Open access peer-reviewed chapter

Silk Fibroin Nanoparticles: A Biocompatible Multi-Functional Polymer for Drug Delivery

Written By

Faith H.N. Howard, Zijian Gao, Hawari Bin Mansor, Zidi Yang and Munitta Muthana

Submitted: 06 October 2022 Reviewed: 21 December 2022 Published: 16 January 2023

DOI: 10.5772/intechopen.109642

From the Annual Volume

Biotechnology - Biosensors, Biomaterials and Tissue Engineering Annual Volume 2023

Edited by Luis Jesús Villarreal-Gómez

Chapter metrics overview

206 Chapter Downloads

View Full Metrics

Abstract

The versatility of nanomedicines allows for various modifications of material type, size, charge and functionalization, offering a promising platform for biomedical applications including tumor targeting. One such material, silk fibroin (SF) has emerged, displaying an excellent combination of mechanical and biological properties characterized by its high tensile and breaking strength, elongation, stiffness and ductility. High stability allows SF to maintain its chemical structure even at high temperatures (around 250°C) and compared with other biological polymers like polylactide (PLA), poly(lactic-co-glycolic acid) (PLGA), and collagen, SF shows excellent biocompatibility and lower immunogenic response making it a very suitable material for drug delivery and tissue engineering. Here we describe the structure, synthesis and properties of SF nanoparticles. We evaluate its emergence as a multi-functional polymer for its utility as a nanocarrier to deliver cancer therapies directly to tumors together with considerations for its clinical use.

Keywords

  • silk fibroin
  • polymer
  • nanomedicine
  • nanocarrier
  • drug delivery

1. Introduction

As stated by the World Health Organization, cancer is the second leading cause of death globally, accounting for an estimated 9.6 million deaths, or 1 in 6 deaths, in 2018 [1]. GLOBOCAN also predicts an increase in cancer rates, with more than 20 million new cancer cases expected annually by 2025 [2]. Over time, as our understanding and knowledge of molecular and tumor biology has increased, the cancer treatment paradigms have notably changed, particularly during the past 20 years. Previously, cancer was listed and treated based on its origin or its unique histomorphologic characteristics. However, in 2002, Schiller et al. reported that third generation chemotherapy administered to non-small-cell lung cancer showed almost similar survival curves [3]. Although the results are only limited to lung cancer, this indicates that cancer treatment using general (non-specific) cytotoxic chemotherapies have reached a therapeutic plateau. Since then, this research area has evolved based on two focus areas: tumor molecular profiling and molecular targets. Together, these efforts have further realized two recent revolutions in cancer research. Firstly, genotype-directed precision oncology which focuses on personalized therapies to treat specific genomic abnormalities regardless of the cancer type. Secondly, the targeting of particular components in the tumor microenvironment. As a result, the discovery of an abundance of anticancer drugs have showed a promising early step for cancer eradication. However, most of these therapeutics agents have undesirable characteristics, limiting their clinical usage and invalidating further drug research [4].

Recent studies have suggested that the use of nanoparticles as a drug carrier may be one of the best alternatives to improve the therapeutic effect of anticancer drugs. Nanotechnology, or in this case, nanomedicine, is the use of materials usually in nanometer scale in the fields of medicine and health [5]. Nanoparticle-based drug delivery systems have shown remarkable progress in overcoming the limitations of conventional drug delivery or drug therapy method. The unique characteristics that usually accompany these potential carriers include nanoscale size, high surface-to-volume ratio, auspicious physical and chemical properties and most importantly, endless possibilities for modifications that support cell targeting, gene delivery etc. [5].

Nanoparticles fall into two different categories, namely soft/organic and hard/inorganic nanoparticles. Soft nanoparticles are based on organic material, typically prepared from polymers or molecules that can self-assemble (coacervation) into large particles. The materials can range from full synthetic polymer to natural materials such as silk proteins [6]. Hard nanoparticles, on the other hand, are inorganic and usually insoluble, e.g. silver, gold nanoparticles, and carbon nanotubes [7, 8]. The benefits of organic nanoparticles as drug nanocarriers have been reported numerously in recent years, citing desirable characteristics including, biodegradability, non-antigenic and superior biocompatibility [9]. Thus, the delivery system’s advancement holds the promise of future precision medicine, which would greatly improve cancer survival rate by treating each cancer patient with the most effective drugs in the most efficient ways [10].

One such natural polymer which promises great potential as a drug delivery system is silk fibroin (SF). Silk has been recognized as a valuable natural material for the fabric industry for centuries, but during the last few decades, it has attracted immense attention as a promising biopolymer for biomedical and pharmaceutical applications [11]. Silk from the domesticated silkworm Bombyx mori (B. mori) is well characterized and has been approved as a safe biomaterial by the US Food and Drug Administration (FDA) [12]. This chapter focuses on the use of B. mori derived SF as a functional material for cancer drug delivery.

Advertisement

2. Silk fibroin

2.1 Structure

Most SF utilized for biomedical and commercial applications are derived from cocoons of B. mori domestic silk moths. Constructed from a continuous fiber strand comprised of two cores of fibroin protein held together by sericin protein [13, 14]. The primary structure of B. mori SF mainly consists of glycine (Gly) (43%), alanine (Ala) (30%) and serine (Ser) (12%) [15]. The secondary structure of SF exists in three different structural forms including silk I, silk II and silk III. Silk I consists of α-helix domains which is in a water-soluble state and easy to convert to silk II structure once treated with organic solvents, electromagnetic fields, or physical spinning environments. Unlike silk I, silk II contains an antiparallel β-sheet/crystal molecular model which has hydrogen side chains from glycine and methyl side chains from the alanines resulting in higher stability and both water and solvent insolubility. Silk III prevails at the water/air interface (Figure 1) [16, 17, 18].

Figure 1.

Cross-sectional and structural composition of silk fibers. Created with BioRender.

2.2 Properties

In the past centuries, silk has been used as a natural material for the fabric industry, but recently SF nanoparticles have been considered as a potential alternative carrier for anticancer drug delivery because of its physicochemical, mechanical, and biological properties [11]. Compared with other materials, SF is characterized by its high tensile and breaking strength, elongation, stiffness and ductility [19]. High stability allows maintenance of SF chemical structure even at high temperatures (around 250°C) [20]. In addition, compared with other biological polymers like polylactide (PLA), poly(lactic-co-glycolic acid) (PLGA), and collagen, silk fibroin shows excellent biocompatibility and lower immunogenic response [21, 22, 23] making it a very suitable material for drug delivery and tissue engineering. It is commonly used as a suture material, demonstrating comparable immunogenicity and biocompatibility to other natural and synthetic suture materials [24]. Moreover, it has consistently shown to be non-toxic and fully resorbable, this includes any degradation products [25]. Surprisingly, SF has been proved to have an intrinsic anti-inflammatory ability which could be used in the treatment of inflammatory bowel disease [26].

Degradability is another important property of silk fibroin. The degradation rate of SF is related to the molecular weight, the degree of crystallinity, morphological features and crosslinking [27]. This affords a tunability from seconds to years, another unique feature of SF. As a result of these properties, SF has been used in different nanosystems, including films, sponges, hydrogels, tubes [28]. Here, we focus on silk nanoparticles.

Advertisement

3. Silk nanoparticles

3.1 Synthesis of silk nanoparticles

Silk nanoparticles are an excellent candidate as a carrier for drugs, targeted therapies and contrast agents. They are synthesized from regenerated SF through a variety of methods based on their self-assembly behavior. Among all of the approaches, desolvation (Figure 2) is the most common method for SF nanoparticle synthesis, in which dissolving agents including ethanol, acetone, dimethyl sulfoxide (DMSO) and methanol could be used to dehydrate and package the silk chain, leading to the change from silk I to silk II structure and forming SF nanoparticles [29]. During the desolvation method, lipophilic active drugs (e.g. curcumin and 5-fluorouracil) can be easily dissolved in the organic solvents allowing nanoencapsulation of anticancer drugs [30]. The main challenge of the desolvation method is to find the ideal SF/dissolving agent ratio and ensure adequate mixing during SF nanoparticle formation which plays an important role in the control of the nanoparticle size.

Figure 2.

Representation of the desolvation and salting-out method for the production of silk fibroin nanoparticles. Created with BioRender.

Salting-out (Figure 2) is another widely used method in the preparation of protein-based nanoparticles by removing the water barriers between protein molecules and increasing the interactions between proteins, leading to aggregation and precipitation from the solution [31]. The salt, pH, and ionic strength are major factors influencing the yield, particle morphology, zeta potential, and nanoparticle stability [30]. The salting-out method has advantages in avoiding the usage of toxic solvents, therefore maintaining the activity of the protein. However, the synthesized particle size is relatively large (500–2000 nm), and the high amounts of salts are difficult to remove [29].

The electrospraying method uses electrical forces to obtain liquid atomization where the liquid flowing out of a capillary nozzle is dispersed into small droplets by an electric field [32, 33]. Similar to the desolvation method, the main limitation of electrospraying is the organic solvents used during the synthesis that may damage the bioactivity of their cargo (e.g. enzymes, genes, and cell vitality) [33]. Other silk nanoparticle synthesis methods which are not used widely used include supercritical fluid technologies [34], mechanical comminution [35], capillary-microdot technique [36], and microemulsion [37].

3.2 Functionalization

Many drug delivery systems lack a targeting mechanism, resulting in poor accumulation at tumor targets. To achieve efficacious concentrations at the tumor, saturating drug doses are often administered which can lead to non-specific or toxic effects. Nanoparticles have been designed to overcome this problem by delivering the drug to specific tissue instead of a more generalized treatment. Currently, four different targeting mechanism have been explored; passive targeting, targeted recognition, triggered release and guided delivery (Figure 3). Historically, the enhanced permeability and retention (EPR) effect (a unique phenomenon of solid tumors) has been utilized whereby nanoparticles can extravasate through the leaky blood vessels of the tumor tissue without the need for surface modification [5, 38, 39]. Some surface modification that affects circulation time may also indirectly affect passive targeting such as PEGylation of the nanoparticles. This is due to EPR effect increases proportional to the circulation time [40]. However, some tissues may also contain fenestrated blood vessels resulting in a similar effect on nanoparticles, causing them to accumulate there [5]. Additionally, the tumor microenvironment varies depending on the tumor type and passive targeting may not be as efficient in that particular condition.

Figure 3.

Schematic of different drug targeting approaches for nanoparticles. Created with BioRender.

Targeted recognition involves the use of targeting molecules as an attachment to the drug-loaded nanoparticles. Targeting molecules such as ligands have high specificity to receptors and other cancer-specific target molecules available on the surface of cancerous tissue such as glycans [4]. Conjugation of nanoparticles with ligands such as transferrin, folic acid, enzymes, antibodies and other macromolecules has demonstrated enhanced uptake of nanoparticles by cancer cells [5, 39]. However, they still rely on passive accumulation at the tumor site.

Traditional cancer treatment methods suffer from a lack of specific regional and temporal activation leading to off-target effects. The concept of smart nanosystems uses the intrinsic and environmental differences between normal cells and cancer cells to trigger activation or release of drugs at a tumor site [41]. Intrinsic activation strategies include using the differences in pH, enzyme expression level, the concentration of membrane proteins and soluble molecules between healthy cells and cancer cells to trigger drug release at a specific location in the body [5, 41, 42, 43]. Extrinsic activation includes using ultrasound, magnetic field, light and photodynamic therapy to provide an activatable system with less toxic, safe, and minimal adverse effects [44, 45, 46]. At the moment, the development of nanoparticles activation strategy is not only satisfied with a single treatment but also presume a multimodal trigger system. Therefore, the synthesis and analysis of multifunctional nanomaterials have been extensively researched in vitro and in vivo for cancer [47, 48].

The above strategies, however, lack a navigational force to the desired target as well as the ability to penetrate tumors beyond diffusion limits. Magnetic tumor targeting, using magnetic carriers and an external magnetic field is demonstrating promise for enhanced tumor accumulation of chemotherapy [49] and virotherapy [50] following their systemic administration. In the presence of a magnetic field, peptide-functionalized magnetic silk nanoparticles demonstrated increased cellular uptake of an anticancer agent (ASC-J9) by HCT116 colorectal cancer spheroids [51]. Additionally, in an orthotopic model of breast cancer, magnetic targeting enriched Doxorubicin-loaded magnetic SF nanoparticles at the tumor site with a concomitant suppression of uptake by the liver, resulting in a significant decrease in tumor volume and survival [52]. The provision of an external driving force expands therapeutic use to a wide variety of tumors, independent of specific receptor expression.

3.3 Nanocarrier for cancer

3.3.1 Chemotherapy delivery

Due to excellent stability in the change of temperature [53], humidity [54], and pH [55], silk-based nanocarriers have been widely studied for the delivery of numerous chemotherapeutic substances such as doxorubicin, cisplatin, paclitaxel, 5′-fluorouracil, and floxuridine for cancer treatment (Table 1). In addition, silk nanoparticles can also be used in the delivery of natural plant-derived therapeutics including curcumin [65], celastrol and triptolide [70], which are limited by their poor water solubility. To reduce systemic toxicity and adverse side effects of chemotherapeutics, targeted delivery can be achieved by conjugation of various targeting ligands to the silk material (as described above), recognizing overexpression of particular epitopes on the surface of target cells [71]. For example, Lei Huang et al. [63] designed a folate (FA) conjugated silk nanoparticle double loaded with doxorubicin (FA-SFPs-DOX-DOX), which provided a pH-dependent targeted drug release, lasting for over 30 hours. This study demonstrated the importance of the target ligand, with FA-SFPs-DOX-DOX inducing greater cytotoxicity against HeLa cells compared with SFPs-DOX-DOX.

Therapeutic agentSilk sourceFunctionalizationCancer TypeIn vitro modelIn vivo modelReference
DoxorubicinSilk ProteinRGD peptideBreast4 T14 T1 (F)[56]
Silk sericinBreast4 T1[57]
B. moriBreast4 T14 T1 (F)[58]
B. moriBreast, colon4 T1, MCF7, Caco2-BBE4 T1 (OT)[59]
Spider silkHerceptinBreastD2F2, D2F2E2D2F2, D2F2E2 (OT)[60]
RSFHyaluronic acidLungA459A459 (X)[61]
B. moriBreast4 T14 T1 (F)[62]
B. moriFolateKB, C2C12[63]
CisplatinB. moriLungA549[64]
CurcuminB. moriBreastMDA-MB-231[65]
Curcumin/ 5-FUB. moriHyaluronic acidBreast4 T14 T1 (F)[66]
B. moriBreast4 T14 T1 (OT)[30]
5-FURSFColorectalHT-29[67]
PaclitaxelB. moriLiverH22H22 (F)[68]
B. moriGastricBGC-823, SGC-7901BGC-823 (X, F)[69]

Table 1.

Silk nanoparticles as nanocarriers for chemotherapy.

RSF = regenerated silk fibroin, 5-FU = 5-fluorouracil, F = flank, OT = orthotopic, and X = xenograft. All in vivo models performed in mice.

3.3.2 Peptide and protein delivery

Silk-based nanocarriers can also bind with peptides and proteins improving their in vivo stability. Lactoferrin is one such protein showing anti-cancer properties, whereby apo-bovine lactoferrin loaded silk nanoparticles induces significantly higher internalization and cytotoxicity towards the MDA-MB-231 and MCF-7 breast cancer cell lines [72]. Peptide-based cancer vaccines are another important therapeutic agent in cancer treatment. However, peptide vaccines suffer from short in vivo stability caused by proteolytic degradation and rapid clearance from the bloodstream [73]. A silk nanoparticle delivery system is an effective way to improve the bioavailability and stability of peptide tumor vaccines [74]. Using engineered spider silk nanoparticles a peptide-based vaccination resulted in successful activation of cytotoxic T-cells, without unspecific immune responses [75]. How these antigens are delivered can also influence the developing vaccination response. It is thought that controlled, persistent antigenic signals elicit stronger responses than transient bolus vaccine exposure [76, 77, 78], such as that seen with microneedle skin patches. Microneedle vaccines exploit the skin’s accessibility, both in terms of ease of administration as well as access to densely populated areas of antigen presenting cells. Silk microneedles therefore represent an attractive prospect due to their tunable release kinetics of encapsulated cargo as well as their overall biodegradability. This system demonstrated a > 10-fold increase in ovalbumin (OVA)-specific T cell and humoral responses in C57/Bl6 mice when compared with parenteral immunization [79], warranting further investigation.

3.3.3 Gene delivery

Viral vectors are traditional carriers for gene delivery, however, their drawbacks in inducing high systemic toxicity and immune responses limit their application in cancer treatment [22]. Thus, non-viral vectors have emerged to address challenges surrounding improving transfection efficiency, target specificity and cytotoxicity [80]. Among various materials, silk-based nanocarriers have been reported to provide biodegradability, biocompatibility, high transfection efficiency, and DNase resistance in gene delivery [9]. Through genetic engineering, the transfection efficiency of silk nanoparticles could be further improved. Numata et al. [81] combined silk protein-based nanocarriers with poly(L-lysine) (PLL) for gene delivery, resulting in improved transfection efficiency of pDNA in human embryonic kidney (HEK) cells. Additionally, to further enhance target specificity of the silk-based gene delivery system, they included tumor homing peptides (THP) [80]; F3 peptide (specifically targeted towards nucleolin expressing tumor and endothelial cells) and Lyp1 peptide (shows target specificity towards the p32 receptor overexpressed in tumor cells) [82, 83]. The use of cationic polymers with silk-based nanocarriers is another popular strategy due to their high cellular uptake efficiency, good water solubility, excellent transferability and easy synthesis [84]. Polyethyleneimine (PEI) is one of the commonly used cationic polymers which easily assembles with gene therapies and demonstrates improved cellular uptake due to their positive charge [85]. Song et al. [65] designed magnetic-silk/PEI core-shell nanoparticles for targeted delivery of c-myc antisense oligodeoxynucleotides (ODNs), which had high uptake efficiencies and significantly inhibit the growth of MDA-MB-231 cells.

3.3.4 Diagnostics and theranostics

In addition to therapeutic delivery, silk nanoparticles are also promising as non-invasive imaging components and provide an opportunity to augment existing imaging modalities for diagnostic purposes. These modalities are often limited by inadequate contrast between healthy and diseased tissue, contributing to failure to detect signs of illness, particularly early signs. Silk nanoparticles can be used as a vehicle for loading magnetic resonance contrast agents, overcoming agglomeration limitations of magnesium oxide nanospheres [86]. Alternatively the use of fluorescent dyes and carbon dots for modification of the silk fibroin itself has applications for live cell imaging or visualizing degradation of silk-fibroin implants [87]. The production of fluorescent silk nanoparticles can be created using simple dyes, chemical modification of the fibroin, conjugation or entrapment of fluorescent proteins and even doping the silkworm larvae’s diet with fluorescent dyes such as rhodamine and fluorescein [88]. Additionally, carbon quantum dots (CQDs) generated from SF are strongly fluorescent, resist photobleaching, can be further functionalized [89, 90, 91] and in comparison to other colloidal materials, avoid the need for toxic heavy metals. However, this process does require controlled and pressurized heating of the fibroin for carbonization into CQDs.

By combining these imaging modalities with their role as a drug carrier, silk nanoparticles are becoming an important theranostic device. Theranostics is an approach that combines cancer treatment and diagnosis, in which efficient imaging guidance of therapy is necessary for detecting the drug loading, targeted delivery, and release123. For example, Levodopa (a PTT agent) and manganese dioxide particles (a contrast agent) were formulated with silk sericin from B. mori cocoons to create a one-step method for MRI-guided photothermal therapy [92]. The composition of spheres made of spider silk and iron oxide nanoparticles have also demonstrated drug loading and release capacity with potential to be used in both hyperthermia and magnetic resonance imaging (MRI) applications combined with drug delivery against tumor cells [93].

Advertisement

4. Conclusion

Nanoparticles used for drug delivery require desired physicochemical properties including size [94], shape [95], structure [96], rigidity [97], and surface modification [98]. Translation and application of nanoparticles, including silk, to the clinic must first overcome a number of challenges including their heterogeneity, reproducibility and upscale production. Silk derived from different sources will possess different amino acid sequences and morphology, whilst LPS contamination of recombinant silk is a major obstacle for progression to clinic, requiring careful characterization of its toxicity and immunogenicity. Additionally, traditional nanoparticle preparation methods involving breaking down of large particles, nanoprecipitation, or self-assembly of monomers, suffer from wide size distribution and large batch-to-batch variability [99]. To obtain more stable and controllable nanoparticles, microfluidics has emerged for manipulating tiny fluids (1 × 10−9 L–1× 10−18 L) in micro-channels with dimensions of tens of micrometers [100]. Several flow patterns including laminar flow, turbulent flow and droplet flow could be achieved under microfluidic control with potential to enhance fluid mixing, reduce reagent consumption and batch-to-batch variations [101, 102]. Interestingly, the introduction of superparamagnetic magnetic nanoparticles (used to provide magnetic targeting capabilities) during the SF formation process provided artificial regulation of this process as well as drug entrapment, preventing agglomeration of SF and resulting in uniform, spherical nanoparticles [52]. Ultimately, SF nanoparticles provide many attractive properties for multi-functional drug delivery strategies but future use relies on reliable, reproducible manufacture to ensure appropriate comparisons can be made for their translation.

Advertisement

Acknowledgments

Financial support provided by Cancer Research UK (CRUK grant reference: C25574/A24321) and Public Service Department Malaysia (JPA).

References

  1. 1. WHO. Cancer: Overview. 3 Feb 2022. Available from: https://www.who.int/health-topics/cancer
  2. 2. GLOBOCAN. Global Cancer Observatory. 2022. Available from: https://gco.iarc.fr/
  3. 3. Schiller JH, Harrington D, Belani CP,Langer C, Sandler A, Krook J, et al. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. The New England Journal of Medicine. 2002;346(2):92-98
  4. 4. Cho K, Wang X, Nie S, Chen ZG, Shin DM. Therapeutic nanoparticles for drug delivery in cancer. Clinical Cancer Research. 2008;14(5):1310-1316
  5. 5. Wicki A, Witzigmann D, Balasubramanian V, Huwyler J. Nanomedicine in cancer therapy: Challenges, opportunities, and clinical applications. Journal of Controlled Release. 2015;200:138-157
  6. 6. Young SWS, Stenzel MH, Yang J-L. Nanoparticle-siRNA: A potential cancer therapy? Critical Reviews in Oncology/Hematology. 2016;98:159-169
  7. 7. Tomalia DA. In quest of a systematic framework for unifying and defining nanoscience. Journal of Nanoparticle Research. 2009;11(6):1251-1310
  8. 8. Kim T, Hyeon T. Applications of inorganic nanoparticles as therapeutic agents. Nanotechnology. 2014;25(1):012001
  9. 9. Zhao Z, Li Y, Xie MB. Silk fibroin-based nanoparticles for drug delivery. International Journal of Molecular Sciences. 2015;16(3):4880-4903
  10. 10. Bernards R. A missing link in genotype-directed cancer therapy. Cell. 2012;151(3):465-468
  11. 11. Wenk E, Merkle HP, Meinel L. Silk fibroin as a vehicle for drug delivery applications. Journal of Controlled Release. 2011;150(2):128-141
  12. 12. Kundu B, Rajkhowa R, Kundu SC, Wang X. Silk fibroin biomaterials for tissue regenerations. Advanced Drug Delivery Reviews. 2013;65(4):457-470
  13. 13. Cao Y, Wang B. Biodegradation of silk biomaterials. International Journal of Molecular Sciences. 2009;10(4):1514-1524
  14. 14. Guidetti G, d’Amone L, Kim T, Matzeu G, Mogas-Soldevila L, Napier B, et al. Silk materials at the convergence of science, sustainability, healthcare, and technology. Applied Physics Reviews. 2022;9(1):50
  15. 15. Kaplan DL, Mello CM, Arcidiacono S, Fossey S, Senecal K, Muller W. Silk. In: McGrath K, Kaplan D, editors. Protein-Based Materials. Boston, MA: Birkhäuser Boston; 1997. pp. 103-131
  16. 16. Jin HJ, Kaplan DL. Mechanism of silk processing in insects and spiders. Nature. 2003;424(6952):1057-1061
  17. 17. Matsumoto A, Lindsay A, Abedian B, Kaplan DL. Silk fibroin solution properties related to assembly and structure. Macromolecular Bioscience. 2008;8(11):1006-1018
  18. 18. Chen X, Shao Z, Knight DP, Vollrath F. Conformation transition kinetics of Bombyx mori silk protein. Proteins: Structure, Function, and Bioinformatics. 2007;68(1):223-231
  19. 19. Zhang C, Song D, Lu Q, Hu X, Kaplan DL, Zhu H. Flexibility regeneration of silk fibroin in vitro. Biomacromolecules. 2012;13(7):2148-2153
  20. 20. Lu Q, Hu X, Wang X, Kluge JA, Lu S, Cebe P, et al. Water-insoluble silk films with silk I structure. Acta Biomaterialia. 2010;6(4):1380-1387
  21. 21. Yucel T, Lovett ML, Kaplan DL. Silk-based biomaterials for sustained drug delivery. Journal of Controlled Release. 2014;190:381-397
  22. 22. Numata K, Hamasaki J, Subramanian B, Kaplan DL. Gene delivery mediated by recombinant silk proteins containing cationic and cell binding motifs. Journal of Controlled Release. 2010;146(1):136-143
  23. 23. Meinel L, Karageorgiou V, Hofmann S, Fajardo R, Snyder B, Li C, et al. Engineering bone-like tissue in vitro using human bone marrow stem cells and silk scaffolds. Journal of Biomedical Materials Research Part A. 2004;71A(1):25-34
  24. 24. Patil PP, Reagan MR, Bohara RA. Silk fibroin and silk-based biomaterial derivatives for ideal wound dressings. International Journal of Biological Macromolecules. 2020;164:4613-4627
  25. 25. Kim DH, Kim YS, Amsden J, Panilaitis B, Kaplan DL, Omenetto FG, et al. Silicon electronics on silk as a path to bioresorbable, implantable devices. Applied Physics Letters. 2009;95(13):133701
  26. 26. Lozano-Perez AA, Rodriguez-Nogales A, Ortiz-Cullera V, Algieri F, Garrido-Mesa J, Zorrilla P, et al. Silk fibroin nanoparticles constitute a vector for controlled release of resveratrol in an experimental model of inflammatory bowel disease in rats. International Journal of Nanomedicine. 2014;9:4507-4520
  27. 27. You R, Zhang Y, Liu Y, Liu G, Li M. The degradation behavior of silk fibroin derived from different ionic liquid solvents. Natural Science. 2013;5:10-19
  28. 28. Crivelli B, Perteghella S, Bari E, Sorrenti M, Tripodo G, Chlapanidas T, et al. Silk nanoparticles: From inert supports to bioactive natural carriers for drug delivery. Soft Matter. 2018;14(4):546-557
  29. 29. Zhang Y-Q, Shen W-D, Xiang R-L, Zhuge L-J, Gao W-J, Wang W-B. Formation of silk fibroin nanoparticles in water-miscible organic solvent and their characterization. Journal of Nanoparticle Research. 2007;9(5):885-900
  30. 30. Li H, Tian J, Wu A, Wang J, Ge C, Sun Z. Self-assembled silk fibroin nanoparticles loaded with binary drugs in the treatment of breast carcinoma. International Journal of Nanomedicine. 2016;11:4373-4380
  31. 31. Lammel AS, Hu X, Park SH, Kaplan DL, Scheibel TR. Controlling silk fibroin particle features for drug delivery. Biomaterials. 2010;31(16):4583-4591
  32. 32. Anatol J, Arkadiusz TS. Electrospraying route to nanotechnology: An overview. Journal of Electrostatics. 2008;66:197-219
  33. 33. Bock N, Woodruff MA, Hutmacher DW, Dargaville TR. Electrospraying, a reproducible method for production of polymeric microspheres for biomedical applications. Polymers. 2011;3(1):131-149
  34. 34. Zhao Z, Li Y, Chen A-Z, Zheng Z-J, Hu J-Y, Li J-S, et al. Generation of silk fibroin nanoparticles via solution-enhanced dispersion by supercritical CO2. Industrial & Engineering Chemistry Research. 2013;52(10):3752-3761
  35. 35. Grbavčić ŽB, Arsenijević ZL, Garić-Grulović RV. Prediction of single particle settling velocities through liquid fluidized beds. Powder Technology. 2009;190(3):283-291
  36. 36. Gupta V, Aseh A, Ríos CN, Aggarwal BB, Mathur AB. Fabrication and characterization of silk fibroin-derived curcumin nanoparticles for cancer therapy. International Journal of Nanomedicine. 2009;4:115-122
  37. 37. Myung SJ, Kim H-S, Kim Y, Chen P, Jin H-J. Fluorescent silk fibroin nanoparticles prepared using a reverse microemulsion. Macromolecular Research. 2008;16(7):604-608
  38. 38. Albanese A, Tang PS, Chan WC. The effect of nanoparticle size, shape, and surface chemistry on biological systems. Annual Review of Biomedical Engineering. 2012;14:1-16
  39. 39. Bregoli L, Movia D, Gavigan-Imedio JD, Lysaght J, Reynolds J, Prina-Mello A. Nanomedicine applied to translational oncology: A future perspective on cancer treatment. Nanomedicine. 2016;12(1):81-103
  40. 40. van Vlerken LE, Vyas TK, Amiji MM. Poly(ethylene glycol)-modified nanocarriers for tumor-targeted and intracellular delivery. Pharmaceutical Research. 2007;24(8):1405-1414
  41. 41. White BD, Duan C, Townley HE. Nanoparticle activation methods in cancer treatment. Biomolecules. 2019;9(5):26
  42. 42. Boron WF. Regulation of intracellular pH. Advances in Physiology Education. 2004;28(4):160-179
  43. 43. Li X, Kim J, Yoon J, Chen X. Cancer-associated, stimuli-driven, turn on Theranostics for multimodality imaging and therapy. Advanced Materials. 2017;29(23):1606857
  44. 44. Mullin LB, Phillips LC, Dayton PA. Nanoparticle delivery enhancement with acoustically activated microbubbles. IEEE Transactions on Ultrasonics, Ferroelectrics, and Frequency Control. 2013;60(1):65-77
  45. 45. Pearce J, Giustini A, Stigliano R, Jack HP. Magnetic heating of nanoparticles: The importance of particle clustering to achieve therapeutic temperatures. Journal of Nanotechnology in Engineering and Medicine. 2013;4(1):110071-1100714
  46. 46. Huang L, Li Z, Zhao Y, Zhang Y, Wu S, Zhao J, et al. Ultralow-power near infrared lamp light operable targeted organic nanoparticle photodynamic therapy. Journal of the American Chemical Society. 2016;138(44):14586-14591
  47. 47. Chen KJ, Liang HF, Chen HL, Wang Y, Cheng PY, Liu HL, et al. A thermoresponsive bubble-generating liposomal system for triggering localized extracellular drug delivery. ACS Nano. 2013;7(1):438-446
  48. 48. Guduru R, Liang P, Runowicz C, Nair M, Atluri V, Khizroev S. Magneto-electric nanoparticles to enable field-controlled high-specificity drug delivery to eradicate ovarian cancer cells. Scientific Reports. 2013;3:2953
  49. 49. Lu YJ, Lin PY, Huang PH, Kuo CY, Shalumon KT, Chen MY, et al. Magnetic graphene oxide for dual targeted delivery of doxorubicin and photothermal therapy. Nanomaterials (Basel). 2018;8(4):20
  50. 50. Howard FHN, Al-Janabi H, Patel P, Cox K, Smith E, Vadakekolathu J, et al. Nanobugs as drugs: Bacterial derived Nanomagnets enhance tumor targeting and oncolytic activity of HSV-1 virus. Small. 2022;18(13):e2104763
  51. 51. Tomeh MA, Hadianamrei R, Xu D, Brown S, Zhao X. Peptide-functionalised magnetic silk nanoparticles produced by a swirl mixer for enhanced anticancer activity of ASC-J9. Colloids and Surfaces B: Biointerfaces. 2022;216:112549
  52. 52. Tian Y, Jiang X, Chen X, Shao Z, Yang W. Doxorubicin-loaded magnetic silk fibroin nanoparticles for targeted therapy of multidrug-resistant cancer. Advanced Materials. 2014;26(43):7393-7398
  53. 53. Liu Y, Zheng Z, Gong H, Liu M, Guo S, Li G, et al. DNA preservation in silk. Biomaterials Science. 2017;5(7):1279-1292
  54. 54. X-m Z, Berghe IV, Wyeth P. Heat and moisture promoted deterioration of raw silk estimated by amino acid analysis. Journal of Cultural Heritage. 2011;12:408-411
  55. 55. Zong X-H, Zhou P, Shao Z-Z, Chen S-M, Chen X, Hu B-W, et al. Effect of pH and copper(II) on the conformation transitions of silk fibroin based on EPR, NMR, and Raman spectroscopy. Biochemistry. 2004;43(38):11932-11941
  56. 56. Bin L, Yang Y, Wang F, Wang R, Fei H, Duan S, et al. Biodegradable silk fibroin Nanocarriers to modulate hypoxia tumor microenvironment favoring enhanced chemotherapy. Frontiers in Bioengineering and Biotechnology. 2022;10:960501
  57. 57. Li X, Hou S, Chen J, He CE, Gao YE, Lu Y, et al. Engineering silk sericin decorated zeolitic imidazolate framework-8 nanoplatform to enhance chemotherapy. Colloids and Surfaces. B, Biointerfaces. 2021;200:111594
  58. 58. Norouzi P, Motasadizadeh H, Atyabi F, Dinarvand R, Gholami M, Farokhi M, et al. Combination therapy of breast cancer by Codelivery of doxorubicin and Survivin siRNA using Polyethylenimine modified silk fibroin nanoparticles. ACS Biomaterials Science & Engineering. 2021;7(3):1074-1087
  59. 59. Chen Q, Ma Y, Bai P, Li Q, Canup BSB, Long D, et al. Tumor microenvironment-responsive Nanococktails for synergistic enhancement of cancer treatment via Cascade reactions. ACS Applied Materials & Interfaces. 2021;13(4):4861-4873
  60. 60. Florczak A, Deptuch T, Lewandowska A, Penderecka K, Kramer E, Marszalek A, et al. Functionalized silk spheres selectively and effectively deliver a cytotoxic drug to targeted cancer cells in vivo. Journal of Nanobiotechnology. 2020;18(1):177
  61. 61. Gou S, Yang J, Ma Y, Zhang X, Zu M, Kang T, et al. Multi-responsive nanococktails with programmable targeting capacity for imaging-guided mitochondrial phototherapy combined with chemotherapy. Journal of Controlled Release. 2020;327:371-383
  62. 62. Tan M, Liu W, Liu F, Zhang W, Gao H, Cheng J, et al. Silk fibroin-coated Nanoagents for acidic lysosome targeting by a functional preservation strategy in cancer chemotherapy. Theranostics. 2019;9(4):961-973
  63. 63. Huang L, Tao K, Liu J, Qi C, Xu L, Chang P, et al. Design and fabrication of multifunctional Sericin nanoparticles for tumor targeting and pH-responsive subcellular delivery of cancer chemotherapy drugs. ACS Applied Materials & Interfaces. 2016;8(10):6577-6585
  64. 64. Kim SY, Naskar D, Kundu SC, Bishop DP, Doble PA, Boddy AV, et al. Formulation of biologically-inspired silk-based drug carriers for pulmonary delivery targeted for lung cancer. Scientific Reports. 2015;5(1):11878
  65. 65. Song W, Muthana M, Mukherjee J, Falconer RJ, Biggs CA, Zhao X. Magnetic-silk Core–Shell nanoparticles as potential carriers for targeted delivery of curcumin into human breast cancer cells. ACS Biomaterials Science & Engineering. 2017;3(6):1027-1038
  66. 66. Huang Y, Xie D, Gou S, Canup BSB, Zhang G, Dai F, et al. Quadruple-responsive nanoparticle-mediated targeted combination chemotherapy for metastatic breast cancer. Nanoscale. 2021;13(11):5765-5779
  67. 67. Hudita A, Radu IC, Galateanu B, Ginghina O, Herman H, Balta C, et al. Bioinspired silk fibroin nano-delivery systems protect against 5-FU induced gastrointestinal mucositis in a mouse model and display antitumor effects on HT-29 colorectal cancer cells in vitro. Nanotoxicology. 2021;15(7):973-994
  68. 68. Wu P, Liu Q, Wang Q, Qian H, Yu L, Liu B, et al. Novel silk fibroin nanoparticles incorporated silk fibroin hydrogel for inhibition of cancer stem cells and tumor growth. International Journal of Nanomedicine. 2018;13:5405-5418
  69. 69. Wu P, Liu Q, Li R, Wang J, Zhen X, Yue G, et al. Facile preparation of paclitaxel loaded silk fibroin nanoparticles for enhanced antitumor efficacy by Locoregional drug delivery. ACS Applied Materials & Interfaces. 2013;5(23):12638-12645
  70. 70. Chen S-R, Dai Y, Zhao J, Lin L, Wang Y, Wang Y. A mechanistic overview of Triptolide and Celastrol, natural products from Tripterygium wilfordii hook F. Frontiers in Pharmacology. 2018;9:13
  71. 71. Gobin AS, Butler CE, Mathur AB. Repair and regeneration of the abdominal wall musculofascial defect using silk fibroin-chitosan blend. Tissue Engineering. 2006;12(12):3383-3394
  72. 72. Roy K, Patel YS, Kanwar RK, Rajkhowa R, Wang X, Kanwar JR. Biodegradable Eri silk nanoparticles as a delivery vehicle for bovine lactoferrin against MDA-MB-231 and MCF-7 breast cancer cells. International Journal of Nanomedicine. 2016;11:25-44
  73. 73. Malonis RJ, Lai JR, Vergnolle O. Peptide-based vaccines: Current Progress and future challenges. Chemical Reviews. 2020;120(6):3210-3229
  74. 74. Oroojalian F, Beygi M, Baradaran B, Mokhtarzadeh A, Shahbazi M-A. Immune cell membrane-coated biomimetic nanoparticles for targeted cancer therapy. Small. 2021;17(12):2006484
  75. 75. Lucke M, Mottas I, Herbst T, Hotz C, Römer L, Schierling M, et al. Engineered hybrid spider silk particles as delivery system for peptide vaccines. Biomaterials. 2018;172:105-115
  76. 76. Mohanan D, Slütter B, Henriksen-Lacey M, Jiskoot W, Bouwstra JA, Perrie Y, et al. Administration routes affect the quality of immune responses: A cross-sectional evaluation of particulate antigen-delivery systems. Journal of Controlled Release. 2010;147(3):342-349
  77. 77. Jewell CM, López SC, Irvine DJ. In situ engineering of the lymph node microenvironment via intranodal injection of adjuvant-releasing polymer particles. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(38):15745-15750
  78. 78. Johansen P, Storni T, Rettig L, Qiu Z, Der-Sarkissian A, Smith KA, et al. Antigen kinetics determines immune reactivity. Proceedings of the National Academy of Sciences. 2008;105:5189-5194
  79. 79. DeMuth PC, Min Y, Irvine DJ, Hammond PT. Implantable silk composite microneedles for programmable vaccine release kinetics and enhanced immunogenicity in transcutaneous immunization. Advanced Healthcare Materials. 2014;3(1):47-58
  80. 80. Numata K, Mieszawska-Czajkowska AJ, Kvenvold LA, Kaplan DL. Silk-based Nanocomplexes with tumor-homing peptides for tumor-specific gene delivery. Macromolecular Bioscience. 2012;12(1):75-82
  81. 81. Numata K, Subramanian B, Currie HA, Kaplan DL. Bioengineered silk protein-based gene delivery systems. Biomaterials. 2009;30(29):5775-5784
  82. 82. Christian S, Pilch J, Akerman ME, Porkka K, Laakkonen P, Ruoslahti E. Nucleolin expressed at the cell surface is a marker of endothelial cells in angiogenic blood vessels. The Journal of Cell Biology. 2003;163(4):871-878
  83. 83. Song N, Zhao L, Zhu M, Zhao J. Recent progress in LyP-1-based strategies for targeted imaging and therapy. Drug Delivery. 2019;26(1):363-375
  84. 84. Zakeri A, Kouhbanani MAJ, Beheshtkhoo N, Beigi V, Mousavi SM, Hashemi SAR, et al. Polyethylenimine-based nanocarriers in co-delivery of drug and gene: A developing horizon. Nano Reviews & Experiments. 2018;9(1):1488497
  85. 85. Chen Z, Krishnamachary B, Bhujwalla ZM. Degradable dextran Nanopolymer as a carrier for choline kinase (ChoK) siRNA cancer therapy. Nanomaterials (Basel). 2016;6(2):8
  86. 86. Li J, Khalid A, Verma R, Abraham A, Qazi F, Dong X, et al. Silk fibroin coated magnesium oxide Nanospheres: A biocompatible and biodegradable tool for noninvasive bioimaging applications. Nanomaterials (Basel). 2021;11(3):20
  87. 87. Kim JH, Park CH, Lee OJ, Lee JM, Kim JW, Park YH, et al. Preparation and in vivo degradation of controlled biodegradability of electrospun silk fibroin nanofiber mats. Journal of Biomedical Materials Research. Part A. 2012;100(12):3287-3295
  88. 88. Sagnella A, Chieco C, Di Virgilio N, Toffanin S, Posati T, Pistone A, et al. Bio-doping of regenerated silk fibroin solution and films: A green route for biomanufacturing. RSC Advances. 2014;4(64):33687-33694
  89. 89. Colusso E, Cicerchia L, Rigon M, Gomes V, Martucci A. Photoluminescence properties of silk–carbon quantum dots composites. Journal of Sol-Gel Science and Technology. 2022:8
  90. 90. Horo H, Saha M, Das H, Mandal B, Kundu LM. Synthesis of highly fluorescent, amine-functionalized carbon dots from biotin-modified chitosan and silk-fibroin blend for target-specific delivery of antitumor agents. Carbohydrate Polymers. 2022;277:118862
  91. 91. Niu L, Shi M, Feng Y, Sun X, Wang Y, Cheng Z, et al. The interactions of quantum dot-labeled silk fibroin micro/nanoparticles with cells. Materials (Basel). 2020;13(15):17
  92. 92. Tian D, Xu H, Xiao B, Zhou X, Liu X, Zhou Z, et al. Single-step formulation of levodopa-based nanotheranostics—Strategy for ultra-sensitive high longitudinal relaxivity MRI guided switchable therapeutics. Biomaterials Science. 2020;8(6):1615-1621
  93. 93. Kucharczyk K, Rybka JD, Hilgendorff M, Krupinski M, Slachcinski M, Mackiewicz A, et al. Composite spheres made of bioengineered spider silk and iron oxide nanoparticles for theranostics applications. PLoS One. 2019;14(7):e0219790
  94. 94. Wang J, Mao W, Lock LL, Tang J, Sui M, Sun W, et al. The role of micelle size in tumor accumulation, penetration, and treatment. ACS Nano. 2015;9(7):7195-7206
  95. 95. Kinnear C, Moore TL, Rodriguez-Lorenzo L, Rothen-Rutishauser B, Petri-Fink A. Form follows function: Nanoparticle shape and its implications for nanomedicine. Chemical Reviews. 2017;117(17):11476-11521
  96. 96. Zhang L, Feng Q, Wang J, Zhang S, Ding B, Wei Y, et al. Microfluidic synthesis of hybrid nanoparticles with controlled lipid layers: Understanding flexibility-regulated cell-nanoparticle interaction. ACS Nano. 2015;9(10):9912-9921
  97. 97. Hui Y, Yi X, Hou F, Wibowo D, Zhang F, Zhao D, et al. Role of nanoparticle mechanical properties in cancer drug delivery. ACS Nano. 2019;13(7):7410-7424
  98. 98. Beck-Broichsitter M, Nicolas J, Couvreur P. Design attributes of long-circulating polymeric drug delivery vehicles. European Journal of Pharmaceutics and Biopharmaceutics. 2015;97(Pt B):304-317
  99. 99. Chen G, Roy I, Yang C, Prasad PN. Nanochemistry and nanomedicine for nanoparticle-based diagnostics and therapy. Chemical Reviews. 2016;116(5):2826-2885
  100. 100. Liu J, Lan Y, Yu Z, Tan CS, Parker RM, Abell C, et al. Cucurbit[n]uril-based microcapsules self-assembled within microfluidic droplets: A versatile approach for supramolecular architectures and Materials. Accounts of Chemical Research. 2017;50(2):208-217
  101. 101. Liu D, Cito S, Zhang Y, Wang CF, Sikanen TM, Santos HA. A versatile and robust microfluidic platform toward high throughput synthesis of homogeneous nanoparticles with tunable properties. Advanced Materials. 2015;27(14):2298-2304
  102. 102. Sackmann EK, Fulton AL, Beebe DJ. The present and future role of microfluidics in biomedical research. Nature. 2014;507(7491):181-189

Written By

Faith H.N. Howard, Zijian Gao, Hawari Bin Mansor, Zidi Yang and Munitta Muthana

Submitted: 06 October 2022 Reviewed: 21 December 2022 Published: 16 January 2023