10°C

[112]

Appearance of three major characteristic bands of silk II at 1085, 1232 and 1667 cm−1

[113]

Shift of amide I band from 1657 to 1667 cm-1 after increasing the ion concentration

[114]

ACCEPTED MANUSCRIPT 3.4 Biodegradation of silk protein

AC CE P

TE

D

MA

NU

SC

RI

PT

In order to develop a successful formulation of nanoparticle based drug carriers, it is necessary to consider their biodegradation rate. Many factors affect the release rate of nanoparticles, including desorption of surface-bond/adsorbed drug molecules, diffusion through the nanoparticle matrix, diffusion through the polymer wall (in the case of nanocapsules), erosion of nanoparticle matrix, and a combined erosion/diffusion process [115]. Generally, in contrast to natural polymers, synthetic polymers such as PLA, PGA, and PLGA produce acidic compounds as by-products of hydrolytic degradation, and this is potentially undesirable at the targeted sites [116]. The sensitivity of natural polymers such as collagen, fibrinogen, hyaluronic acid to enzymatic degradation is sometimes higher than the synthetic polymers [117]. As a natural polymer, SF usually undergoes proteolytic degradation resulting in non-toxic by-products [26]. However, US Pharmacopeia has classified SF as non-degradable material because it maintains 50% of its tensile integrity 60 days post-implantation [95]. However, we have recently reported that immersing freeze-dried SF scaffold in phosphate buffered saline (PBS) solution can induce hydrolysis degradation. In our study, the structural, physical, and mechanical properties of the SF scaffold were different 12 weeks after incubation in PBS (Figure 1). Surprisingly, hydrolysisinduced changes to the surface of the scaffold affected the behavior of osteoblast cells in terms of biocompatibility, alkaline phosphatase production, and even the expression of some bone gene markers [118].

Figure 1. Scanning electron micrographs of: (a) Fresh SF scaffold, (b) Treated SF scaffold in PBS after 12 weeks. The interconnection between pores is treated and larger pores with unusual structure are formed as a function of exposure time. Wenk et al. have also reported that in the absence of proteolytic degradation, only 4% of SF scaffold weight is degraded within 7 weeks by hydrolysis [119]. In contrast to synthetic polymers like PLGA, SF usually undergoes surface erosion and degradation by-products diffuse into the active site. This phenomenon results in a reduction of drug stability, drug activity, and safety [120]. Moreover, Wang et al. have reported an SF porous scaffold that is not only biodegradable but also bioresorbable due to degradation by macrophages [121]. In general, the possibility of enzymatic, surface-mediated biodegradation and controlled biodegradation of SF could be considered an important factor in its role as a drug delivery agent. An overview of various enzymatic methods used for SF degradation is shown in Table 4. Generally, Biodegradable nanoparticles as well as silk polymer have shown great interest for imaging, cancer treatment, medical tools, bone treatment, drug delivery, diagnostic tests, and drug

12

ACCEPTED MANUSCRIPT development [122-124]. These particles have the potential to enable early detection, prevention, and to essentially improve diagnosis, treatment and follow-up of diseases [125-129]. Table 4. An overview of various enzymatic methods used for SF degradation

Recombinant honeybee silk protein (AmelF3)

Nanofiber

In vitro

Microtube

Intervals (1, 3, 8, 24, 48 or 96 h)

Bright field microscopy, SDS-PAGE spectra

Protease XIV (2 U/ml)

56 days

NU

In vitro

Technical characterization

In vitro

Protease XIV (5.3 units/mg)

Mass loss evaluation, SEM, histology, cell metabolism study

10 days

Mass loss evaluation

AC CE P

TE

D

Bombyx mori

3D porous scaffold

Incubation time

MA

Bombyx mori

Enzyme model Alphachymotrypsin (54 U mg–1) and trypsin (313 U mg–1)

PT

In vivo/ In vitro

RI

Silk structure

SC

Type of silk

Key findings

Ref.

Cleavage of protein by both proteases

[130]

High degradation rate of scaffold affected osteogenesis and cell metabolism Production of more brittle silk microtubes, after incubation and loss of mechanical integrity Higher degradation rate during 9 days, slower degradation until 18 days, morphology changes after in vivo degradation

[131]

[132]

[133]

Conduit

In vitro and in vivo

Protease XIV (1.0 U/ml)

10 days for in vitro and 24 weeks for in vivo

UV spectroscopy, gross observation, SEM analysis, mass loss evaluation, SDSPAGE spectra, histology

Bombyx mori

Porous SF sheets

In vitro

Alphachymotrypsin collagenase IA and protease XIV (1.0 U/ml)

1, 3, 6, 9, 12, and 15 days

Mass loss evaluation, XRD, SEM, Molecularweight distribution

Degradation of sheets by collagenase IA and protease XIV in contrast to Alphachymotrypsin

[134]

Bombyx mori

SF mats

In vitro and in vivo

Protease XIV (1 U/mL)

Within 24 days for in vitro and 8-weeks

Mass loss evaluation, SEM, FTIR, XRD, histology

Degradation of 65% of the electrospun SF scaffolds within 24 day

[135]

Bombyx mori

13

ACCEPTED MANUSCRIPT

In vivo

-

Within one-year

NU MA

Histology, Histochemistry

RI

Porous SF scaffold

SC

Bombyx mori

PT

for in vivo

in protease XIV, complete degradation after 8 weeks in vivo Complete degradation of prepared scaffolds in aqueous medium between 2 and 6 months, beyond 1 year degradation rate for scaffolds prepared in organic solvents

[121]

D

3.5 Particulate silk protein preparation techniques

AC CE P

TE

In recent years, many micro and nanoparticulate systems have been used for biomedical and pharmaceutical applications. These systems are suitable as drug carriers as they have the ability to control the release rate of drugs and increase the likelihood of positive therapeutic outcomes [136]. When designing particles for drug delivery applications, it is important to consider their biocompatibility, biodegradability, size, drug loading and release [137]. However, selecting the appropriate types of biomaterials and finding the correct processing methods needed to prepare particles is challenging. For instance, it is necessary to avoid organic solvents, surfactants, initiators, or cross-linking agents during particle preparation [138-140]. Silk as a flexible polymer is commonly prepared in a powder form by using a chemical process; this is known as the bottom-up approach [141]. During preparation via this method, the intermolecular forces between β-sheets within the silk proteins are broken down by dissolving in different solvents (e.g. chaotropic salts, ionic liquids, and fluorinated solvents) leading to the generation of silk particles [142-149]. Consequently, in order to make the silk particles water insoluble, the particles are usually treated with kosmotropic salts like potassium phosphate or alcohols (usually methanol) to induce the formation of β-sheets [150]. Another method to prepare silk particles is the top-down approach. For this method, silk fibers are cut with different milling machines to mechanically prepare fine silk particles in the powder form [151]. The key drawback of using chemical procedures is the denaturation of the silk protein structure. In addition, the removal of chemical agents from silk particles is a lengthy process [151]. This drawback makes the use of mechanical methods more appealing as it avoids the limitations inherent in the chemical approaches and allows the direct preparation of silk particles. However, this method is limited by the viscoelastic nature of silk proteins, which need a milling time of up to 40 h to produce fine particles [152]. A brief description of some important research activities concerning silk powder preparation are summarized in Table 5.

14

ACCEPTED MANUSCRIPT Table 5. A brief description of important research activities concerning silk powder preparation. Loaded molecule

Key findings

Ref.

[146]

Formic acid

< 80 nm

Spherical nanopowder

FTIR1, XRD2, SEM3

200 nm

Spherical powder

Laser particle analyzer and SEM

500 nm2 µm

Spherical powder

Potassium phosphate

Rotary and planetary ball milling

Water-in-oil emulsificationdiffusion

Self-assembly

TE

Ethanol and PVA

980 nm

AC CE P

Self-assembly

Decreasing the average size of SF particle

-

Narrow size distribution of SF particles at pH 10

Alcian blue, rhodamine B, and crystal violet

Negative surface charge, higher loading capacity of charged small molecules with controlled release

[153]

UV-Vis spectrometry, FTIR, SEM, light microscopy, DLS4

Rhodamine B, dextran and BSA

Appropriate drug loading capacity, controlled release of hydrophobic drugs, higher uptake efficiency

[154]

UV–Vis spectrometry, SEM, light microscopy, particle size analysis

-

RI

Potassium hydrogen phthalate and hydrochloric acid

PT

Characteristic method

D

Phase separation

Particle type

SC

Bead milling

Particle size

NU

Electro-spraying

Chemical solution

MA

Method

Spherical powder

[152]

-

200 nm

Fibrous particle

Laser particle analyzer and SEM

-

Ethyl acetate, Diethyl ether, DCM5, Chloroform

48- 148 µm

Bowl-like and spherical

FTIR and SEM

-

Ethanol

0.2 to 1.5 µm

[151]

Spherical

Photoncorrelation spectroscopy, particle size analysis, SEM, AFM6, FTIR

Water-in-oil 15

-

Fine particles production

Relation of shape and size of SF microparticles to the type of organic phase

[155]

Particle size and size distribution dependence on SF microspheres to the amount of ethanol, SF concentration, freezing, and temperature

[156]

High BSA absorption

[157]

ACCEPTED MANUSCRIPT

Spherical

µm

BSA8

FTIR, SEM, Particle size analysis

-

101-440 μm

Spherical

FTIR, SEM, Particle size analysis

Phase separation

PVA9

300 nm20 μm

Spherical

FTIR, SEM, DSC10, DLS

BSA, rhodamine B

efficiency, lower size of SF microsphere Bioactivity preservation of growth factor, sustained release over 7 weeks Controlling the size and distribution of SF sphere by varying the concentrations of SF and PVA

[110]

[158]

NU

SC

Laminar jet break-up

Salicylic acid, propranolol hydrochloride , IGF-I7

PT

Paraffin

RI

emulsion solvent evaporation

Fourier transform infrared spectroscopy, 2X-ray diffraction, 3Scanning electron microscopy, 4Dynamic light scattering, 5Dichloromethane, 6Atomic force microscopy, 7Insulin-like growth factor, 8Bovine serum albumin, 9 Polyvinyl alcohol, 10Differential scanning calorimetry.

MA

1

3.6 Potential characteristics of silk fibroin for drug delivery applications

AC CE P

TE

D

SF has many unique properties that established its reputation among other synthetic and natural polymers for controlled drug delivery. For this, the investigation of SF as a drug carrier has widely expanded over the last few years due to highly controllable composition and sequence, structure and architecture, mechanical properties and function. One of the main advantages of using SF as a carrier is performing mild all-aqueous processes for loading sensitive drugs such as protein and nucleic acid therapeutics in order to provide good resistance to dissolution, thermal and enzymatic degradation [159, 160]. This can be achieved by conformational transition of αhelix and random coil to highly crystalline β-sheets through water vapor annealing, mechanical stretching and ultrasonic treatments. This avoids the use of any harsh processing conditions which make silk as a potential system for drug delivery applications [161]. Additionally, SF protein consists of a diverse range of amino acids with functional groups including amines, alcohols, phenols, carboxyl groups, and thiols that simplify the attachment of different biomolecules or antibodies for specific cell types. It is suggested that different drugs with different kinetics could be introduced to SF biomaterial by varying the degree of functionalization per SF molecule that could provide a variety of drug release systems [25]. This is the significant advantage of using SF compared with many other relatively inert polymeric systems [25, 162]. Moreover, it is possible to modify the properties of SF by genetic manipulation. These novel structures are consisting of silk sequence that self-assemble into the desired morphological structures and the sequence of a polypeptide that endow novel functionalities. The functional domains can provide binding sites for receptors, enzymes, drugs, metals or sugars, among others [163]. Another main feature of silk among other polymers is its potential as a lysosomotropic drug delivery platform. A wide range of synthetic and natural polymers have been used as lysosomotropic delivery systems. However, silk as a natural polymer has an intrinsic capability to response to pH changes in order to initiate drug release. Therefore, drug release is achieved in response to pH without any chemical modifications [164]. When using biomaterials for drug delivery applications, it is necessary to consider their clearance mechanism from the body. Absorption is the main mechanism for clearing of some biomaterials. Additionally, by-products of biomaterials could be cleared by liver, kidneys or lungs [165]. As 16

ACCEPTED MANUSCRIPT

SC

3.7 Silk fibroin nanoparticles vs. other SF based carriers

RI

PT

silk can be degraded by such proteolytic enzymes, absorption is the most likely mechanism of elimination from the body without any side effects. The binding mechanism of biomolecules to silk is another important feature for controlling drug release kinetics. It is assumed that electrostatic interaction is the main possible mechanism for release and loading drug on SF biomaterial [153]. The negatively charged silk particles (−24 to −26 mV) provide opportunities for strong electrostatic force with hydrophobic and positively charged drugs compared with hydrophilic or negatively charged drugs. These strong interactions electrostatic could avoid the significant burst release as is normally seen with many polymeric carrier particles [158, 166].

AC CE P

TE

D

MA

NU

To date, different structures of SF including micro and nanospheres, hydrogels, micro- and nanoparticles have been explored for drug delivery applications. Generally, SF microspheres with mucoadhesive properties are more relevant for long-acting delivery of depot drug. They are able to adhere to the mucous membrane of oral and nasal routes and release encapsulated drug [167]. The common drug release mechanism of microspheres is diffusion of drug molecules from degraded polymeric matrix. Silk microspheres can be processed using different methods but some of these approaches (e.g. spray-drying method) apply harsh processing conditions and high temperature. In addition, the microspheres generated by this mode of processing are large, above 100 μm, which is suboptimal for drug delivery [148]. Another useful structure of SF for drug delivery applications is SF nanosphere that is usually used as short-acting delivery carriers. The other advantage of using SF nanospheres as a drug carrier is that they could be used either by fluidizing with a liquid carrier or as a solid powder [168, 169]. Drug release mechanism from nanosphers is identical to microspheres as described above. In some cases, the small size of nanospheres is not suitable for targeted drug delivery such as pulmonary drug delivery. Therefore, these spheres are incorporated to larger spheres using flocculation, spray drying and etc. to have a desired size in order target a specific disease site [170]. Rather than size limitation of nanospheres for drug delivery applications, the shape of this structure has also a significant impact on degradation of polymeric matrix and drug release [171]. Therefore, it seems that fabricating SF nanopsheres is more challenging for drug delivery. The main challenges of fabricating SF nanospheres are high molecular weight and protein nature of SF. Moreover, when expose to heat, salt, pH change and high shear, the SF tends to self-assemble into fibers or gels [158, 172]. Moreover, in both nanosphere and microsphere structures, uncontrollable size and shape and using harsh preparation conditions such as organic solvent are challenging for using these structures in drug delivery applications. Therefore, researchers have developed a new SF based structure applicable as drug carriers. For example, hydrogels are attracted more attention in the field of drug delivery. Generally, many factors including hydrogel hydration, crosslinking, pore size, degradability, hydrophobicity, charge and polymer concentration affect the release rate of different drug molecules from hydrogel. It is confirmed that the β-sheet content is increased after SF gelation. So, there is no need to post-treat the SF hydrogels with solvents in order to induce water insolubility. SF hydrogels are simply prepared by lowering the pH of a SF solution (with pI of about 4.2) in the presence of the drug with acidic solution to pH 4 in order to induce gelation. Although, this pH may be suitable for some drugs, but this may have harmful effects on others. Therefore, it is considered that despite of favorable characteristics of SF hydrogel for drug delivery, applying this structure is related to the type of incorporated drug. Moreover, the low mechanical properties are another limitation of using hydrogels in biomedical applications [173-176]. Microparticulate SF carriers have also considerable potential to be used as 17

ACCEPTED MANUSCRIPT

MA

NU

SC

RI

PT

therapeutics delivery platform [25, 177]. Using SF microparticles have many advantages in comparison to other mentioned structures including preservation of drug from degradation and denaturation, controlling the release rate of drugs and the high potential for targeted drug delivery [153]. Despite of many potential properties of using microparticles as drug carriers, the harsh preparation processes including non-aqueous solvents, water/solvent interfaces, crosslinking reagents for hardening, or high temperatures have limited their applications [178, 179]. In this regards, it is necessary to optimize formulation protocols in order to preserve the stability and potency of drugs. Despite of potential advantages of above SF based delivery systems, their limitation tend to investigate SF nanoparticles based delivery systems. As mentioned above, the application of SF nanoparticles has expanded due to advantages such as biocompatibility, controlled degradation, size and shape as well as drug loading and release. The small size of SF nanoparticles allows them to penetrate through small capillaries which enhance the cellular uptake of an encapsulated drug or therapeutic molecule [165]. Moreover, SF nanoparticles have high potential for targeted drug delivery as they could get into cells and deliver anticancer to the tumor site. Highly efficient clearance systems are also applied for unused nanoparticles or their degraded products from the body. Therefore, SF nanoparticles are suggested as potential delivery platform in comparison to other conventional systems [165, 166]. 4. Silk protein as a drug carrier

D

4.1 Silk fibroin nanoparticulate for protein delivery

AC CE P

TE

In the context of protein-based drug delivery systems, silk is a potentially useful natural polymer, which has been used to deliver peptide and protein molecules (Table 6). Unfortunately, few studies have discussed the role of silk nanostructure in protein delivery systems. Hofer et al. investigated the use of recombinant spider silk protein eADF4 (C16) particles for carrying the high molecular weight protein, lysozyme. With an efficiency of almost 100% lysozyme (positively charged) was loaded onto negatively charged eADF4 (C16) particles (size: 521 ± 8.3 nm) taking advantage of the electrostatic interaction between the negatively charged and positively charged groups. They also reported that substantial quantities of lysozyme diffused into the matrix of these particles rather than simply absorbing onto the surface. The release of lysozyme from eADF4 (C16) particles was related to the pH and ionic strength of the medium used. The maximum release of lysozyme was observed after 24 h in medium with acidic pH, and no significant release was observed in medium with a neutral pH even after 28 days [180]. It was reported that SF and biological molecules could interact via electrostatic interaction, hydrophobic attraction and Coulomb forces [25, 181, 182]. However, there is still less knowledge about the exact mechanism of these interactions and needs additional investigations. In this regards, Germershaus et al. have investigated the mechanism of SF interaction with polylysine and protamine in cosmotropic or chaotropic environment. The positive net charge of these biomolecules was responsible for inducing electrostatic interaction with negatively charged SF protein. The ionic strength and the type of deployed affected the interaction between SF nanoparticle and two basic model proteins. Increasing ionic strength using sodium chloride has decreased the zeta potential of SF and its electrostatic interactions but the impact on micelle stability was minimal. In contrast, chaotropic environments were lead to micelle destabilization due to hydrophobic collapse of SF and efficient coacervates formation. The opposite was observed for cosmotropic conditions (micelle stability and abolished coacervate formation)

18

ACCEPTED MANUSCRIPT

MA

NU

SC

RI

PT

(Illustration 1). As a result, the interaction of SF with basic model proteins can be optimized by rationale salt selection [183].

Illustration 1. The effect of chaotropic and cosmotropic salts on SF micelle stability.

AC CE P

TE

D

The loading efficiency and release patterns of hydrophobic and protein drugs from SF particles were evaluated by Shi et al [154]. Self-assembled SF particles with average size of 980 nm were prepared in order to evaluate the drug loading capacity. The actual loading of fluorescein isothiocyanate-labeled bovine serum albumin (FITC–BSA) was 0.27% with 80.06% encapsulation efficiency, while these for rhodamine B (RhB) were 0.153% and 45.87%, respectively. The high encapsulation efficiency of drug was related to protein structure and high molecular weight of FITC–BSA than RhB. The low molecular weight of RhB (479 Da) in comparison to FITC–BSA (66,000) was lead to easy diffuse out from SF particles and thus less encapsulation efficiency. Additionally, this study showed that about 23% FITC-BSA and 34% RhB were released from the silk particles in 50 days [154]. It was well known that the half-life of an enzyme could be increased by immobilizing it on a polymeric substrate by using covalent conjugation or physical attachment [184-186]. There are many reports of the immobilization of enzymes on woven silk and diazotized silk fiber [187]. Besides these substrates, there are only a few investigations concerning the use of silk powder for enzyme immobilization. For instance, it was reported in 1981 that both neutral and alkaline proteases could be immobilized in silk powder at their isoelectric point in pH 5.0–5.5 [143, 188]. In addition, some researchers have described that covalently binding of insulin, L-asparaginase (ASNase), and β-glucosidase to SF nanoparticles enhances their biological stability and activity in vitro [189-191]. Zhang et al. have shown that rapid introduction of SF solution containing ASNase into excess acetone could not inactivate the enzyme and was able to embed and immobilize the enzyme in simultaneously formed SF nanoparticles. The crystalline globular SF nanoparticle–ASNase bioconjugates had 50–120 nm diameter with 90% enzyme activity recovery. The enzyme-entrapped SF nanoparticles produce high temperature resistance in dry conditions, trypsin digestion resistance, higher stability in serum and greater storage stability in solution. Although, the exact formation mechanism, and the reason of using acetone as a preferable agent to prepare SF nanoparticles or SF nanoparticles–ASNases with good bioactivity, is unknown, but the authors stated that this 19

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

phenomena might be attributed to the cleaved micelles aggregated by these degraded polypeptide chains of the regenerated SF and the moderate polarity and hydrophilicity of acetone [192]. Another important application of protein delivery is in tissue engineering. Many growth factors have been incorporated in different scaffolds to enhance tissue regeneration. In addition, there has been extensive research on improving vascularization during the development and repair of tissue [193, 194]. Many strategies can be used for this purpose such as enhancing the pore size and interconnectivity of scaffolds [195], promoting in vitro prevascularization by using cocultures of endothelial cells or mesenchymal stem cells [196, 197], and incorporating angiogenic factors, including vascular endothelial growth factor (VEGF), transforming growth factor (TGF), platelet derived growth factor (PDGF), and fibroblast growth factor (FGF) [198]. Recently, SF nanoparticles with an average size of 150–170 nm have been used to control the release rate of VEGF in a sustained manner. In this study, it was shown that the SF nanoparticles that had accumulated in the cytoplasm of the cells were nontoxic and that normal cell cycle distribution was maintained [166]. We have recently fabricated a bio-hybrid SF/calcium phosphate/PLGA nanocomposite scaffold for controlling the release profile of VEGF in order to enhance bone regeneration. In this study, we first prepared a porous scaffold based on SF and calcium phosphate by using freeze-drying method. After that, VEGF/SF was electrospun onto the surface of the freeze-dried scaffold. PLGA nanofibers were electrospun onto the VEGF loaded SF/calcium phosphate scaffold in order to control the release profile of VEGF (Figure 2). The release profile of VEGF that we obtained over 28 days established the efficacy of this scaffold as a sustained delivery system. The bioactivity of the released VEGF was measured to be about 83% [199].

Figure 2. Schematic illustration of scaffold fabrication processes. (a) SF solution containing CaP powder, (b) freeze-drying of SF/CaP solution, (c) loading VEGF/SF nanofibers on porous SF/CaP substrate using electrospinning, (d) electrospinning of PLGA on VEGF loaded SF/CaP scaffold. Despite the potential of VEGF as an angiogenic factor, it produces immature blood vessels with high permeability [3, 200]. Thus, the incorporation of mature angiogenic factor such as PDGF is required [200]. It seems that the delivery of VEGF and PDGF within scaffolds could mimic the process of natural tissue regeneration. Therefore, in another study, we designed a nanocomposite scaffold based on silk/calcium phosphate/PLGA for the sustained release of PDGF and VEGF, concomitantly [201]. PDGF showed a slower release rate than VEGF because it was loaded deeper in the scaffold and was embedded within two layers of PLGA. In a rabbit model, neovascularization occurred 10 weeks after the implantation of the scaffold (Figure 3). These studies have shown that the electrospinning of SF with different types of angiogenic factors such as PDGF and VEGF could be an effective nanosystem for drug delivery and tissue regeneration applications [201]. 20

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

TE

D

Figure 3. Histological examination of rabbit bones without scaffolds (a and b), SF/CaP scaffold (c and d) and angiogenic loaded scaffold (e and f). Preexisted bone (*), newly formed bone (B), fibrous tissue (F), newly formed capillaries (arrows) and the materials (----). Bars: 50µm (a, c and e) and 30 µm (b, d and f). Magnification: 400× (a, c and e), 500× (b, d and f). Table 6. SF protein as matrix for protein delivery in different tissue constructs.

NGF1

IGF-I2

FGF-23 BMP5

IgG16 monoclonal antibody

Application

Type of delivery system

AC CE P

Types of peptide or protein

Preparation method

Release time

Axon

Nerve conduit

Freeze drying

More than 3 weeks

Cartilage

Porous scaffold

Freeze drying

For 25 days

Not reported

Film

Bone

Disk shaped scaffold

-

Hydrogels and lyogel

Key findings

Ref.

Maintenance of fully bioactive NGF after release, induction of PC12 cell differentiation and neurite outgrowth

[202]

[203]

Diazonium coupling reaction

-

Sonication

For 6 days

Not reported

For 80 days

Chondrogenesis induction Reduced metabolic activities of hMSC4, increased levels of pERK1/2 Promotion of neoosteogenesis after 8 week implantation Antibody release dependence on hydrophobic interactions, hydration resistance, and ionic repulsions

[204]

[205]

[181]

[206]

21

ACCEPTED MANUSCRIPT

-

EGF7

Brain

Film

Adenosine-releasing scaffold with neuroprotective, anti-ictogenic and anti-epileptogenic properties

PT

Adenosine

For 2 Weeks

[207]

Silk films, Casting and Not lamellar electrospinning reported porous silk films and electrospun silk nanofibers 1 Nerve growth factor, 2Insulin-like growth factor I, 3Fibroblast growth factor 2, 4Human mesenchymal stem cell, 5Bone morphogenetic protein, 6Immunoglobulin G, 7Epidermal growth factor.

NU

SC

RI

Skin

Increasing wound healing rate, re-epithelialization, dermis proliferation

MA

4.2 Silk fibroin nanoparticulate for small molecules delivery

AC CE P

TE

D

Nowadays, the use of nanoparticles to deliver small molecules is rapidly growing in many fields [208-211]. Although small molecules have been used as chemotherapeutic agent for many years, there are several disadvantages to their use, including hydrophobicity, nonspecific targeting and biodistribution, and drug resistance shortly after initial treatment. The unique properties of nanoparticles could overcome the limitations of using small molecules as therapeutic agents in biomedical applications [212]. The first polymeric carrier used to deliver small molecules was PGA [213]. To date, there are two PGA based carriers approved for clinical trials: Xyotax (PGA–paclitaxel) [214] and CT-2106 (PGA–camptothecin) [215, 216]. As a natural polymer, silk attracts extensive attention as a possible method of delivering bioactive small molecules [110, 217-219]. Silk is used as a vehicle to deliver small molecules in various forms, including films, 3D porous scaffolds, microspheres, microcapsules, transdermal microneedles, nanospheres, and hydrogels [10]. Different small molecules such as alcian blue, rhodamine B, and crystal violet could be incorporated to silk particles as reported by Lammel et al.[153]. Silk particles produced with 1.25 M potassium phosphate has shown dominate silk II (crystalline) and silk I (less crystalline) in pH 6 and pH 9, respectively. Additionally, they showed that small molecules had 95% loading efficiency because of a charge-charge interaction with silk particles (The negative surface charge of silk particles with positively charged small molecules). They also have found that the release of these three molecules is highly related to their charge and the structure of silk. For instance, the structure of silk II and the pH of solution could induce the burst release of molecules at all-time intervals [153]. It is necessary in many diseases to administer two or more drugs concomitantly. For this purpose, dual drug delivery systems have been introduced in recent years to control the release rate of all the incorporated drugs and achieve optimal therapeutic efficacy [2, 3, 220]. However, no significant successes in clinical trials have been reported regarding the design of suitable carriers with the ability to control the release rate of each molecule independently with all the drugs embedded in the same polymeric matrix [221]. Hydrogels are a versatile polymeric network that could be used for the delivery of cells, drugs, antibodies, proteins, peptides, and genes because their structure changes depending on salt concentration, pH, and temperature [222-224]. Kaplan et al. showed that silk hydrogels could be prepared by initiating the gelation of silk solutions by changing pH or applying ultrasonication and vortexing [158, 225, 226]. Similarly, Naumata et al. have fabricated a dual 22

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

delivery system based on silk nanoparticles incorporated into silk hydrogels consisting of silkmicrofibril networks. Silk nanoparticles had approximately 175 ± 3 nm diameter. The atomic force microscopy (AFM) has shown that silk nanoparticles were aggregated prior to ethanol treatment while no aggregation was observed after ethanol treatment and homogeneous size distribution. It was reported that silk nanoparticles had encapsulation efficiency of approximately 35% for Texas Red (TR) as well as that of around 55% for RhB and FITC. They have also claimed that the release rate of dyes from silk nanoparticles was related to the presence of protease XIV, which is known to degrade the β-sheet structures of silk proteins, because no significant release of RhB was observed from silk nanoparticles without using protease. None of the dyes exhibited burst release, indicating that they were not attached at the surface but rather had been incorporated into the silk particles. However, after enzymatic treatment, significant amounts of RhB and TR were released from the silk nanoparticles through enzymatic degradation especially after 9 h. RhB and TR have shown similar release pattern as a result of similar hydrophobicity and size. On the contrary, more hydrophobic nature of FITC was responsible for slower release behavior in 24 h due to stronger hydrophobic interactions between the silk molecules and FITC. The authors claimed that the release rate of molecules from their system was highly dependent on physical properties such as β-sheet content, the size of the silk nanoparticles and the network size of the silk hydrogels [225]. In addition to hydrogels, micro and nanoparticulate systems also have unique properties, which make them a great candidate for biomedical applications [136, 227]. Generally, the fabrication of silk nanospheres is a more challenging area of research than the fabrication of silk microspheres because of the high molecular weight and protein nature of silk [110, 148, 228]. However, it was reported that by using 70% (v/v) water-miscible protonic and polar aprotonic organic solvents, it is possible to fabricate silk nanospheres in a size range of 300–400 nm [172]. Silk micro and nanospheres could also be prepared with controlled size and shape by using poly(vinyl alcohol) (PVA) as the continuous phase. Silk and PVA were phase separated at a weight ratio of 1:1 and 1:4, respectively. It was observed that using 1/4 ratio sample could generate more homogenous spheres with size distribution ranging from 300 nm to 20 µm. The concentration of silk and PVA played an important role in generating spheres of different sizes and shapes (Figure 4). The loading efficiency of RhB, tetramethylrhodamine conjugated bovine serum albumin (TMR-BSA) and tetramethylrhodamine conjugated dextran (TMR-Dextran) were 95%, 51% and 1.2% respectively. It is suggested that strong binding of RhB to silk is due to hydrophobic and electrostatic attraction that lead to high loading efficiency and a slow release rate. The release rate of TMR-BSA and RhB were less than 5% of total loading during 2 weeks. However, total loading for dextran was 60% with faster release profile within 2 weeks. Since the molecular weight of RhB is much lower than that of TMR-BSA and TMR-dextran, the results confirmed that the interaction between silk and encapsulated drug is controlled by drug release rather than diffusion [158].

23

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

AC CE P

TE

D

Figure 4. SEM images of silk spheres with size controlled by varying silk and PVA concentration. Silk/PVA weight ratio was 1/4. The samples of 5 and 1 wt% silk and PVA concentration were dominated by silk microspheres with a size range of 1–30 µm (A,B,D,E) whereas the samples of 0.2 wt% was dominated by nanospheres with a size lower than 400 nm (C,F). Upper panel shows low magnification images, scale bar; 10 µm in A, B to show multiple microspheres in general; 1 µm in C to show multiple nanospheres. Lower panel show high magnification images, scale bar; 2 µm in D,E; 200 nm in F to show detailed structure of nanospheres 4.3 Silk fibroin nanoparticulate for anticancer delivery Despite the investment of billions of dollars into the investigation of the mechanisms of tumorigenesis, more than 10 million people are exposed to different cancers, annually. Cancer is the leading cause of death worldwide, and its rate of incidence is increasing every year. It is estimated that 15 million people will suffer from cancer until 2020 [229-232]. Many strategies have been developed for cancer treatment: immunotherapy, thermal therapy, phototherapy [233, 234], surgery, gene therapy, chemotherapy, and radiotherapy. Each of these methods has its own advantages and disadvantages [235]. Many researchers are keen to develop new strategies for improved cancer therapy with fewer side effects and improved tumor-targeting ability [236]. Recently, nanotechnology has offered a new avenue for cancer treatment by providing unique nanoparticles based on synthetic and natural polymers for drug delivery applications. Some of these vehicles are in the pre-clinical or clinical phases of development [237-239]. Silk has many unique properties that make it suitable for preparing drug delivery vehicles, including fibers, films, 3D scaffold, and gels [25]. For example, it was reported that by using a capillary-microdot technique, SF nanoparticles could be produced with an average size (> 6.0 > 7.4) (Illustration 2). Two important features of SF nanoparticles including pH-dependent drug release and lysosomal accumulation of SF nanoparticles have confirmed the ability of drug-loaded SF nanoparticles to serve as a lysosomotropic anticancer nanomedicine [164].

Illustration 2. Schematic representation of silk pH dependent release of doxorubicin from a silk nanoparticle. Consistent with this study, another publication revealed that a greater release of doxorubicin was observed at pH 4.5 in comparison to pH 7.4 from folate conjugated SF nanoparticles. It was 25

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

suggested that the greater release of doxorubicin at pH 4.5 was due to many factors, including weak binding between drug and the carboxylic group of silk, reprotonation of the amino groups of doxorubicin, and higher solubility of the drug at this pH [242]. SF nanoparticles are also useful for other anticancers delivery. For example, paclitaxel-encapsulated SF nanoparticles were used for locoregional gastric cancer therapy. The drug loading and encapsulation efficiency were 10 ± 2% and 52 ± 2%, respectively. Paclitaxel had a burst release of 40.9 ± 2.7% during the first 8 h followed by steady and sustained release about 47.2 ± 1.5% for 100 h. The authors have claimed that this release behavior could be related to locating paclitaxel at the hydrophobic/hydrophilic interface inside of the nanoparticles that resulted in initial paclitaxel diffusion from the nanoparticles. Another reason was that the release speed was depended on the drug payload and drug density in the nanoparticles, which was higher paclitaxel loading leading to faster release. Additionally, SF nanoparticles could be taken up by human gastric cancer cells and paclitaxel released from paclitaxel-SF-nanoparticles kept its pharmacological activity [243]. SF nanospheres have also been used for paclitaxel delivery. The maximum loading of paclitaxel in 270- to 520-nm nanospheres was about 6.9%, which was released over 9 days as reported by Chen and colleagues. The authors described that the drug loading, encapsulation efficiency, and release rate of paclitaxel-loaded SF nanospheres was highly dependent on SF concentration and the initial paclitaxel-loading capacity [244]. It is beyond doubt that combining SF with other natural and synthetic polymers could improve its structural, mechanical, and degradation properties along with its drug release profile, drug retention, and encapsulation efficiency [245, 246]. For example, the synthesis of self-aggregating SF-albumin nanoparticles for delivering methotrexate as an antitumor drug was evaluated by Subia and co-workers. Drug loading and encapsulation efficiency were 15–24% and 83–87%., respectively. It was observed that bulk SF and SF-albumin blends (1:1, 2:1) have higher drug loading and encapsulation efficiency than any other blended particles (1:2 and bulk albumin). These observations may be related to hydrophobic nature of SF protein and the greater electrostatic interaction between SF and albumin. Moreover, 35% of drug was released from nanoparticles during the first 48 h and this was increased with time. The drug release from SF-albumin (2:1) was 82%, while 70% of the total drug was released from SF-albumin (1:1). This suggested the higher drug loading ability and controlled release rate of the particles mainly due to surface localization. Increasing the SF content in nanoparticles causes faster diffusion of entrapped drug into the polymer matrix, which may be due to the hydrophobicity of SF protein [247]. Finally, more intensive examinations of the potential role of SF in drug delivery, and its ability to mediate sustained release may lead to further applications of SF-based drug delivery vehicles. 5. Conclusions Many efforts have been made to improve the therapeutic efficacy of biomolecules for various biomedical applications. Nanotechnology has led to the development of nanoparticle-based drug delivery vehicles in the ―nanometer‖ size range in order to overcome the side effects associated with chemotherapeutics. Silk-based nanoparticles have been developed to deliver proteins, small molecules, and anticancer drugs as described in this review. SF has many unique characteristics, including appropriate mechanical properties, versatile processability in an aqueous environment, biocompatibility, and a controlled degradation rate that make it an excellent candidate for drug delivery applications. For this reason, SF nanoparticles have been successfully designed and are able to control the release rate of biomolecules in a sustained manner with high stability. Overall, 26

ACCEPTED MANUSCRIPT applications of SF in drug delivery promise further opportunities for the future use of both natural and engineered silk proteins.

PT

Acknowledgments

AC CE P

TE

D

MA

NU

SC

RI

This study was funded by the National Science Council of Taiwan through research grant NSC 1032221-E-011-032.

27

ACCEPTED MANUSCRIPT References

AC CE P

TE

D

MA

NU

SC

RI

PT

[1] J. Lehár, A.S. Krueger, W. Avery, A.M. Heilbut, L.M. Johansen, E.R. Price, R.J. Rickles, G.F. Short Iii, J.E. Staunton, X. Jin, Synergistic drug combinations tend to improve therapeutically relevant selectivity, Nature biotechnology, 27 (2009) 659-666. [2] J. Wei, F. Chen, J.-W. Shin, H. Hong, C. Dai, J. Su, C. Liu, Preparation and characterization of bioactive mesoporous wollastonite–polycaprolactone composite scaffold, Biomaterials, 30 (2009) 1080-1088. [3] T.P. Richardson, M.C. Peters, A.B. Ennett, D.J. Mooney, Polymeric system for dual growth factor delivery, Nature biotechnology, 19 (2001) 1029-1034. [4] Z. Ghasemi, R. Dinarvand, F. Mottaghitalab, M. Esfandyari-Manesh, E. Sayari, F. Atyabi, Aptamer decorated hyaluronan/chitosan nanoparticles for targeted delivery of 5-fluorouracil to MUC1 overexpressing adenocarcinomas, Carbohydrate Polymers, 121 (2015) 190-198. [5] Y. Zhang, H.F. Chan, K.W. Leong, Advanced materials and processing for drug delivery: the past and the future, Advanced drug delivery reviews, 65 (2013) 104-120. [6] L.S. Nair, C.T. Laurencin, Polymers as biomaterials for tissue engineering and controlled drug delivery, in: Tissue engineering I, Springer, 2006, pp. 47-90. [7] N. Aboudzadeh, M. Imani, M.A. Shokrgozar, A. Khavandi, J. Javadpour, Y. Shafieyan, M. Farokhi, Fabrication and characterization of poly (D, L‐lactide‐co‐glycolide)/hydroxyapatite nanocomposite scaffolds for bone tissue regeneration, Journal of Biomedical Materials Research Part A, 94 (2010) 137-145. [8] M. Farokhi, S. Sharifi, Y. Shafieyan, Z. Bagher, F. Mottaghitalab, A. Hatampoor, M. Imani, M. Shokrgozar, Porous crosslinked poly (ε‐caprolactone fumarate)/nanohydroxyapatite composites for bone tissue engineering, Journal of Biomedical Materials Research Part A, 100 (2012) 1051-1060. [9] S. Mao, C. Guo, Y. Shi, L.C. Li, Recent advances in polymeric microspheres for parenteral drug delivery-part 1, Expert opinion on drug delivery, 9 (2012) 1161-1176. [10] T. Yucel, M.L. Lovett, D.L. Kaplan, Silk-Based Biomaterials for Sustained Drug Delivery, Journal of Controlled Release, (2014). [11] T. Estey, J. Kang, S.P. Schwendeman, J.F. Carpenter, BSA degradation under acidic conditions: a model for protein instability during release from PLGA delivery systems, Journal of pharmaceutical sciences, 95 (2006) 1626-1639. [12] A. Giteau, M.-C. Venier-Julienne, A. Aubert-Pouëssel, J.-P. Benoit, How to achieve sustained and complete protein release from PLGA-based microparticles?, International journal of pharmaceutics, 350 (2008) 14-26. [13] H. Tamber, P. Johansen, H.P. Merkle, B. Gander, Formulation aspects of biodegradable polymeric microspheres for antigen delivery, Advanced drug delivery reviews, 57 (2005) 357376. [14] C.F. van der Walle, G. Sharma, M. Ravi Kumar, Current approaches to stabilising and analysing proteins during microencapsulation in PLGA, (2009). [15] A.O. Elzoghby, W.M. Samy, N.A. Elgindy, Protein-based nanocarriers as promising drug and gene delivery systems, Journal of Controlled Release, 161 (2012) 38-49. [16] P.B. Malafaya, G.A. Silva, R.L. Reis, Natural–origin polymers as carriers and scaffolds for biomolecules and cell delivery in tissue engineering applications, Advanced drug delivery reviews, 59 (2007) 207-233. [17] J. Mano, G. Silva, H.S. Azevedo, P. Malafaya, R. Sousa, S. Silva, L. Boesel, J.M. Oliveira, T. Santos, A. Marques, Natural origin biodegradable systems in tissue engineering and 28

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

regenerative medicine: present status and some moving trends, Journal of the Royal Society Interface, 4 (2007) 999-1030. [18] H. Hosseinkhani, M. Hosseinkhani, H. Kobayashi, Design of tissue-engineered nanoscaffold through self-assembly of peptide amphiphile, Journal of bioactive and compatible polymers, 21 (2006) 277-296. [19] H. Hosseinkhani, T. Kushibiki, K. Matsumoto, T. Nakamura, Y. Tabata, Enhanced suppression of tumor growth using a combination of NK4 plasmid DNA-PEG engrafted cationized dextran complex and ultrasound irradiation, Cancer Gene Therapy, 13 (2006) 479489. [20] H. Hosseinkhani, T. Aoyama, O. Ogawa, Y. Tabata, Ultrasound enhances the transfection of plasmid DNA by non-viral vectors, Current pharmaceutical biotechnology, 4 (2003) 109-122. [21] M. Konishi, Y. Tabata, M. Kariya, H. Hosseinkhani, A. Suzuki, K. Fukuhara, M. Mandai, K. Takakura, S. Fujii, In vivo anti-tumor effect of dual release of cisplatin and adriamycin from biodegradable gelatin hydrogel, Journal of controlled release, 103 (2005) 7-19. [22] Y. Tabata, Tissue regeneration based on growth factor release, Tissue Engineering, 9 (2003) 5-15. [23] F. Mottaghitalab, M. Farokhi, V. Mottaghitalab, M. Ziabari, A. Divsalar, M.A. Shokrgozar, Enhancement of neural cell lines proliferation using nano-structured chitosan/poly (vinyl alcohol) scaffolds conjugated with nerve growth factor, Carbohydrate Polymers, 86 (2011) 526535. [24] M.A. Shokrgozar, F. Mottaghitalab, V. Mottaghitalab, M. Farokhi, Fabrication of porous chitosan/poly (vinyl alcohol) reinforced single-walled carbon nanotube nanocomposites for neural tissue engineering, Journal of biomedical nanotechnology, 7 (2011) 276-284. [25] E. Wenk, H.P. Merkle, L. Meinel, Silk fibroin as a vehicle for drug delivery applications, Journal of Controlled Release, 150 (2011) 128-141. [26] A.D. Bangham, Membrane models with phospholipids, Progress in biophysics and molecular biology, 18 (1968) 29-95. [27] J. Kreuter, S. Mills, S. Davis, C. Wilson, Polybutylcyanoacrylate nanoparticles for the delivery of [< sup> 75 Se] norcholestenol, International Journal of Pharmaceutics, 16 (1983) 105-113. [28] D. Scherer, Einfluß von Polybutylcyanoacrylat-Nanopartikeln auf die orale Absorption von Arzneistoffen, Johann Wolfgang Goethe Universitèat, 1992. [29] C.S. Maia, W. Mehnert, M. Schäfer-Korting, Solid lipid nanoparticles as drug carriers for topical glucocorticoids, International journal of pharmaceutics, 196 (2000) 165-167. [30] B. Müller, J. Kreuter, Enhanced transport of nanoparticle associated drugs through natural and artificial membranes—a general phenomenon?, International journal of pharmaceutics, 178 (1999) 23-32. [31] M. Ricci, C. Puglia, F. Bonina, C.D. Giovanni, S. Giovagnoli, C. Rossi, Evaluation of indomethacin percutaneous absorption from nanostructured lipid carriers (NLC): in vitro and in vivo studies, Journal of pharmaceutical sciences, 94 (2005) 1149-1159. [32] C. Burda, X. Chen, R. Narayanan, M.A. El-Sayed, Chemistry and properties of nanocrystals of different shapes, Chemical reviews, 105 (2005) 1025-1102. [33] A.K. Boal, V.M. Rotello, Fabrication and self-optimization of multivalent receptors on nanoparticle scaffolds, Journal of the American Chemical Society, 122 (2000) 734-735. [34] C. Murray, D.J. Norris, M.G. Bawendi, Synthesis and characterization of nearly monodisperse CdE (E= sulfur, selenium, tellurium) semiconductor nanocrystallites, Journal of 29

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

the American Chemical Society, 115 (1993) 8706-8715. [35] J. McKinnie, Nanobiotechnology offers a promising solution overcoming common drug delivery failures, Drug Delivery Technology, 5 (2006) 11-15. [36] S.M. Moghimi, A.C. Hunter, J.C. Murray, Long-circulating and target-specific nanoparticles: theory to practice, Pharmacological reviews, 53 (2001) 283-318. [37] M.R. Avadi, A.M.M. Sadeghi, N. Mohammadpour, S. Abedin, F. Atyabi, R. Dinarvand, M. Rafiee-Tehrani, Preparation and characterization of insulin nanoparticles using chitosan and Arabic gum with ionic gelation method, Nanomedicine: Nanotechnology, Biology and Medicine, 6 (2010) 58-63. [38] H. Hosseinzadeh, F. Atyabi, R. Dinarvand, S.N. Ostad, Chitosan–Pluronic nanoparticles as oral delivery of anticancer gemcitabine: preparation and in vitro study, International journal of nanomedicine, 7 (2012) 1851. [39] A. Tahamtan, A. Tabarraei, A. Moradi, M. Dinarvand, M. Kelishadi, A. Ghaemi, F. Atyabi, Chitosan nanoparticles as a potential nonviral gene delivery for HPV-16 E7 into mammalian cells, Artificial cells, nanomedicine, and biotechnology, (2014) 1-7. [40] H. Hosseinkhani, Y. Tabata, Self assembly of DNA nanoparticles with polycations for the delivery of genetic materials into cells, Journal of nanoscience and nanotechnology, 6 (2006) 2320-2328. [41] S. Abdullah, W.Y. Wendy-Yeo, H. Hosseinkhani, M. Hosseinkhani, E. Masrawa, R. Ramasamy, R. Rosli, S.A. Rahman, A.J. Domb, Gene transfer into the lung by nanoparticle dextran-spermine/plasmid DNA complexes, BioMed Research International, 2010 (2010). [42] T. Lammers, F. Kiessling, W.E. Hennink, G. Storm, Drug targeting to tumors: principles, pitfalls and (pre-) clinical progress, Journal of controlled release, 161 (2012) 175-187. [43] Y. Ding, S. Li, G. Nie, Nanotechnological strategies for therapeutic targeting of tumor vasculature, Nanomedicine, 8 (2013) 1209-1222. [44] S. Nie, Y. Xing, G.J. Kim, J.W. Simons, Nanotechnology applications in cancer, Annu. Rev. Biomed. Eng., 9 (2007) 257-288. [45] F. Alexis, E.M. Pridgen, R. Langer, O.C. Farokhzad, Nanoparticle technologies for cancer therapy, in: Drug Delivery, Springer, 2010, pp. 55-86. [46] M. Kanapathipillai, A. Brock, D.E. Ingber, Nanoparticle targeting of anti-cancer drugs that alter intracellular signaling or influence the tumor microenvironment, Advanced drug delivery reviews, (2014). [47] S.A. Galindo-Rodriguez, E. Allemann, H. Fessi, E. Doelker, Polymeric nanoparticles for oral delivery of drugs and vaccines: a critical evaluation of in vivo studies, Critical Reviews™ in Therapeutic Drug Carrier Systems, 22 (2005). [48] D.F. Emerich, C.G. Thanos, The pinpoint promise of nanoparticle-based drug delivery and molecular diagnosis, Biomolecular engineering, 23 (2006) 171-184. [49] J. Shi, A.R. Votruba, O.C. Farokhzad, R. Langer, Nanotechnology in drug delivery and tissue engineering: from discovery to applications, Nano letters, 10 (2010) 3223-3230. [50] D.W. Bartlett, H. Su, I.J. Hildebrandt, W.A. Weber, M.E. Davis, Impact of tumor-specific targeting on the biodistribution and efficacy of siRNA nanoparticles measured by multimodality in vivo imaging, Proceedings of the National Academy of Sciences, 104 (2007) 15549-15554. [51] M.P. Desai, V. Labhasetwar, E. Walter, R.J. Levy, G.L. Amidon, The mechanism of uptake of biodegradable microparticles in Caco-2 cells is size dependent, Pharmaceutical research, 14 (1997) 1568-1573. [52] J. Panyam, S.K. Sahoo, S. Prabha, T. Bargar, V. Labhasetwar, Fluorescence and electron 30

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

microscopy probes for cellular and tissue uptake of poly (d, l-lactide-< i> co-glycolide) nanoparticles, International journal of pharmaceutics, 262 (2003) 1-11. [53] F. Greco, M.J. Vicent, Combination therapy: opportunities and challenges for polymer–drug conjugates as anticancer nanomedicines, Advanced drug delivery reviews, 61 (2009) 1203-1213. [54] S. Mitragotri, Synergistic effect of enhancers for transdermal drug delivery, Pharmaceutical Research, 17 (2000) 1354-1359. [55] C. Walsh, Molecular mechanisms that confer antibacterial drug resistance, Nature, 406 (2000) 775-781. [56] M.P. Desai, V. Labhasetwar, G.L. Amidon, R.J. Levy, Gastrointestinal uptake of biodegradable microparticles: effect of particle size, Pharmaceutical research, 13 (1996) 18381845. [57] B.D. Chithrani, W.C. Chan, Elucidating the mechanism of cellular uptake and removal of protein-coated gold nanoparticles of different sizes and shapes, Nano letters, 7 (2007) 15421550. [58] R. Singh, J.W. Lillard Jr, Nanoparticle-based targeted drug delivery, Experimental and molecular pathology, 86 (2009) 215-223. [59] I. Brigger, C. Dubernet, P. Couvreur, Nanoparticles in cancer therapy and diagnosis, Advanced drug delivery reviews, 54 (2002) 631-651. [60] R. Müller, S. Maaben, H. Weyhers, W. Mehnert, Phagocytic uptake and cytotoxicity of solid lipid nanoparticles (SLN) sterically stabilized with poloxamine 908 and poloxamer 407, Journal of drug targeting, 4 (1996) 161-170. [61] M. Roser, D. Fischer, T. Kissel, Surface-modified biodegradable albumin nano-and microspheres. II: effect of surface charges on in vitro phagocytosis and biodistribution in rats, European journal of pharmaceutics and biopharmaceutics, 46 (1998) 255-263. [62] D.A. Tomalia, H. Baker, J. Dewald, M. Hall, G. Kallos, S. Martin, J. Roeck, J. Ryder, P. Smith, A new class of polymers: starburst-dendritic macromolecules, Polymer Journal, 17 (1985) 117-132. [63] G.S. Kwon, T. Okano, Polymeric micelles as new drug carriers, Advanced Drug Delivery Reviews, 21 (1996) 107-116. [64] E. Locatelli, M.C. Franchini, Biodegradable PLGA-b-PEG polymeric nanoparticles: synthesis, properties, and nanomedical applications as drug delivery system, Journal of Nanoparticle Research, 14 (2012) 1-17. [65] S.J. Holland, B.J. Tighe, P.L. Gould, Polymers for biodegradable medical devices. 1. The potential of polyesters as controlled macromolecular release systems, Journal of Controlled Release, 4 (1986) 155-180. [66] M. Roussaki, A. Gaitanarou, P.C. Diamanti, S. Vouyiouka, C. Papaspyrides, P. Kefalas, A. Detsi, Encapsulation of the natural antioxidant aureusidin in biodegradable PLA nanoparticles, Polymer Degradation and Stability, 108 (2014) 182-187. [67] M. Chen, H. Ouyang, S. Zhou, J. Li, Y. Ye, PLGA-nanoparticle mediated delivery of antiOX40 monoclonal antibody enhances anti-tumor cytotoxic T cell responses, Cellular immunology, 287 (2014) 91-99. [68] J.S. Chawla, M.M. Amiji, Biodegradable poly (ε-caprolactone) nanoparticles for tumortargeted delivery of tamoxifen, International journal of pharmaceutics, 249 (2002) 127-138. [69] T. Akagi, T. Kaneko, T. Kida, M. Akashi, Preparation and characterization of biodegradable nanoparticles based on poly (γ-glutamic acid) with L-phenylalanine as a protein carrier, Journal of Controlled Release, 108 (2005) 226-236. 31

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

[70] J.K. Li, N. Wang, X.S. Wu, Poly (vinyl alcohol) nanoparticles prepared by freezing–thawing process for protein/peptide drug delivery, Journal of controlled release, 56 (1998) 117-126. [71] K. Kisich, S. Gelperina, M. Higgins, S. Wilson, E. Shipulo, E. Oganesyan, L. Heifets, Encapsulation of moxifloxacin within poly (butyl cyanoacrylate) nanoparticles enhances efficacy against intracellular Mycobacterium tuberculosis, International journal of pharmaceutics, 345 (2007) 154-162. [72] B. Petri, A. Bootz, A. Khalansky, T. Hekmatara, R. Müller, R. Uhl, J. Kreuter, S. Gelperina, Chemotherapy of brain tumour using doxorubicin bound to surfactant-coated poly (butyl cyanoacrylate) nanoparticles: revisiting the role of surfactants, Journal of Controlled Release, 117 (2007) 51-58. [73] M. Peracchia, E. Fattal, D. Desmaele, M. Besnard, J. Noel, J. Gomis, M. Appel, J. d’Angelo, P. Couvreur, Stealth® PEGylated polycyanoacrylate nanoparticles for intravenous administration and splenic targeting, Journal of Controlled Release, 60 (1999) 121-128. [74] M. Hans, A. Lowman, Biodegradable nanoparticles for drug delivery and targeting, Current Opinion in Solid State and Materials Science, 6 (2002) 319-327. [75] K. Löw, M. Wacker, S. Wagner, K. Langer, H. von Briesen, Targeted human serum albumin nanoparticles for specific uptake in EGFR-Expressing colon carcinoma cells, Nanomedicine: Nanotechnology, Biology and Medicine, 7 (2011) 454-463. [76] L. Zhao, Y. Zhou, Y. Gao, S. Ma, C. Zhang, J. Li, D. Wang, X. Li, C. Li, Y. Liu, Bovine serum albumin nanoparticles for delivery of tacrolimus to reduce its kidney uptake and functional nephrotoxicity, International journal of pharmaceutics, 483 (2015) 180-187. [77] M. Nicklas, W. Schatton, S. Heinemann, T. Hanke, J. Kreuter, Preparation and characterization of marine sponge collagen nanoparticles and employment for the transdermal delivery of 17β-estradiol-hemihydrate, Drug development and industrial pharmacy, 35 (2009) 1035-1042. [78] Z. Lu, T.-K. Yeh, M. Tsai, J.L.-S. Au, M.G. Wientjes, Paclitaxel-loaded gelatin nanoparticles for intravesical bladder cancer therapy, Clinical cancer research, 10 (2004) 7677-7684. [79] I. Pali-Schöll, H. Szöllösi, P. Starkl, B. Scheicher, C. Stremnitzer, A. Hofmeister, F. RothWalter, A. Lukschal, S.C. Diesner, A. Zimmer, Protamine nanoparticles with CpGoligodeoxynucleotide prevent an allergen-induced Th2-response in BALB/c mice, European Journal of Pharmaceutics and Biopharmaceutics, 85 (2013) 656-664. [80] I. Ezpeleta, J.M. Irache, S. Stainmesse, C. Chabenat, J. Gueguen, Y. Popineau, A.-M. Orecchioni, Gliadin nanoparticles for the controlled release of all-trans-retinoic acid, International Journal of Pharmaceutics, 131 (1996) 191-200. [81] R. Penalva, I. Esparza, M. Agüeros, C.J. Gonzalez-Navarro, C. Gonzalez-Ferrero, J.M. Irache, Casein nanoparticles as carriers for the oral delivery of folic acid, Food Hydrocolloids, 44 (2015) 399-406. [82] T. Mirshahi, J. Irache, J. Gueguen, A. Orecchioni, Development of drug delivery systems from vegetal proteins: legumin nanoparticles, Drug development and industrial pharmacy, 22 (1996) 841-846. [83] P.C. Bessa, R. Machado, S. Nürnberger, D. Dopler, A. Banerjee, A.M. Cunha, J.C. Rodríguez-Cabello, H. Redl, M. van Griensven, R.L. Reis, Thermoresponsive self-assembled elastin-based nanoparticles for delivery of BMPs, Journal of Controlled Release, 142 (2010) 312318. [84] A.M. De Campos, A. Sánchez, M.a.J. Alonso, Chitosan nanoparticles: a new vehicle for the improvement of the delivery of drugs to the ocular surface. Application to cyclosporin A, 32

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

International Journal of Pharmaceutics, 224 (2001) 159-168. [85] J.O. You, C.A. Peng, Calcium‐Alginate Nanoparticles Formed by Reverse Microemulsion as Gene Carriers, in: Macromolecular Symposia, Wiley Online Library, 2005, pp. 147-153. [86] C. Chittasupho, M. Jaturanpinyo, S. Mangmool, Pectin nanoparticle enhances cytotoxicity of methotrexate against hepG2 cells, Drug delivery, 20 (2013) 1-9. [87] D. Song, Y.S. Thio, Y. Deng, Starch nanoparticle formation via reactive extrusion and related mechanism study, Carbohydrate Polymers, 85 (2011) 208-214. [88] K. Kaewprapan, P. Inprakhon, E. Marie, A. Durand, Enzymatically degradable nanoparticles of dextran esters as potential drug delivery systems, Carbohydrate Polymers, 88 (2012) 875-881. [89] H. Guo, Y. Liu, Y. Wang, J. Wu, X. Yang, R. Li, Y. Wang, N. Zhang, pH-sensitive pullulanbased nanoparticle carrier for adriamycin to overcome drug-resistance of cancer cells, Carbohydrate polymers, 111 (2014) 908-917. [90] K.Y. Choi, H. Chung, K.H. Min, H.Y. Yoon, K. Kim, J.H. Park, I.C. Kwon, S.Y. Jeong, Selfassembled hyaluronic acid nanoparticles for active tumor targeting, Biomaterials, 31 (2010) 106114. [91] K. Numata, D.L. Kaplan, Silk-based delivery systems of bioactive molecules, Advanced drug delivery reviews, 62 (2010) 1497-1508. [92] S. Kundu, Silk Biomaterials for Tissue Engineering and Regenerative Medicine, Elsevier, 2014. [93] B. Mahendran, S.K. Ghosh, S.C. Kundu, Molecular phylogeny of silk producing insects based on internal transcribed spacer DNA1, Journal of biochemistry and molecular biology, 39 (2006) 522. [94] M.S. Jolly, S. Sen, M.M. Ahsan, Tasar culture, Ambika publishers Bombay, 1974. [95] G.H. Altman, F. Diaz, C. Jakuba, T. Calabro, R.L. Horan, J. Chen, H. Lu, J. Richmond, D.L. Kaplan, Silk-based biomaterials, Biomaterials, 24 (2003) 401-416. [96] K. McGrath, D. Kaplan, Protein-based materials, Springer, 1997. [97] S. Inoue, K. Tanaka, F. Arisaka, S. Kimura, K. Ohtomo, S. Mizuno, Silk fibroin of Bombyx mori is secreted, assembling a high molecular mass elementary unit consisting of H-chain, Lchain, and P25, with a 6: 6: 1 molar ratio, Journal of Biological Chemistry, 275 (2000) 4051740528. [98] E. Bini, D.P. Knight, D.L. Kaplan, Mapping domain structures in silks from insects and spiders related to protein assembly, Journal of molecular biology, 335 (2004) 27-40. [99] T. Gamo, T. Inokuchi, H. Laufer, Polypeptides of fibroin and sericin secreted from the different sections of the silk gland in< i> Bombyx mori, Insect Biochemistry, 7 (1977) 285295. [100] M. urovec, C. ang, D. Kodr k, F. Sehnal, Identification of a novel type of silk protein and regulation of its expression, Journal of Biological Chemistry, 273 (1998) 15423-15428. [101] M.-p. Ho, H. Wang, K.-t. Lau, Effect of degumming time on silkworm silk fibre for biodegradable polymer composites, Applied Surface Science, 258 (2012) 3948-3955. [102] S. Hofmann, C. Wong Po Foo, F. Rossetti, M. Textor, G. Vunjak-Novakovic, D. Kaplan, H. Merkle, L. Meinel, Silk fibroin as an organic polymer for controlled drug delivery, Journal of Controlled Release, 111 (2006) 219-227. [103] O. Kratky, E. Schauenstein, A. Sekora, An unstable lattice in silk fibroin, Nature, 165 (1950) 319-320. [104] L.F. Drummy, D.M. Phillips, M.O. Stone, B. Farmer, R.R. Naik, Thermally induced αhelix to β-sheet transition in regenerated silk fibers and films, Biomacromolecules, 6 (2005) 33

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

3328-3333. [105] T. Asakura, J. Ashida, T. Yamane, T. Kameda, Y. Nakazawa, K. Ohgo, K. Komatsu, A repeated β-turn structure in Poly (Ala-Gly) as a model for silk I of< i> Bombyx mori silk fibroin studied with two-dimensional spin-diffusion NMR under off magic angle spinning and rotational echo double resonance, Journal of molecular biology, 306 (2001) 291-305. [106] J.P. Anderson, Morphology and crystal structure of a recombinant silk-like molecule, SLP4, (1998). [107] C. Zhao, J. Yao, H. Masuda, R. Kishore, T. Asakura, Structural characterization and artificial fiber formation of Bombyx mori silk fibroin in hexafluoro‐iso‐propanol solvent system, Biopolymers, 69 (2003) 253-259. [108] P. Garside, P. Wyeth, Crystallinity and degradation of silk: correlations between analytical signatures and physical condition on ageing, Applied Physics A, 89 (2007) 871-876. [109] P. Monti, G. Freddi, A. Bertoluzza, N. Kasai, M. Tsukada, Raman spectroscopic studies of silk fibroin from Bombyx mori, Journal of Raman spectroscopy, 29 (1998) 297-304. [110] E. Wenk, A.J. Wandrey, H.P. Merkle, L. Meinel, Silk fibroin spheres as a platform for controlled drug delivery, Journal of Controlled Release, 132 (2008) 26-34. [111] A. Motta, L. Fambri, C. Migliaresi, Regenerated silk fibroin films: thermal and dynamic mechanical analysis, Macromolecular Chemistry and Physics, 203 (2002) 1658-1665. [112] J. Nam, Y.H. Park, Morphology of regenerated silk fibroin: Effects of freezing temperature, alcohol addition, and molecular weight, Journal of Applied Polymer Science, 81 (2001) 30083021. [113] F. Xie, H. Zhang, H. Shao, X. Hu, Effect of shearing on formation of silk fibers from regenerated< i> Bombyx mori silk fibroin aqueous solution, International journal of biological macromolecules, 38 (2006) 284-288. [114] X.-H. Zong, P. Zhou, Z.-Z. Shao, S.-M. Chen, X. Chen, B.-W. Hu, F. Deng, W.-H. Yao, Effect of pH and copper (II) on the conformation transitions of silk fibroin based on EPR, NMR, and Raman spectroscopy, Biochemistry, 43 (2004) 11932-11941. [115] K.S. Soppimath, T.M. Aminabhavi, A.R. Kulkarni, W.E. Rudzinski, Biodegradable polymeric nanoparticles as drug delivery devices, Journal of controlled release, 70 (2001) 1-20. [116] K.A. Athanasiou, A.R. Shah, R.J. Hernandez, R.G. LeBaron, Basic science of articular cartilage repair, Clinics in sports medicine, 20 (2001) 223-247. [117] E. Dawson, G. Mapili, K. Erickson, S. Taqvi, K. Roy, Biomaterials for stem cell differentiation, Advanced drug delivery reviews, 60 (2008) 215-228. [118] M. Farokhi, F. Mottaghitalab, J. Hadjati, R. Omidvar, M. Majidi, A. Amanzadeh, M. Azami, S.M. Tavangar, M.A. Shokrgozar, J. Ai, Structural and functional changes of silk fibroin scaffold due to hydrolytic degradation, Journal of Applied Polymer Science, 131 (2014). [119] E. Wenk, A.J. Meinel, S. Wildy, H.P. Merkle, L. Meinel, Microporous silk fibroin scaffolds embedding PLGA microparticles for controlled growth factor delivery in tissue engineering, Biomaterials, 30 (2009) 2571-2581. [120] R.L. Horan, K. Antle, A.L. Collette, Y. Wang, J. Huang, J.E. Moreau, V. Volloch, D.L. Kaplan, G.H. Altman, In vitro degradation of silk fibroin, Biomaterials, 26 (2005) 3385-3393. [121] Y. Wang, D.D. Rudym, A. Walsh, L. Abrahamsen, H.-J. Kim, H.S. Kim, C. Kirker-Head, D.L. Kaplan, < i> In vivo degradation of three-dimensional silk fibroin scaffolds, Biomaterials, 29 (2008) 3415-3428. [122] H. Hosseinkhani, M. Hosseinkhani, Biodegradable polymer-metal complexes for gene and drug delivery, Current Drug Safety, 4 (2009) 79-83. 34

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

[123] K. Subramani, H. Hosseinkhani, A. Khraisat, M. Hosseinkhani, Y. Pathak, Targeting nanoparticles as drug delivery systems for cancer treatment, Current Nanoscience, 5 (2009) 135140. [124] H. Hosseinkhani, M. Hosseinkhani, E.V. Farahani, M.N. Haghighi, In vitro sustained release and degradation study of biodegradable poly (d, l-lactic acid) microspheres loading theophylline, Advanced Science Letters, 2 (2009) 70-77. [125] M. Mahmoudi, H. Hosseinkhani, M. Hosseinkhani, S. Boutry, A. Simchi, W.S. Journeay, K. Subramani, S. Laurent, Magnetic resonance imaging tracking of stem cells in vivo using iron oxide nanoparticles as a tool for the advancement of clinical regenerative medicine, Chemical reviews, 111 (2010) 253-280. [126] R. Amini, H. Hosseinkhani, A. Jalilian, S. Abdullah, R. Rosli, Engineered smart biomaterials for gene delivery, Gene Ther Mol Biol, 14 (2012) 72-86. [127] H. Hosseinkhani, 3D in vitro technology for drug discovery, Current drug safety, 7 (2012) 37-43. [128] K. Subramani, S. Pathak, H. Hosseinkhani, Recent trends in diabetes treatment using nanotechnology, Dig J Nanomat Biostructures, 7 (2012) 85-95. [129] H. Hosseinkhani, P.-D. Hong, D.-S. Yu, Self-assembled proteins and peptides for regenerative medicine, Chemical reviews, 113 (2013) 4837-4861. [130] C.R. Wittmer, X. Hu, P.-C. Gauthier, S. Weisman, D.L. Kaplan, T.D. Sutherland, Production, structure and in vitro degradation of electrospun honeybee silk nanofibers, Acta biomaterialia, 7 (2011) 3789-3795. [131] S.-H. Park, E.S. Gil, H. Shi, H.J. Kim, K. Lee, D.L. Kaplan, Relationships between degradability of silk scaffolds and osteogenesis, Biomaterials, 31 (2010) 6162-6172. [132] M. Lovett, C. Cannizzaro, L. Daheron, B. Messmer, G. Vunjak-Novakovic, D.L. Kaplan, Silk fibroin microtubes for blood vessel engineering, Biomaterials, 28 (2007) 5271-5279. [133] Y. Yang, Y. Zhao, Y. Gu, X. Yan, J. Liu, F. Ding, X. Gu, Degradation behaviors of nerve guidance conduits made up of silk fibroin in vitro and in vivo, Polymer Degradation and Stability, 94 (2009) 2213-2220. [134] M. Li, M. Ogiso, N. Minoura, Enzymatic degradation behavior of porous silk fibroin sheets, Biomaterials, 24 (2003) 357-365. [135] J. Zhou, C. Cao, X. Ma, L. Hu, L. Chen, C. Wang, In vitro and in vivo degradation behavior of aqueous-derived electrospun silk fibroin scaffolds, Polymer Degradation and Stability, 95 (2010) 1679-1685. [136] F. Chiellini, A.M. Piras, C. Errico, E. Chiellini, Micro/nanostructured polymeric systems for biomedical and pharmaceutical applications, (2008). [137] D.S. Kohane, Microparticles and nanoparticles for drug delivery, Biotechnology and bioengineering, 96 (2007) 203-209. [138] M. Vandelli, F. Rivasi, P. Guerra, F. Forni, R. Arletti, Gelatin microspheres crosslinked with D, L-glyceraldehyde as a potential drug delivery system: preparation, characterisation, in vitro and in vivo studies, International journal of pharmaceutics, 215 (2001) 175-184. [139] R. Arshady, Review: Biodegradable microcapsular drug delivery systems: manufacturing methodology, release control and targeting prospects, Journal of Bioactive and Compatible Polymers, 5 (1990) 315-342. [140] G.A. Digenis, T.B. Gold, V.P. Shah, Cross‐linking of gelatin capsules and its relevance to their in vitro‐in vivo performance, Journal of pharmaceutical sciences, 83 (1994) 915-921. [141] J. Gosline, P. Guerette, C. Ortlepp, K. Savage, The mechanical design of spider silks: from 35

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

fibroin sequence to mechanical function, Journal of Experimental Biology, 202 (1999) 32953303. [142] Y. Horikawa, K. Ohtomo, Finely powdered fibroin and process for producing same, in, Google Patents, 1980. [143] D. Akiyama, K. Hirabayashi, Effect of preparation method of powdered silk on the mechanical properties of moulded silk, Polymer, 35 (1994) 2355-2358. [144] T. Hino, M. Tanimoto, S. Shimabayashi, Change in secondary structure of silk fibroin during preparation of its microspheres by spray-drying and exposure to humid atmosphere, Journal of Colloid and interface science, 266 (2003) 68-73. [145] J.-H. Yeo, K.-G. Lee, Y.-W. Lee, S.Y. Kim, Simple preparation and characteristics of silk fibroin microsphere, European Polymer Journal, 39 (2003) 1195-1199. [146] A. Gholami, H. Tavanai, A. Moradi, Production of fibroin nanopowder through electrospraying, Journal of Nanoparticle Research, 13 (2011) 2089-2098. [147] P. Shi, J.C. Goh, Self-assembled silk fibroin particles: Tunable size and appearance, Powder Technology, 215 (2012) 85-90. [148] X. Wang, E. Wenk, A. Matsumoto, L. Meinel, C. Li, D.L. Kaplan, Silk microspheres for encapsulation and controlled release, Journal of Controlled Release, 117 (2007) 360-370. [149] J.G. Hardy, T.R. Scheibel, Composite materials based on silk proteins, Progress in Polymer Science, 35 (2010) 1093-1115. [150] C. Fu, Z. Shao, V. Fritz, Animal silks: their structures, properties and artificial production, Chemical Communications, (2009) 6515-6529. [151] M. Kazemimostaghim, R. Rajkhowa, T. Tsuzuki, X. Wang, Production of submicron silk particles by milling, Powder Technology, 241 (2013) 230-235. [152] R. Rajkhowa, L. Wang, X. Wang, Ultra-fine silk powder preparation through rotary and ball milling, Powder technology, 185 (2008) 87-95. [153] A.S. Lammel, X. Hu, S.-H. Park, D.L. Kaplan, T.R. Scheibel, Controlling silk fibroin particle features for drug delivery, Biomaterials, 31 (2010) 4583-4591. [154] P. Shi, J.C. Goh, Release and cellular acceptance of multiple drugs loaded silk fibroin particles, International journal of pharmaceutics, 420 (2011) 282-289. [155] Y. Baimark, P. Srihanam, Y. Srisuwan, P. Phinyocheep, Preparation of porous silk fibroin microparticles by a water‐in‐oil emulsification‐diffusion method, Journal of Applied Polymer Science, 118 (2010) 1127-1133. [156] Z. Cao, X. Chen, J. Yao, L. Huang, Z. Shao, The preparation of regenerated silk fibroin microspheres, Soft Matter, 3 (2007) 910-915. [157] Y. Srisuwan, P. Srihanam, Y. Baimark, Preparation of silk fibroin microspheres and its application to protein adsorption, Journal of Macromolecular Science®, Part A: Pure and Applied Chemistry, 46 (2009) 521-525. [158] X. Wang, T. Yucel, Q. Lu, X. Hu, D.L. Kaplan, Silk nanospheres and microspheres from silk/pva blend films for drug delivery, Biomaterials, 31 (2010) 1025-1035. [159] V. Karageorgiou, L. Meinel, S. Hofmann, A. Malhotra, V. Volloch, D. Kaplan, Bone morphogenetic protein‐2 decorated silk fibroin films induce osteogenic differentiation of human bone marrow stromal cells, Journal of Biomedical Materials Research Part A, 71 (2004) 528-537. [160] V. Karageorgiou, M. Tomkins, R. Fajardo, L. Meinel, B. Snyder, K. Wade, J. Chen, G. Vunjak‐Novakovic, D.L. Kaplan, Porous silk fibroin 3‐D scaffolds for delivery of bone morphogenetic protein‐2 in vitro and in vivo, Journal of Biomedical Materials Research Part A, 78 (2006) 324-334. 36

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

[161] B. Kundu, R. Rajkhowa, S.C. Kundu, X. Wang, Silk fibroin biomaterials for tissue regenerations, Advanced drug delivery reviews, 65 (2013) 457-470. [162] R. Rajkhowa, X. Wang, S. Kundu, Silk powder for regenerative medicine, Silk Biomaterials for Tissue Engineering and Regenerative Medicine, (2014) 191. [163] K. Jastrzebska, K. Kucharczyk, A. Florczak, E. Dondajewska, A. Mackiewicz, H. DamsKozlowska, Silk as an innovative biomaterial for cancer therapy, Reports of Practical Oncology & Radiotherapy, (2014). [164] F.P. Seib, G.T. Jones, J. Rnjak‐Kovacina, Y. Lin, D.L. Kaplan, pH‐Dependent Anticancer Drug Release from Silk Nanoparticles, Advanced healthcare materials, 2 (2013) 1606-1611. [165] A.B. Mathur, V. Gupta, Silk fibroin-derived nanoparticles for biomedical applications, Nanomedicine, 5 (2010) 807-820. [166] J. Kundu, Y.-I. Chung, Y.H. Kim, G. Tae, S. Kundu, Silk fibroin nanoparticles for cellular uptake and control release, International journal of pharmaceutics, 388 (2010) 242-250. [167] K.P.R. Chowdary, Y. Srinivasa Rao, Mucoadhesive microspheres for controlled drug delivery, Biological and pharmaceutical Bulletin, 27 (2004) 1717-1724. [168] P.H. Hoet, I. Brüske-Hohlfeld, O.V. Salata, Nanoparticles–known and unknown health risks, Journal of nanobiotechnology, 2 (2004) 12. [169] R.C. Mundargi, V.R. Babu, V. Rangaswamy, P. Patel, T.M. Aminabhavi, Nano/micro technologies for delivering macromolecular therapeutics using poly (D, L-lactide-co-glycolide) and its derivatives, Journal of Controlled Release, 125 (2008) 193-209. [170] E. Rytting, J. Nguyen, X. Wang, T. Kissel, Biodegradable polymeric nanocarriers for pulmonary drug delivery, (2008). [171] J.A. Champion, Y.K. Katare, S. Mitragotri, Particle shape: a new design parameter for micro-and nanoscale drug delivery carriers, Journal of Controlled Release, 121 (2007) 3-9. [172] Y.-Q. Zhang, W.-D. Shen, R.-L. Xiang, L.-J. Zhuge, W.-J. Gao, W.-B. Wang, Formation of silk fibroin nanoparticles in water-miscible organic solvent and their characterization, Journal of Nanoparticle Research, 9 (2007) 885-900. [173] K. Numata, 19 - Silk hydrogels for tissue engineering and dual-drug delivery, in: S.C. Kundu (Ed.) Silk Biomaterials for Tissue Engineering and Regenerative Medicine, Woodhead Publishing, 2014, pp. 503-518. [174] X. Hu, Q. Lu, L. Sun, P. Cebe, X. Wang, X. Zhang, D.L. Kaplan, Biomaterials from ultrasonication-induced silk fibroin− hyaluronic acid hydrogels, Biomacromolecules, 11 (2010) 3178-3188. [175] X. Wang, J.A. Kluge, G.G. Leisk, D.L. Kaplan, Sonication-induced gelation of silk fibroin for cell encapsulation, Biomaterials, 29 (2008) 1054-1064. [176] U.-J. Kim, J. Park, C. Li, H.-J. Jin, R. Valluzzi, D.L. Kaplan, Structure and properties of silk hydrogels, Biomacromolecules, 5 (2004) 786-792. [177] P.C. Bessa, E.R. Balmayor, H.S. Azevedo, S. Nürnberger, M. Casal, M. Van Griensven, R. Reis, H. Redl, Silk fibroin microparticles as carriers for delivery of human recombinant BMPs. Physical characterization and drug release, Journal of tissue engineering and regenerative medicine, 4 (2010) 349-355. [178] L.-Y. Wang, Y.-H. Gu, Z.-G. Su, G.-H. Ma, Preparation and improvement of release behavior of chitosan microspheres containing insulin, International journal of pharmaceutics, 311 (2006) 187-195. [179] U. Bilati, E. Allémann, E. Doelker, Strategic approaches for overcoming peptide and protein instability within biodegradable nano-and microparticles, European Journal of 37

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

Pharmaceutics and Biopharmaceutics, 59 (2005) 375-388. [180] M. Hofer, G. Winter, J. Myschik, Recombinant spider silk particles for controlled delivery of protein drugs, Biomaterials, 33 (2012) 1554-1562. [181] N.A. Guziewicz, A.J. Massetti, B.J. Perez-Ramirez, D.L. Kaplan, Mechanisms of monoclonal antibody stabilization and release from silk biomaterials, Biomaterials, 34 (2013) 7766-7775. [182] A. Matsumoto, J. Chen, A.L. Collette, U.-J. Kim, G.H. Altman, P. Cebe, D.L. Kaplan, Mechanisms of silk fibroin sol-gel transitions, The Journal of Physical Chemistry B, 110 (2006) 21630-21638. [183] O. Germershaus, V. Werner, M. Kutscher, L. Meinel, Deciphering the mechanism of protein interaction with silk fibroin for drug delivery systems, Biomaterials, 35 (2014) 34273434. [184] L.M. Holle, Pegaspargase: an alternative?, Annals of Pharmacotherapy, 31 (1997) 616-624. [185] S.L. Berg, F.M. Balis, C.L. McCully, K.S. Godwin, D.G. Poplack, Pharmacokinetics of PEG-l-asparaginase and plasma and cerebrospinal fluidl-asparagine concentrations in the rhesus monkey, Cancer chemotherapy and pharmacology, 32 (1993) 310-314. [186] M.J. Keating, R. Holmes, S. Lerner, D.H. Ho, L-asparaginase and PEG asparaginase-past, present, and future, Leukemia & lymphoma, 10 (1993) 153-157. [187] L. Grasset, D. Cordier, A. Ville, Woven silk as a carrier for the immobilization of enzymes, Biotechnology and bioengineering, 19 (1977) 611-618. [188] S. Inoue, Y. Matsunaga, H. Iwane, M. Sotomura, T. Nose, Entrapment of phenylalanine ammonia-lyase in silk fibroin for protection from proteolytic attack, Biochemical and biophysical research communications, 141 (1986) 165-170. [189] H.-B. Yan, Y.-Q. Zhang, Y.-L. Ma, L.-X. Zhou, Biosynthesis of insulin-silk fibroin nanoparticles conjugates and in vitro evaluation of a drug delivery system, Journal of Nanoparticle Research, 11 (2009) 1937-1946. [190] Y.-Q. Zhang, R.-L. Xiang, H.-B. Yan, X.-X. Chen, Preparation of silk fibroin nanoparticles and their application to immobilization of L-asparaginase, Chemical Journal of Chinese Universities, 29 (2008) 628-633. [191] . . hou, .Q. hang, Biosynthesis of β-glucosidase-silk fibroin nanoparticles conjugates and enzymatic characteristics, Advanced Materials Research, 175 (2011) 186-191. [192] Y.-Q. Zhang, Y.-J. Wang, H.-Y. Wang, L. Zhu, Z.-Z. Zhou, Highly efficient processing of silk fibroin nanoparticle-l-asparaginase bioconjugates and their characterization as a drug delivery system, Soft Matter, 7 (2011) 9728-9736. [193] M. Grellier, L. Bordenave, J. Amedee, Cell-to-cell communication between osteogenic and endothelial lineages: implications for tissue engineering, Trends in biotechnology, 27 (2009) 562-571. [194] J.J. Moon, J.L. West, Vascularization of engineered tissues: approaches to promote angiogenesis in biomaterials, Current topics in medicinal chemistry, 8 (2008) 300. [195] S. Yang, K.-F. Leong, Z. Du, C.-K. Chua, The design of scaffolds for use in tissue engineering. Part I. Traditional factors, Tissue engineering, 7 (2001) 679-689. [196] R.E. Unger, A. Sartoris, K. Peters, A. Motta, C. Migliaresi, M. Kunkel, U. Bulnheim, J. Rychly, C. James Kirkpatrick, Tissue-like self-assembly in cocultures of endothelial cells and osteoblasts and the formation of microcapillary-like structures on three-dimensional porous biomaterials, Biomaterials, 28 (2007) 3965-3976. [197] J. Rouwkema, J.D. Boer, C.A.V. Blitterswijk, Endothelial cells assemble into a 338

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

dimensional prevascular network in a bone tissue engineering construct, Tissue engineering, 12 (2006) 2685-2693. [198] D. Kaigler, Z. Wang, K. Horger, D.J. Mooney, P.H. Krebsbach, VEGF scaffolds enhance angiogenesis and bone regeneration in irradiated osseous defects, Journal of Bone and Mineral Research, 21 (2006) 735-744. [199] M. Farokhi, F. Mottaghitalab, M.A. Shokrgozar, J. Ai, J. Hadjati, M. Azami, Bio-hybrid silk fibroin/calcium phosphate/PLGA nanocomposite scaffold to control the delivery of vascular endothelial growth factor, Materials Science and Engineering: C, 35 (2014) 401-410. [200] N. Ferrara, H.-P. Gerber, J. LeCouter, The biology of VEGF and its receptors, Nature medicine, 9 (2003) 669-676. [201] M. Farokhi, F. Mottaghitalab, J. Ai, M.A. Shokrgozar, Sustained release of platelet-derived growth factor and vascular endothelial growth factor from silk/calcium phosphate/PLGA based nanocomposite scaffold, International journal of pharmaceutics, 454 (2013) 216-225. [202] L. Uebersax, M. Mattotti, M. Papaloïzos, H.P. Merkle, B. Gander, L. Meinel, Silk fibroin matrices for the controlled release of nerve growth factor (NGF), Biomaterials, 28 (2007) 44494460. [203] L. Uebersax, H.P. Merkle, L. Meinel, Insulin-like growth factor I releasing silk fibroin scaffolds induce chondrogenic differentiation of human mesenchymal stem cells, Journal of controlled release, 127 (2008) 12-21. [204] E. Wenk, A.R. Murphy, D.L. Kaplan, L. Meinel, H.P. Merkle, L. Uebersax, The use of sulfonated silk fibroin derivatives to control binding, delivery and potency of FGF-2 in tissue regeneration, Biomaterials, 31 (2010) 1403-1413. [205] C. Kirker-Head, V. Karageorgiou, S. Hofmann, R. Fajardo, O. Betz, H. Merkle, M. Hilbe, B. Von Rechenberg, J. McCool, L. Abrahamsen, BMP-silk composite matrices heal critically sized femoral defects, Bone, 41 (2007) 247-255. [206] C. Szybala, E.M. Pritchard, T.A. Lusardi, T. Li, A. Wilz, D.L. Kaplan, D. Boison, Antiepileptic effects of silk-polymer based adenosine release in kindled rats, Experimental neurology, 219 (2009) 126-135. [207] E.S. Gil, B. Panilaitis, E. Bellas, D.L. Kaplan, Functionalized silk biomaterials for wound healing, Advanced healthcare materials, 2 (2013) 206-217. [208] R.-D. Hofheinz, S.U. Gnad-Vogt, U. Beyer, A. Hochhaus, Liposomal encapsulated anticancer drugs, Anti-Cancer Drugs, 16 (2005) 691-707. [209] R. Duncan, The dawning era of polymer therapeutics, Nature Reviews Drug Discovery, 2 (2003) 347-360. [210] R. Duncan, Polymer conjugates as anticancer nanomedicines, Nature Reviews Cancer, 6 (2006) 688-701. [211] D.J. Bharali, M. Khalil, M. Gurbuz, T.M. Simone, S.A. Mousa, Nanoparticles and cancer therapy: a concise review with emphasis on dendrimers, International journal of nanomedicine, 4 (2009) 1. [212] Z.G. Chen, Small-molecule delivery by nanoparticles for anticancer therapy, Trends in molecular medicine, 16 (2010) 594-602. [213] C. Li, Poly (L-glutamic acid)–anticancer drug conjugates, Advanced drug delivery reviews, 54 (2002) 695-713. [214] P. Sabbatini, C. Aghajanian, D. Dizon, S. Anderson, J. Dupont, J.V. Brown, W.A. Peters, A. Jacobs, A. Mehdi, S. Rivkin, Phase II study of CT-2103 in patients with recurrent epithelial ovarian, fallopian tube, or primary peritoneal carcinoma, Journal of clinical oncology, 22 (2004) 39

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

4523-4531. [215] K. Cho, X. Wang, S. Nie, D.M. Shin, Therapeutic nanoparticles for drug delivery in cancer, Clinical cancer research, 14 (2008) 1310-1316. [216] J. Homsi, G.R. Simon, C.R. Garrett, G. Springett, R. De Conti, A.A. Chiappori, P.N. Munster, M.K. Burton, S. Stromatt, C. Allievi, Phase I trial of poly-L-glutamate camptothecin (CT-2106) administered weekly in patients with advanced solid malignancies, Clinical Cancer Research, 13 (2007) 5855-5861. [217] A.S. Gobin, R. Rhea, R.A. Newman, A.B. Mathur, Silk-fibroin-coated liposomes for longterm and targeted drug delivery, International journal of nanomedicine, 1 (2006) 81. [218] X. Wang, X. Hu, A. Daley, O. Rabotyagova, P. Cebe, D.L. Kaplan, Nanolayer biomaterial coatings of silk fibroin for controlled release, Journal of Controlled release, 121 (2007) 190-199. [219] E.M. Pritchard, T. Valentin, B. Panilaitis, F. Omenetto, D.L. Kaplan, Antibiotic‐Releasing Silk Biomaterials for Infection Prevention and Treatment, Advanced functional materials, 23 (2013) 854-861. [220] C.M. Owens, C. Mawhinney, J.M. Grenier, R. Altmeyer, M.S. Lee, A.A. Borisy, J. Lehár, L.M. Johansen, Chemical combinations elucidate pathway interactions and regulation relevant to Hepatitis C replication, Molecular systems biology, 6 (2010). [221] K. Karthikeyan, S. Guhathakarta, R. Rajaram, P.S. Korrapati, Electrospun zein/eudragit nanofibers based dual drug delivery system for the simultaneous delivery of aceclofenac and pantoprazole, International journal of pharmaceutics, 438 (2012) 117-122. [222] J.K. Oh, D.J. Siegwart, K. Matyjaszewski, Synthesis and biodegradation of nanogels as delivery carriers for carbohydrate drugs, Biomacromolecules, 8 (2007) 3326-3331. [223] M.S. Muthu, M.K. Rawat, A. Mishra, S. Singh, PLGA nanoparticle formulations of risperidone: preparation and neuropharmacological evaluation, Nanomedicine: Nanotechnology, Biology and Medicine, 5 (2009) 323-333. [224] J.T. Zhang, S.W. Huang, Y.N. Xue, R.X. Zhuo, Poly (N‐isopropylacrylamide) Nanoparticle‐Incorporated PNIPAAm Hydrogels with Fast Shrinking Kinetics, Macromolecular rapid communications, 26 (2005) 1346-1350. [225] K. Numata, S. Yamazaki, N. Naga, Biocompatible and biodegradable dual-drug release system based on silk hydrogel containing silk nanoparticles, Biomacromolecules, 13 (2012) 1383-1389. [226] D.N. Rockwood, R.C. Preda, T. Yücel, X. Wang, M.L. Lovett, D.L. Kaplan, Materials fabrication from Bombyx mori silk fibroin, Nature protocols, 6 (2011) 1612-1631. [227] M.E. Davis, Nanoparticle therapeutics: an emerging treatment modality for cancer, Nature reviews Drug discovery, 7 (2008) 771-782. [228] X. Wang, E. Wenk, X. Hu, G.R. Castro, L. Meinel, X. Wang, C. Li, H. Merkle, D.L. Kaplan, Silk coatings on PLGA and alginate microspheres for protein delivery, Biomaterials, 28 (2007) 4161-4169. [229] J. Ferlay, H.R. Shin, F. Bray, D. Forman, C. Mathers, D.M. Parkin, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008, International journal of cancer, 127 (2010) 28932917. [230] A. Jemal, R. Siegel, E. Ward, Y. Hao, J. Xu, T. Murray, M.J. Thun, Cancer statistics, 2008, CA: a cancer journal for clinicians, 58 (2008) 71-96. [231] L. Brannon-Peppas, J.O. Blanchette, Nanoparticle and targeted systems for cancer therapy, Advanced drug delivery reviews, 64 (2012) 206-212. [232] S.-S. Feng, Nanoparticles of biodegradable polymers for new-concept chemotherapy, 40

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

(2004). [233] X. Jia, L. Jia, Nanoparticles improve biological functions of phthalocyanine photosensitizers used for photodynamic therapy, Current drug metabolism, 13 (2012) 1119-1122. [234] J. Shao, Y. Dai, W. Zhao, J. Xie, J. Xue, J. Ye, L. Jia, Intracellular distribution and mechanisms of actions of photosensitizer Zinc (II)-phthalocyanine solubilized in Cremophor EL against human hepatocellular carcinoma HepG2 cells, Cancer letters, 330 (2013) 49-56. [235] Y. Gao, J. Xie, H. Chen, S. Gu, R. Zhao, J. Shao, L. Jia, Nanotechnology-based intelligent drug design for cancer metastasis treatment, Biotechnology advances, (2013). [236] X. Wang, L. Yang, Z.G. Chen, D.M. Shin, Application of nanotechnology in cancer therapy and imaging, CA: a cancer journal for clinicians, 58 (2008) 97-110. [237] O.C. Farokhzad, R. Langer, Impact of nanotechnology on drug delivery, ACS nano, 3 (2009) 16-20. [238] S. Biswas, V.P. Torchilin, Nanopreparations for organelle-specific delivery in cancer, Advanced drug delivery reviews, 66 (2014) 26-41. [239] J.K. Vasir, V. Labhasetwar, Targeted drug delivery in cancer therapy, Technology in cancer research & treatment, 4 (2005) 363-374. [240] V. Gupta, A. Aseh, C.N. Ríos, B.B. Aggarwal, A.B. Mathur, Fabrication and characterization of silk fibroin-derived curcumin nanoparticles for cancer therapy, International journal of nanomedicine, 4 (2009) 115. [241] X.-X. Xia, M. Wang, Y. Lin, Q. Xu, D.L. Kaplan, Hydrophobic Drug-Triggered SelfAssembly of Nanoparticles from Silk-Elastin-Like Protein Polymers for Drug Delivery, Biomacromolecules, 15 (2014) 908-914. [242] B. Subia, S. Chandra, S. Talukdar, S.C. Kundu, Folate conjugated silk fibroin nanocarriers for targeted drug delivery, Integrative Biology, 6 (2014) 203-214. [243] P. Wu, Q. Liu, R. Li, J. Wang, X. Zhen, G. Yue, H. Wang, F. Cui, F. Wu, M. Yang, Facile preparation of paclitaxel loaded silk fibroin nanoparticles for enhanced antitumor efficacy by locoregional drug delivery, ACS applied materials & interfaces, 5 (2013) 12638-12645. [244] M. Chen, Z. Shao, X. Chen, Paclitaxel‐loaded silk fibroin nanospheres, Journal of Biomedical Materials Research Part A, 100 (2012) 203-210. [245] N. Vadia, S. Rajput, Study on formulation variables of methotrexate loaded mesoporous MCM-41 nanoparticles for dissolution enhancement, European Journal of Pharmaceutical Sciences, 45 (2012) 8-18. [246] A. Taheri, R. Dinarvand, F.S. Nouri, M.R. Khorramizadeh, A.T. Borougeni, P. Mansoori, F. Atyabi, Use of biotin targeted methotrexate–human serum albumin conjugated nanoparticles to enhance methotrexate antitumor efficacy, International journal of nanomedicine, 6 (2011) 1863. [247] B. Subia, S. Kundu, Drug loading and release on tumor cells using silk fibroin–albumin nanoparticles as carriers, Nanotechnology, 24 (2013) 035103.

41

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

AC CE P

TE

D

Graphical abstract

42

Silk fibroin nanoparticle as a novel drug delivery system.

Design and synthesis of efficient drug delivery systems are of vital importance for medicine and healthcare. Nanocarrier-based drug delivery systems, ...
1MB Sizes 1 Downloads 12 Views