Skip to content
BY-NC-ND 3.0 license Open Access Published by De Gruyter September 26, 2016

Gelatin nanoparticles: a potential candidate for medical applications

  • Rehana Yasmin

    Rehana Yasmin has earned her MS degree in Biotechnology (Medical Biotechnology) from COMSATS, Pakistan. Previously she has done BS (Hons) in Biotechnology and Genetic Engineering from Kohat University of Science and Technology, Pakistan. Presently she is enrolled as a PhD scholar at Quaid-E-Azam University, Pakistan. Her major research focus is “Genetics and Epigenetics of cancer” cancer prognosis and the use of nanotechnology tools as potential therapeutics for cancer.

    , Mohsin Shah

    Mohsin Shah earned his PhD at Gyeongsang National University, South Korea, and he got his post-doctoral research trainings at Nano-Biomaterials Science Laboratory, Gyeongsang National University, South Korea. Now he is an Assistant Professor of Physiology at Khyber Medical University, Peshawar, Pakistan. His previous research is focused on the field of nanoparticles as drug-delivery systems, with special focus on polymers. Research activities in Dr. Mohsin laboratory included endocrinology and reproductive physiology in conjunction with nano-delivery systems.

    EMAIL logo
    , Saeed Ahmad Khan

    Saeed Ahmad Khan graduated in pharmacy from University of Peshawar, Pakistan. He served in Abbott Labs (Pakistan) ltd. and Parke-Davis & co ltd., Pakistan, for a couple of years. He studied MS at Daegu University, South Korea, and eventually obtained PhD at Philipps University Marburg, Germany. He is currently serving at the Department of Pharmacy, Kohat University of Science and Technology as Assistant Professor. A substantial portion of his research interests are focused on designing nanoparticulate delivery system from biopolymers like gelatin and other biodegradable polymers for delivery of macromolecules.

    and Roshan Ali

    Roshan Ali is working as Assistant Professor of Biochemistry at Khyber Medical University, Peshawar, Pakistan. He has done his PhD in Biotechnology from University of Peshawar, Pakistan. He has also worked in the Penn State University USA under the supervision of Dr. Claude dePamphilis. His fields of expertise are computer-aided drug designing and computer programming. He has published a number of international papers in different areas of biochemistry and bioinformatics in world-leading journals.

From the journal Nanotechnology Reviews

Abstract

Gelatin is a protein obtained from the hydrolysis of collagen. Gelatin is an attractive biodegradable material for use in nano-biotechnology and nano-pharmaceutics. Gelatin nanoparticles (NPs) have been widely used as drug and gene carrier to targeted sick tissues including cancer, tuberculosis, HIV infection along with the treatment of vasospasm and restenosis, due to its biocompatibility and biodegradability. For instance, coating with gelatin lowers the cytotoxicity of quantum dots. Moreover, gelatin NPs have the ability to cross the blood-brain barrier, hence proven as a promising candidate to target brain disorders. Macrophage targeting with gelatin NPs for remedy of different diseases is repeatedly reported in previous years. In tissue engineering gelatin is actively utilized for construction of biological and life-long 3D scaffolds for bio-artificial tissues and organ production. Gelatins have a wide range of potential applications which needs to be unraveled in more detail. This review is mainly focused on the applications of gelatin NPs in biomedical sciences.

1 Introduction

Recently nanoparticles (NPs) have offered immense advantages in improving drug delivery systems in terms of increasing efficacy and potency of drugs [1], [2]. In biological sciences, NPs are submicron particles. This small size renders NPs with unique promising properties in terms of drug delivery systems [3]. Consequently, from drug delivery point of view NPs are considered as superior over micro-particles. The surface of NPs is amicable to chemical modifications with specific recognition ligands, functional groups, and polymers of different sizes and charges in order to increase their specificity towards target sites [4]. Drugs or other bioactive ingredients encapsulated in NPs show increased solubility, bioavailability, and bioactivity [5], [6]. The delivery of genes and drugs to their selective targets and their cellular trafficking has been revolutionized with the advent of NPs like micelles, liposomes, and polymeric systems [7].

Nanomaterials derived from biopolymers have certain unique properties such as biodegradability, bioavailability, and low immunogenicity. Biopolymers are isolated from living organisms. They have been immensely used in pharmaceutical and medical fields. Biopolymers for pharmaceutical use are divided into two main classes: (a) polysaccharides and (b) proteins, as summarized in Table 1. Both groups have their distinctive properties; however, protein-based biopolymers posses great loading efficiency which is increased when the drug is loaded onto protein-based NPs at or near its isoelectric point (IEP) [9].

Table 1:

Examples of important biopolymers used as nanomaterial in drug delivery [8].

ClassSourceStructure
Proteins
 Proteins from animals
   AlbuminBloodNot available
   GelatinBones or skin of animals
 Proteins from plants
   GliadinWheat gluten
   Soy proteinsSoybeansNot available
   ZeinMaizeNot available
 Polysaccharides
   ChitosanCrustaceans
   CelluloseAgriculture by product
   DextranAlgae Leuconostocmesenteroides
   HyaluronanSkin and cartilage of rooster combs, etc.,

Bacteria, e.g. Streptococcus zooepidemicus
   Chondroitin sulfateAnimal cartilage
   AlginateBacteria, e.g. Azotobacter and Pseudomonas species.
   PullulanBacterium Aureobasidiumpullulans

1.1 Gelatin and gelatin nanoparticles

Gelatin is a natural versatile biopolymer; it has several important applications due to its low cost, easy availability, biodegradable and biocompatible nature as well as the presence of abundant active groups [10]. Gelatin is poly-ampholyte in nature because it contains both cationic and anionic groups. Gelatin polypeptide is composed of repeating triplets of alanine, glycine and proline residues, responsible for typical triple helical structure of gelatin [11]. Gelatin is a protein obtained from the hydrolysis of collagen. The type of gelatin obtained (type A or type B) is governed by the process of collagen hydrolysis (i.e. acidic and basic hydrolysis, respectively). Types A and B have different IEPs [12]. Type A has IEP 7–9 while type B has IEP 4–5. Each type of gelatin has different drug release potential for different kinds of NPs. It is shown that type B gelatine nanoparticles (GNPs) show better potential in terms of drug delivery than type A [13]. Moreover, gelatin type B physically captures DNA molecule, thus increasing the transfection efficiency of the carriers. Commercially available gelatin is a heterogeneous mixture of polypeptides, presented in varying molecular weights in different ranges, e.g. thousand to million Daltons [14].

The GNPs are nontoxic, biodegradable, bioactive, and inexpensive and are very promising in terms of drug delivery and controlled drug release. Different mechanical properties of GNPs like thermal range and swelling behavior are dependent on the amphetatic interactions on gelatin. Different desired GNPs can be prepared by exploiting the physical and chemical properties of gelatin [15], [16]. Gelatin-based materials need to be cross-linked with glutaraldehyde (GA) or other bifunctional cross-linker such as genipin, carbodiimide/N-hydroxysuccinimide, aldehyde groups, and transglutaminase [10] to render insolubility at high temperatures, reduced swelling in water, and hence drug release from NPs [17]. Drug release was suggested to be dependent on the cross-linking density of gelatin [18].

1.2 Preparation of gelatin nanoparticles

The GNPs can be prepared by several different techniques, including desolvation, coacervation-phase separation, emulsification-solvent evaporation, reverse phase microemulsion, and nanoprecipitation. The preparation methods of GNPs have been elaborated in a recently published review article [10]. Briefly, we describe here some of the main methods for the preparation of GNPs.

1.2.1 Two-step desolvation

In this technique a desolvating agent is added to aqueous gelatin solution to dehydrate gelatin molecules. Low molecular weight gelatin is discarded and remaining high molecular weight portion is dissolved in water and then acetone is added to the solution dropwise, at a controlled pH. Cross-linker is added and stirred at 600 rpm at 40°C for 12 h to obtain hardened uniform spherical NPs [19]. The purification is done with acetone:water (30:70 ratios) followed by extensive centrifugation and finally lyophilization.

1.2.2 Simple coacervation

Simple coacervation is used to prepare stable and small-sized particles [20]. After liquid-liquid phase separation, the polymer settles down in the solution producing two visible, immiscible phases [21]. Natural salts (sodium chloride/sodium sulfate) or alcohols (such as ethanol) are usually used to obtain NPs. However, oppositely charged macromolecules such as proteins or polyelectrolytes are also suggested for coacervation (called complex coacervation) [22]. Dehydration of gelatin molecules finally results in the formation of GNPs which are then cross-linked with other cross-linking agents such as GA [23].

1.2.3 Solvent evaporation

This technique uses single emulsions or double emulsions for the formation of GNPs. Gelatin and drugs in aqueous phase are homogenized with oil phase (polymethyl methacrylate or paraffin oil) and then cross-linked with GA or genipin [13], [24]. The solvent is then evaporated. The solidified NPs are then collected and washed with distilled water to remove additives such as surfactants, and then lyophilized [25].

1.2.4 Microemulsion

In microemulsion, aqueous gelatin solution is poured into surfactant solution [sodium bis (2-ethylhexyl) sulfosuccinate] in n-hexane, cross-linked with GA and finally n-hexane is evaporated and GNPs are recovered [26]. Microemulsion is considered advantageous because it can control the size of NPs.

1.2.5 Nanoprecipitation/solvent displacement method

In this technique water is used as a solvent phase in which gelatin and drug molecules are dissolved. This aqueous phase is then added to ethanol as the nonsolvent phase containing poloxamer as a stabilizer. GNPs are formed on the junction of water and ethanol because of interfacial turbulence generated during solvent displacement [27] and are subsequently cross-linked using GA (schematically shown in Figure 1).

Figure 1: Schematic illustration of nanoprecipitation technique for the formation of GNPs [28].
Figure 1:

Schematic illustration of nanoprecipitation technique for the formation of GNPs [28].

1.3 Mechanisms of drug release from GNPs and GNPs clearance

The GNPs should ideally have improved loading capacity in order to deliver desired amount of drug. Drug can be loaded onto GNPs, either by soaking the particles in drug solution after preparation, or by dissolving the drug in the polymer solution before NPs preparation. With regard to drug loading outcomes, various behaviors of loading efficiency were observed for different drugs (given in Table 2).

Table 2:

Drug loading studies performed on gelatin nanoparticles [8].

Drug usedEncapsulation techniquePreparation techniqueE.E. (%)
CyclosporineDrug mixed with polymer prior to nanoparticle formationEmulsion method90%
Methotrexate==5–15
BSA==
SulphamethaxazoleSwelling in drug solution after nanoparticles formation=18–39%
Chloroquine phosphate==15–20%
Cytarabine==
PaclitaxelDrug mixed with desolvating agentDesolvation with sodium sulfate33–78%
PilocarpineDrug mixed with polymer prior to nanoparticle formationDesolvation with ethanol50%
Doxorubicin==42%
Hydrocortisone==35%
Rosiglitazone=Two-step desolvation90%
Didanosine==70%
Cycloheximide==25–40%
Iron oxide==
Amphotericin B==45%
FITC-dextran=Nanoprecipitation50%
FITC-dextran=One-step desolvation10–80%

There are several possible mechanisms of drug release from gelatin (protein) NPs: (a) liberation due to polymer erosion or degradation; (b) self-diffusion through pores; (c) release via surface erosion of the polymer; and (d) pulsed delivery initiated by the application of an oscillating magnetic or sonic field [29]. In most of the drug release pattern, biphasic release is most common, it includes two stages – an initial phase of burst release of immediate discharge surface associated drugs (weakly interacted) from GNPs and then a second phase of covalently bound (tightly bonded) drugs releases in slow diffusion from the matrix, exhibiting prolonged and sustained release [30]. The initial burst release occurs due to weak adsorption of drugs onto NPs surface [31]. The efficacy of drug release is dependent on the size and loading efficiency of the GNPs as well as drug solubility. Smaller particle tends to achieve big burst effect while larger particles show sustained release thoroughly. Apart from the size, density of gelatin cross-linking is also known to have effect on swelling ratio and drug release pattern from the matrix [10]. pH is another factor which contributes to drug release from GNPs. Proteolytic enzymes also accelerates drug release by GNPs, as shown in Figure 2 [28]. The different mechanism of drug release from gelatin NPs is shown schematically in Figure 3. In the case of gelatin nano-spheres, drug is uniformly distributed in the NP matrix and release occurs by diffusion or erosion of the matrix. If diffusion of the drug is faster than matrix degradation, then the mechanism of drug release occurs mainly by diffusion, otherwise it depends upon degradation. Different methods for in vitro drug quantification are also suggested such as: (i) side-by-side diffusion; (ii) equilibrium dialysis technique; (iii) reverse dialysis sac technique; (iv) ultracentrifugation; (v) ultrafiltration; or (vi) centrifugal ultrafiltration technique [32]. Drug release profiles of GNPs can be deployed by manipulating the size, cross-linker density, and IEPs of the particle [33]. Sustained and targeted release of drug from GNPs inside biological fluids can be accomplished by modifying GNPs surface with site-specific ligands, polyethyl glycols coating, and surface cationization with amine derivatives [34]. GNPs are biodegradable and are cleared easily from the body. Hence no hematological toxicity and nephrotoxcity were observed with GNPs [35]. Moreover, the cells adhesion capacity and cytoskeleton organization were unaffected by GNPs, which depicts the nontoxic nature of GNPs [26].

Figure 2: Model drug (FITC-dextran) release from GNPs after tryptic digestion [28].
Figure 2:

Model drug (FITC-dextran) release from GNPs after tryptic digestion [28].

Figure 3: Drug release patterns from gelatin nanoparticles: (A) Simple diffusion of drug particles; (B) Degradation release; (C) Cleavage of the gelatin matrix by proteolytic enzymes.
Figure 3:

Drug release patterns from gelatin nanoparticles: (A) Simple diffusion of drug particles; (B) Degradation release; (C) Cleavage of the gelatin matrix by proteolytic enzymes.

2 Applications of GNPs in nano-biotechnology and biomedical sciences

The use of nano-biotechnology in molecular diagnostics and life sciences offers new options for clinical diagnostic procedures. Nano-materials have been actively used in different applications like cancer therapy, drug delivery systems, tissue scaffolds, tissue engineering devices, diagnostics, and therapeutics in medicine [36], [37], [38]. Strong association exists between drug and its carrier system, thereby allowing modulation of the pharmacokinetic properties and bio-distribution of the drugs. NPs are the most investigated system amongst different drug carriers and drug delivery systems. Controlled and targeted drug delivery are readily used in trails to enhance localization of drugs to specific tissues and cells, decreasing their interaction and potential toxicity thus increasing the therapeutic index of the existing drug. Receptor-mediated drug targeting is one of the promising, effective, and safe approach used in drug delivery system specifically to its site of action [39]. In pharmaceutical industries most of the potent anticancer drugs pose a major problem of poor solubility along with other unfavorable physicochemical and pharmaceutical properties, which are major limitations in their clinical administration [40]. Controlled drug delivery technology is one of the effective methods for targeted drug delivery of anticancer drugs to tumorigenic sites. Patient convenience, improved efficacy, and reduced toxicity of the existing drugs are some of the major important breakthrough of controlled drug delivery systems over the conventional ones. The effectiveness of drug therapies are expected to be greatly improved by controlled drug delivery systems. GNPs are one of the most suitable candidates in reducing the toxicity issue associated with most of the drugs and could be used as a promising candidate for controlled drug release.

2.1 Targeting human malignancies with nanoparticles

When tumor cells multiply and reach a size of 2–3 mm, the demand for high nutrition and oxygen in the blood increases. A tumor tissue also exhibits distinctive pathophysiological properties that are different from normal tissues or organs. They have defective and leaky vascular architecture, extensive angiogenesis leading to hyper-vasculature, impaired lymphatic drainage, and overproduction of various permeability mediators. This anatomical defectiveness, along with functional abnormalities, results in extensive leakage of blood plasma components, such as macromolecules, NPs and lipidic particles, into the tumor tissue. The poor lymphatic clearance results macromolecules to retain in the tumor for longer time. This process of enhanced particles movement and their retention there for longer duration is called enhanced permeability and retention (EPR) effect as explained by Maeda et al. [41]. The NPs with long circulatory half-life were suggested to be more suitable as a cargo to deliver encapsulated drugs/genes to the target tumors passively by exploiting the EPR effect of the tumor vasculature. Administration of tumor suppressor genes, e.g. p53 (wt-p53), is the main focus of anti-tumor gene therapies, which can help cell to potentially restore its pro-apoptotic function [42]. Cancer cells have the ability to develop resistance against anticancer drugs similar to that of microbial pathogens. This resistance is more commonly seen in solid tumors [43]. Inside experimental models of human tumors transplanted to nude mice, the efficacy of the conjugated cytostatic drugs such as 5-fluorouracil, paclitaxel [44], and doxorubicin [23] with albumin or GNPs were not only shown to be enhanced significantly in comparison to free drug but also reduced cytotoxicity of these drugs substantially.

Ellagitannins, derived from pomegranate have been claimed to prevent cardiovascular diseases and cancer. Gelatin and pomegranate ellagitannins can bind together and assemble into NPs. Ellagitannins encapsulated in GNPs had decreased apoptotic effects on leukemia cells HL-60 [45]. The GNPs conjugated with biotinylated CD3 specific antibodies showed great specificity in cellular uptake in leukemic T lymphocytes (CD3 expressing) [46]. Among the drugs used for treatment of acute leukemia, the effective one is cytarabine (ara-C), which actually affects replication of DNA [47]. Cross-linked GNPs form a swelling controlled drug release system, which effectively delivers cytarabine via diffusion controlled pathway [13]. Lung cancer kills around 25% of the patients each year, and is one of the major cause of cancer leading to mortality in underdeveloped and developed countries [48]. Non-small cell lung cancer constitutes 75–80% of all lung cancers and is the most frequent tumor found in the elders [49]. Administration of chemotherapeutic drugs directly into the lungs not only increases the drug exposure to lung cancers but also causes cytotoxicity to normal cells. Ligand-targeted nano-particulate system could be very effective in the treatment of the lung cancer. A study suggested that epidermal growth factor-modified gelatin nanoparticles could be administered directly to lungs via inhalation and could improve the therapeutic options in lung cancer therapy [50]. Resveratrol (RSV) is a naturally occurring polyphenolic phytoalexin having valuable effects against many diseases including cancer [51] and inflammatory diseases [52]. The prepared RSV-GNPs were reported to exhibit very rapid and more efficient cellular uptake than free RSV. Free RSV treatment shows less anti-proliferative properties than RSV-GNPs treatment in NCI-H460 cells accompanied with more ROS production, DNA damage apoptotic incidence, least toxic response, more bioavailability, and longer half-life [53]. Similarly, Rubus Coreanus Miquel (black raspberry) is known to have anti-oxidative and anticancer properties [54], [55]. Rubus Coreanus Miquel-GNPs were suggested as amazing transport vehicle to immune cells, which activates immune system by enhancing antibody and cytokine production along with inducing proliferation of immunity-related cells [56]. Similarly, Lu et al., has reported the effectiveness of paclitaxel-loaded GNPs against human RT4 bladder transitional cancer cells. Using paclitaxel-loaded NPs, the concentration of drug in the regions of Ta and T1 tumors (i.e. urothelium and lamina propria tissue layers, respectively) was 2.6 times higher than free drug in solution [44]. Beside this, other anticancer drugs such as cycloheximide [15], cyclosporine-A8 [57], and cytarabine [13] have also been loaded into GNPs for potential anticancer therapy. All these attempts give an insight that NPs prepared from gelatin can promise better to enhance therapeutic index of partially water-soluble drugs.

2.2 Crossing the blood-brain barrier

The blood-brain barrier (BBB) is a complex physiological structure in central nervous system (CNS) that inhibits free diffusion of circulating molecules from the blood into the brain. Because of this strong barrier across the entire CNS majority of the brain disorders remain untreatable. Nanoparticluate system has the ability to overcome this barrier [58]. The drugs administered through intravenous injection can be transported across BBB with the help of NPs drug delivery system more efficiently, especially using soluble protein NPs. Drugs like hexapeptide, dalargin, endorphin, dipeptide, kyotorphin, loperamide, doxorubicin, and tubocurarine could be loaded onto these nano-protein particles. Also protein NPs systems are now well known for efficient delivery of several other drugs, e.g. loperamide [59], temozolomide [60] and doxorubicin [61], which, normally prone resistant to cross the BBB. NPs have the ability to easily flow through capillaries and can penetrate their walls to reach their site of action due to their small size and perfect surface functionalization [62], [63]. A study reported that SynB-PEG-GS NPs can effectively cross BBB and can transport drugs across the membrane. SynB-PEG-GS NPs have suitable size distribution, morphology, and show less cytotoxicity. Moreover, the SynB-PEG-GS NPs cross the BBB more efficiently along with improved endothelial uptake [64] (Figure 4).

Figure 4: Diagrammatic representation elaborating the gelatin nanoparticles crossing the blood brain barrier, owing to their small size readily move towards brain tissues.
Figure 4:

Diagrammatic representation elaborating the gelatin nanoparticles crossing the blood brain barrier, owing to their small size readily move towards brain tissues.

Inactivation via short living, Si-RNAs are the recently discovered advanced technique used in gene silencing. However, this technique lacks the desired penetration of small RNA in to the membrane [65], [66]. GNPs provide an efficient vehicle for delivery of these RNAs to the cell, provide safety against degradation, and also have nontoxic degradation products [67]. NO is viewed as an important therapeutic target. Therapeutic efficacy of iNOSsiRNA/GNPs is reported to be significantly higher and long sustainable, and also show neuroprotective effect, which is not the case when treated with the bare drug [68] (Figure 5).

Figure 5: NO-SiRNA-GNPs internalization to neuronal cells and their subsequent release of SiRNA and NO after degradation.
Figure 5:

NO-SiRNA-GNPs internalization to neuronal cells and their subsequent release of SiRNA and NO after degradation.

2.3 Macrophages targeting; chasing HIV by GNPs

Mannose-PEG modified GNPs have shown increased drug loading and double cellular uptake in contrast to unmodified GNPs. This shows that coupling of mannose with GNPs enhances the cellular uptake of drug to different organs [69]. Coupling of the GNPs with mannose (MN-GNPs) significantly enhances the lung, liver, and lymph nodes uptake of the drugs [70]. These surface-modified NPs can effectively deliver biopharmaceuticals to macrophages during macrophage targeting therapies [71]. Biotinylated peptide nucleic acid is reported to be effectively complexed by the avidin-conjugated GNPs. Avidin-conjugated GNPs were suggested as potential carrier system for biotinylated drug derivatives in antisense therapy particularly for HIV treatment [72]. The surface-modified GNPs using novel ligand 4-sulfated N-acetyl galactosamine (4-SO4GalNAc) assures for targeting resident macrophages specifically [73]. In a recent study macrophage cell-line was shown to exhibit two-fold greater internalization of anti-HIV drug, stavudine conjugated with mannosylated GNPs through macrophage mannose-receptor, far better than unconjugated GNP. Thus mannosylated GNPs could be useful as a carrier system in the treatment of macrophage-mediated intracellular infections [74]. Mannan-coated GNPs were also claimed to efficiently target macrophage with didanosine (an anti-HIV drug), with the help of mannosyl receptor mediated endocytosis [75]. Macrophages represent a key target for HIV in addition to CD4 positive lymphocyte cells (CD4+). Mannan-coated didanosine GNPs are reported to be highly accumulated inside spleen, lymph nodes, and brain in comparison to free didanosine injections [75].

2.4 GNPs as DNA carrier

Nanoparticulate systems hold promising importance among the approaches used to deliver DNA due to their easy preparation, versatility, and protection of plasmid DNA via encapsulation [4]. A good gene delivery system should be stable inside plasma and resist reticulo-endothelial system (RES) uptake in order to get its target, e.g. solid tumor. This is achieved by surface modification of NPs with PEG which inhibit macrophages uptake due to steric repulsion of proteins of complement system [76]. GNPs have been studied by many researchers as boat for gene delivery [77]. Gene expression in mice subsequent to intramuscular injection of DNA-gelatin nanospheres was greater and more prolonged compared to an equal amount of naked DNA or DNA complexed with lipofectamine [78]. Human CFTR-gene has been transported to human tracheal epithelial cells for expression using nanospheres composed of gelatin and DNA [79]. The PEG-modified gelatin NPs were also used in BT/20 human breast cancer cells for intracellular absorption [80]. Surface modification of thiolated GNPs with PEG increases their circulation and retention time inside plasma and tumor [81]. Another study also claimed that PEGylated GNPs were absorbed in sufficient number inside the cells (murine embryo fibroblast cell line) by nonspecific endocytosis, remains in the cytoplasm for longer duration and shows good transfection efficiency [82]. Expression of β-galactosidase was also reported in vivo after transformation with respective reporter plasmid DNA-loaded PEGylated GNPs inside Lewis lung carcinoma (LLC) in mice. The in vitro and in vivo results of this study clearly showed that the long-circulating, biocompatible and biodegradable, DNA-encapsulated PEGylated GNPs efficiently transfects LLC cells [83]. Inside pancreatic cancer, p53 mutations are the most common abnormalities [84], which correlate with progression of cancer, apoptotic halt, and chemo-resistance [85], [86]. The combination of gene/drug treatment shows better therapeutic performance of the delivery system, rather than treatment alone with free drug or gene. Thiolated gelatin loaded wt-p53 plasmid or gemcitabine-based therapy of pancreatic cancer was also reported to cause cell apoptosis leading to over-expression of apoptotic transcription factors, protein apoptotic biomarker expression, and under expression of anti-apoptotic transcription factors [87]. Over-expression of the epidermal growth factor receptor (EGFR) on panc-1 human pancreatic adenocarcinoma cells was also previously documented [88]. Another study showed that EGFR-targeted thiolated GNPs could be used as a treatment option for pancreatic cancer, where it served as safe DNA delivery system for gene therapy [89]. The PEG-modified thiolated GNPs as nano-particulate vector system can efficiently deliver DNA to tumorous tissues, containing high glutathione concentration at the tumor site [69]. Cerebral vasospasm is also well known as a major cause of morbidity and death in patients after subarachnoid haemorrhage. An essential vasodilator gene, calcitonin gene-related peptide (CGRP) is in clinical trials in therapy of vasospasm. An important technique used for vasospasm prevention is the injection of recombinant adenovirus, carrying CGRP gene into the cisterna magna, after subarachnoid hemorrhage in rabbits and dogs [90], [91]. Reduction in vasospasm was reported inside rat model of subarachnoid haemorrhage with the over-expression of CGRP along with other neurological improvements. Therefore, the use of Tat-GS NP mediated CGRP gene delivery is thought to have potential for treatment of cerebral vasospasm after subarachnoid hemorrhage [92] (Figure 6). Other bioactive hybrid compounds like siloxane conjugated with gelatin were also reported as efficient and safe novel nonviral vector for gene delivery [93], [94].

Figure 6: Gelatin nanoparticles carry CGRP to hemorrhaged vessels. The expression of CGRP inside the vessel eventually relaxes vasospasm.
Figure 6:

Gelatin nanoparticles carry CGRP to hemorrhaged vessels. The expression of CGRP inside the vessel eventually relaxes vasospasm.

2.5 Gelatin-CpG nucleotide complexes as DNA vaccines

Immune system of a vertebrate recognizes foreign DNA by specific nucleotide sequences carried by the invader DNA. Especially, CpG islands, which are CpG-rich regions, are particularly detected as danger signal. Inside animal models, the innate immune response amplification is achieved by using synthetic oligodeoxy-nucleotides containing CpG motifs (CpG-ODN), complimentary to Toll-like receptor [95]. Clinical studies elaborated that CpG-ODN have a strong potential as adjuvant in antiviral vaccination, treatment of asthma, allergic diseases, and cancer [95]. Inside murine system, CpG-ODN show better uptake and immune-stimulation when administered in the form of CpG-ODN-loaded cationized gelatin NPs (CpG-GNPs) both in vivo and in vitro. Increased production of cytokines like IFN-α in primary human plasmacytoid dendritic cells (DCs), after transformation with cat-ionized GNPs of CpG-ODN suggest that CpG-ODN can be delivered to the cell with the help of GNPs to activate and boost-up the immune system. The same perception was suggested to act better as novel adjuvant for antitumoral and antiviral vaccines [96]. GNPs have been used in immunotherapy, to target lymph nodes with CpG-oligonucleotides to activate antitumoral immunity [96]. Powerful immune-stimulation was reported due to cationized GNPs CpG-oligonucleotides complex [96].

2.6 Management of infectious tuberculosis via GNPs

Tuberculosis (TB), a bacterial infectious disease is also included as a major killer worldwide, infecting both youth and adults. TB stands second to acquired immunodeficiency syndrome (AIDS) in terms of mortality. Although complete treatment of TB is possible, but nowadays a serious negative set back in chemotherapy is rising, due to emergence of multiple drug therapy, longer duration of treatment, and quality of available drugs to common folk [97].

New strategies like nanotechnology are in dire need to enhance drug availability and control release to the infected cells for an extended period of time with the help of an efficient delivery system [98]. The cytotoxicity studies revealed that rifampicin (RIF)-loaded GNPs do much less harm to normal cells than free drug. RIF-GNPs conjugates have been found to accumulate highly in various organs in vivo, while plain RIF lack the desired biodistribution profile. Inside TB-infected mice, administration of RIF-GNPs resulted in reduced bacterial counts in the lungs and spleen. Unlike plain drug, the NPs drug delivery system possibly improves pharmacokinetics of drugs along with sustained plasma level, by increasing mean residence time and area under the curve. GNPs is an attractive option for efficient management of tuberculosis not only because it is expected to have great potential for increasing drug targetability, but also due to reduced dose requirement, which ultimately minimizes side effects associated with multiple use of the drug [99].

2.7 GNPs targeting Leishmaniasis

Leishmaniasis is a microbial disease, caused by intracellular protozoan parasite Leishmania donovanianLeishmania infantum (Leishmania infantum chagasi). In leishmaniasis, the parasite invades and uses macrophages and RES for their replication. Leishmaniasis can be cutaneous or visceral, but the visceral becomes deadly if not treated properly. Macrophages readily engulf NPs and this makes the NPs a more promising carrier for drug delivery to target diseased tissues especially where macrophages are involved [100]. Amphoteracin-B (AmB) is a potent polyene antibiotic, used for the treatment of visceral leishmaniasis (VL) and some other systemic fungal infections like candidiasis. AmB is highly potent against VL which is a protozoan infection disseminated by the parasite called Leishmania donovani, transmitted by sand-fly bite. In VL the macrophages of the lymph node, spleen, liver, and bone marrow are preferentially parasitized and support intracellular replication of the parasite [101]. A study indicated that surface functionalized GNPs (f-GNPs) could be a promising carrier for specific delivery of AmB to macrophages for effective treatment of VL. Furthermore, f-GNPs show reduced cytotoxicity and increased uptake by macrophages [71]. A recent study was conducted to monitor the 1, 2-diacyl-sn-glycero-3-phospho-l-serine-coated GNPs carrying amphotericin B (AmB) for specific targeting to the macrophages in VL. The in vitro and in vivo results of this study show high accumulations of AmB in liver and spleen and resultantly enhanced anti-leishmanial activity [102].

2.8 GNPs as bactericidal bodies

Silver ions or silver NPs have long been known to show antimicrobial activity and can be effectively used for curing infectious wounds [103]. Gelatin fiber mats containing silver NPs are reported to possess higher antibacterial activity against Pseudomonas aeroginosa, Staphylococcus aureus, Escherichia coli, and methicillin-resistant S. aureus [104]. The silver NPs (nAg)-loaded gelatin hydrogel pads were also found to be effective in elimination of E. coli, P. aeruginosa, and S. aureus with about 100% inhibition rate. But these hydrogels have toxic effects on normal skin. However, gelatin hydrogels that contained AgNO3 at lower concentrations (0.75, 1.0 wt%) were considered to be more safe for the skin cells [105]. Similarly, gelatin-based sponge incorporated with silver NPs (AgNPs) was prepared by a solution plasma process. It was shown that the gelatin/AgNP solutions and sponges exhibited excellent bactericidal activity against two bacteria, i.e. E. coli and S. aureus [106]. Gelatin NPs were also suggested as immunological adjuvant to activate and boost immunity against antigen, either cellular or humoral [107].

2.9 GNPs monitoring restenosis (vascular malady)

Restenosis is a term referred to lumen narrowing of arterial vessels after injury caused by multiple factors including surgical procedures such as arterial angioplasty and possess a permanent threat to the failure of the procedure [108], [109]. Nitric oxide (NO) has been found to have an important role in regulation of vascular smooth muscle cells proliferation; therefore, it has been considered as an effective agent for relaxing restenosis. Endogenous production of NO badly suffers from injuries during surgical interventions [110], and supply of exogenous NO is harmful due to its highly reactive nature. Different carrier systems have been proposed to deliver NO to the injured points. Among these, NPs are more advantageous, not only because they can conjugate with different products but also due to potential of controlled release of NO. GS-NO NPs have great potential of NO delivery to the targeted region, to regulate vascular restenosis, and can be administered as restenosis prevention agent after surgery [111] (Figure 7).

Figure 7: Gelatin nanoparticles transport can supply NO in plenty of amount, a vasodilator, to narrowed vessels in order to eliminate the deposited plaque.
Figure 7:

Gelatin nanoparticles transport can supply NO in plenty of amount, a vasodilator, to narrowed vessels in order to eliminate the deposited plaque.

2.10 The role and potential of gelatin in tissue engineering

Tissue engineering is one the most interesting fields of nano-biotechnology. Tissue engineering focuses on the generation of biocompatible tissues in vitro. The use of nano-composites scaffolds in tissue engineering has attracted broad research interest in recent years due to their biological origin [112]. Presently various types of nanocomposites are tried in tissue engineering strategies, to make porous scaffolds. Nano-composite scaffolds have been used in reconstruction of tissues like muscles, bone, tendons, cartilage, ligaments, other multi-tissue organs, and tissues interfaces [113]. Composite systems synergistically combine two or more materials in order to produce a new system with novel properties. Gelatin-based composite systems provide new options for tissue engineering. As gelatin is a denatured biopolymer, the selection of gelatin as a scaffolding material can circumvent the concerns of immunogenicity and pathogen transmission associated with collagen [114]. Most of the work is based on the use of gelatin nano-composite focus in bone defects. Wound infection and bone nonunion are the two most well-known challenges of bone defects. Production and application of gelatin-based ceramics for drug delivery and bone regeneration are in active progression. Gelatin nano-composites can be formed with different types of polymers, both natural and synthetic, according to the purpose of their use. The insulin like growth factor-1 (IGF-1) is reported to get five times increase in quantity inside the cells, when treated with the cationized GNPs containing the IGF-1 plasmid compared with the unmodified particles [115].

It was found that calcium phosphate composites (PCL)/hydroxyapatite (Hap)/gelatin composite fibrous scaffolds are very elastic and let the bone cells (osteoblasts) to penetrate inside them [116]. Synthetic polymers are always surface modified in order to make them more biological, biocompatible, and safe to cells. The porous scaffolds imitate bone extracellular matrix not only in structure and composition but also in function. It is claimed that the preosteoblastic MC3T3-E1 cells attached, spread, and proliferated well with a flat morphology on the mineralized scaffolds, which clearly indicates that scaffolds composed of poly(ε-caprolactone), gelatin, and calcium phosphate are biomimetic and could serve as worthy scaffolds for bone tissue engineering [117]. Gelatin is composed of a unique sequence of amino acids such as glycine, proline, and hydroxyproline which promotes cell adhesion [118]. However, because of its weak mechanical strength it lacks the desired properties. Surface modification with other supportive materials such as hydroxyapatite, aluminosilicate (geopolymers), BaG ceramic was used to improve its strength before using it for scaffold construction. BaG is well known to bind with hard and soft tissues and to boost the bone cells growth [119]. Therefore, it is preferentially used in engineering bone repair scaffolds [120], [121]. Novel, three-dimensional (3D) porous nanocomposites formed by inter-connected microstructure is formatted by fusion of BaG with gelatin which showed great biocompatibility with SaOS-2 cell line in vitro [122].

Commonly alveolar bone loss is strongly linked to periodontal degeneration [123]. Bioactive glasses are osteoconductive and biodegradable biomaterial utilized in bone repair [124]. Bioactive glass ceramic scaffolds containing chitosan-gelatin can attach and spread cells more efficiently. Therefore, gelatine-conjugated nanocomposite scaffolds are thought to be a better option for regeneration of alveolar bone in tissue engineering [125]. The proliferation rate of the cells on the mineralized scaffolds [poly (ε-caprolactone) fibers with gelatin and calcium phosphate] is reported significantly higher than that on the pristine fibrous scaffolds. Different gelatin nano-composites and their use in tissue engineering is enlisted in Table 3.

Table 3:

Different types of gelatin nano-composites used in tissue engineering.

MaterialBiological natureConventional useGNPs conjugatesPurposeReference
Ceramics (hydroxyapatite)Inorganic mineralBone matrixHA-drug-GNPs (nanocomposites and scaffolds)Mechanical properties

Drug delivery
[126], [127]
Calcium phosphateMixtures of calcium ortho-phosphatesCraniofacial, orthopedic fillingsCPC-GNPs

Nanocomposites
Porogens

Drug delivery
[128], [129]
Natural polymers
 HyaluronanUbiquitous polysaccharideBinding site for moleculesNanocompositesControlled delivery of growth factors, drugs, biodegradable sheet[130], [131], [132]
 Chitin and chitosanNatural polysaccharideStructural componentGelatin-chitosan sponges, HA-Chitosan-GNPsDrug delivery[133], [134], [135]
 SilkProtein based polymerInsect excretionGelatin-silk compositesDrug delivery

Designing drug delivery vehicles
[136], [137]
Derived polymers
 Poly(lactic-co-glycolic acid) (PLGA)Synthetic polymersDrug delivery tissue engineeringGelatin-PLGA fibers/compositesCell adhesion and proliferation, drug delivery[138], [139]
 Oligo(poly(ethylene glycol) fumarate) (OPF)PEG-based hydrogelControlled drug delivery, tissue regenerationGelatin-OPF compositesDNA delivery

Minerals, growth factors
[140], [141], [142], [143]
 Poly(propylene fumarate) (PPF)Synthetic polymerBone regeneration, hard tissue regenerationGelatin-PPF compositeGrowth factors (bone regeneration)[144]

2.11 Gelatin lowers quantum dots related toxicity

In recent years nanomolecules, especially quantum dots (QDs) are mainly focused as potential tumor diagnostic and drug delivery system [145], [146], [147]. Although QDs are considered as striking tool both in diagnostics and therapeutics, they are inherently cytotoxic which limits their use in various fields of research and diagnostics [148], [149]. Their toxicity can be reduced in vivo for short time by coating them with certain biocompatible polymers [150]. Coating QDs with gelatin has been reported to effectively reduce its cytotoxic effects on cells. Gelatin coating on QDs surface works as a barrier, which prevent toxic ion leakage from the core of QDs for longer duration inside differentiated pheochromocytoma 12 (PC12) cells [151].

In the field of diagnostics, gelatin-coated TGA-CdTe quantum dots have shown good potential for in vitro diagnosis of cancer [152]. Along with lowering cytotoxicity gelatin coating of QDs has been shown to provide substantially improved biocompatibility to QDs [150], [151]. Theranostics is a blend of diagnostics and therapy. NPs, including QDs, have been investigated for their theranostical potential especially in cell tracking and drug delivery [153], [154]. Recently CdSe/ZnS quantum dots (QDs)-fluorescent gelatin nanospheres (GNs) conjugated with anti-human immunoglobulin-G Fab (QDs-GNs-anti-IgG Fab) were found to be an effective theranostic tool for direct imaging and targeted drug delivery in human osteosarcoma [155] (as shown in Figure 8).

Figure 8: Imaging of QDs and IGg decorated GNPs in the bone tumor site.
Figure 8:

Imaging of QDs and IGg decorated GNPs in the bone tumor site.

3 Conclusion and recommendation

Gelatin NPs have many more applications which are out of scope of this article. This review mainly focuses on the summarization of frontline applications of GNPs in various fields of biomedical sciences. Naturally isolated gelatins pose some problems such as poor batch-to-batch reproducibility and potential antigenicity. Animal-origin gelatins can have risk of contamination with transmissible spongiform encephalopathies. Although, major potentials undesired effects were not attributed to GNPs either in vivo or in vitro, but two major concerns are: safety of the starting material and safety of the cross-linking agents. The limitations and deficiencies in the properties of gelatin can be improved by conjugating it with other suitable compounds to make better nano-carriers, in order to utilize the real potential of GNPs in future research. Therefore, replacing animal-derived gelatins by recombinant human gelatins and GA by genipin were suggested as more safe options [156]. Thus, using the integrated research involving researchers from diverse field of sciences one could reduce the potentials hazards in the application of gelatinous compounds. In the near future, a great value of GNPs is expected in the field of biomedical sciences and cutting edge research.

About the authors

Rehana Yasmin

Rehana Yasmin has earned her MS degree in Biotechnology (Medical Biotechnology) from COMSATS, Pakistan. Previously she has done BS (Hons) in Biotechnology and Genetic Engineering from Kohat University of Science and Technology, Pakistan. Presently she is enrolled as a PhD scholar at Quaid-E-Azam University, Pakistan. Her major research focus is “Genetics and Epigenetics of cancer” cancer prognosis and the use of nanotechnology tools as potential therapeutics for cancer.

Mohsin Shah

Mohsin Shah earned his PhD at Gyeongsang National University, South Korea, and he got his post-doctoral research trainings at Nano-Biomaterials Science Laboratory, Gyeongsang National University, South Korea. Now he is an Assistant Professor of Physiology at Khyber Medical University, Peshawar, Pakistan. His previous research is focused on the field of nanoparticles as drug-delivery systems, with special focus on polymers. Research activities in Dr. Mohsin laboratory included endocrinology and reproductive physiology in conjunction with nano-delivery systems.

Saeed Ahmad Khan

Saeed Ahmad Khan graduated in pharmacy from University of Peshawar, Pakistan. He served in Abbott Labs (Pakistan) ltd. and Parke-Davis & co ltd., Pakistan, for a couple of years. He studied MS at Daegu University, South Korea, and eventually obtained PhD at Philipps University Marburg, Germany. He is currently serving at the Department of Pharmacy, Kohat University of Science and Technology as Assistant Professor. A substantial portion of his research interests are focused on designing nanoparticulate delivery system from biopolymers like gelatin and other biodegradable polymers for delivery of macromolecules.

Roshan Ali

Roshan Ali is working as Assistant Professor of Biochemistry at Khyber Medical University, Peshawar, Pakistan. He has done his PhD in Biotechnology from University of Peshawar, Pakistan. He has also worked in the Penn State University USA under the supervision of Dr. Claude dePamphilis. His fields of expertise are computer-aided drug designing and computer programming. He has published a number of international papers in different areas of biochemistry and bioinformatics in world-leading journals.

References

[1] Kreuter J. Nanoparticulate systems for brain delivery of drugs. Adv. Drug Deliv. Rev. 2001, 47, 65–81.10.1016/S0169-409X(00)00122-8Search in Google Scholar

[2] Vijayanathan V, Thomas T, Thomas T. DNA nanoparticles and development of DNA delivery vehicles for gene therapy. Biochemistry 2002, 41, 14085–14094.10.1021/bi0203987Search in Google Scholar

[3] Buzea C, Pacheco II, Robbie K. Nanomaterials and nanoparticles: sources and toxicity. Biointerphases 2007, 2, MR17–MR71.10.1116/1.2815690Search in Google Scholar

[4] Panyam J, Labhasetwar V. Biodegradable nanoparticles for drug and gene delivery to cells and tissue. Adv. Drug Deliv. Rev. 2003, 55, 329–347.10.1016/S0169-409X(02)00228-4Search in Google Scholar

[5] Morimoto K, Katsumata H, Yabuta T, Iwanaga K, Kakemi M, Tabata Y, Ikada Y. Gelatin microspheres as a pulmonary delivery system: evaluation of salmon calcitonin absorption. J. Pharm. Pharmacol. 2000, 52, 611–617.10.1211/0022357001774444Search in Google Scholar PubMed

[6] Tran PH-L, Tran TT-D, Lee B-J. Enhanced solubility and modified release of poorly water-soluble drugs via self-assembled gelatin–oleic acid nanoparticles. Int. J. Pharm. 2013, 455, 235–240.10.1016/j.ijpharm.2013.07.025Search in Google Scholar PubMed

[7] Shenoy D, Little S, Langer R, Amiji M. Poly (ethylene oxide)-modified poly (β-amino ester) nanoparticles as a pH-sensitive system for tumor-targeted delivery of hydrophobic drugs: part 2. In vivo distribution and tumor localization studies. Pharm. Res. 2005, 22, 2107–2114.10.1007/s11095-005-8343-0Search in Google Scholar PubMed PubMed Central

[8] Khan SA. Gelatin Nanoparticles as Potential Nanocarriers for Macromolecular Drugs, Philipps University Marburg: Germany, 2014.Search in Google Scholar

[9] Ofokansi K, Winter G, Fricker G, Coester C. Matrix-loaded biodegradable gelatin nanoparticles as new approach to improve drug loading and delivery. Eur. J. Pharm. Biopharm. 2010, 76, 1–9.10.1016/j.ejpb.2010.04.008Search in Google Scholar PubMed

[10] Elzoghby AO. Gelatin-based nanoparticles as drug and gene delivery systems: reviewing three decades of research. J. Controll. Release 2013, 172, 1075–1091.10.1016/j.jconrel.2013.09.019Search in Google Scholar PubMed

[11] Azarmi S, Huang Y, Chen H, McQuarrie S, Abrams D, Roa W, Finlay WH, Miller GG, Löbenberg R. Optimization of a two-step desolvation method for preparing gelatin nanoparticles and cell uptake studies in 143B osteosarcoma cancer cells. J. Pharm. Pharm. Sci. 2006, 9, 124–132.Search in Google Scholar

[12] Sawicka J. [Microencapsulization of cholecalciferol by coacervation]. Die Pharmazie 1990, 45, 264–265.Search in Google Scholar

[13] Bajpai A, Choubey J. In vitro release dynamics of an anticancer drug from swellable gelatin nanoparticles. J. Appl. Polymer Sci. 2006, 101, 2320–2332.10.1002/app.23761Search in Google Scholar

[14] Fraunhofer W, Winter G, Coester C. Asymmetrical flow field-flow fractionation and multiangle light scattering for analysis of gelatin nanoparticle drug carrier systems. Anal. Chem. 2004, 76, 1909–1920.10.1021/ac0353031Search in Google Scholar

[15] Saxena A, Sachin K, Bohidar H, Verma AK. Effect of molecular weight heterogeneity on drug encapsulation efficiency of gelatin nano-particles. Colloids Surf. B Biointerfaces 2005, 45, 42–48.10.1016/j.colsurfb.2005.07.005Search in Google Scholar

[16] Zwiorek K, Kloeckner J, Wagner E, Coester C. Gelatin nanoparticles as a new and simple gene delivery system. J. Pharm. Pharm. Sci. 2004, 7, 22–28.Search in Google Scholar

[17] Jameela S, Jayakrishnan A. Glutaraldehyde cross-linked chitosan microspheres as a long acting biodegradable drug delivery vehicle: studies on the in vitro release of mitoxantrone and in vivo degradation of microspheres in rat muscle. Biomaterials 1995, 16, 769–775.10.1016/0142-9612(95)99639-4Search in Google Scholar

[18] Levy M-C, Andry M-C. Microcapsules prepared through interfacial cross-linking of starch derivatives. Int. J. Pharm. 1990, 62, 27–35.10.1016/0378-5173(90)90028-3Search in Google Scholar

[19] Farrugia CA, Groves MJ. Gelatin behaviour in dilute aqueous solution: designing a nanoparticulate formulation. J. Pharm. Pharmacol. 1999, 51, 643–649.10.1211/0022357991772925Search in Google Scholar

[20] Mohanty B, Bohidar H. Systematic of alcohol-induced simple coacervation in aqueous gelatin solutions. Biomacromolecules 2003, 4, 1080–1086.10.1021/bm034080lSearch in Google Scholar

[21] Mohanty B, Aswal V, Kohlbrecher J, Bohidar H. Synthesis of gelatin nanoparticles via simple coacervation. J. Surf. Sci. Technol. 2005, 21, 149.Search in Google Scholar

[22] Marty J, Oppenheim R, Speiser P. Nanoparticles–a new colloidal drug delivery system. Pharm. Acta Helv. 1978, 53, 17.Search in Google Scholar

[23] Leo E, Vandelli MA, Cameroni R, Forni F. Doxorubicin-loaded gelatin nanoparticles stabilized by glutaraldehyde: involvement of the drug in the cross-linking process. Int. J. Pharm. 1997, 155, 75–82.10.1016/S0378-5173(97)00149-XSearch in Google Scholar

[24] Choubey J, Bajpai A. Investigation on magnetically controlled delivery of doxorubicin from superparamagnetic nanocarriers of gelatin crosslinked with genipin. J. Mater. Sci. Mater. Med. 2010, 21, 1573–1586.10.1007/s10856-010-3997-5Search in Google Scholar PubMed

[25] Rao JP, Geckeler KE. Polymer nanoparticles: preparation techniques and size-control parameters. Prog. Polym. Sci. 2011, 36, 887–913.10.1016/j.progpolymsci.2011.01.001Search in Google Scholar

[26] Gupta AK, Gupta M, Yarwood SJ, Curtis AS. Effect of cellular uptake of gelatin nanoparticles on adhesion, morphology and cytoskeleton organisation of human fibroblasts. J. Control. Release 2004, 95, 197–207.10.1016/j.jconrel.2003.11.006Search in Google Scholar PubMed

[27] Lee E, Khan S, Lim K-H. Gelatin nanoparticle preparation by nanoprecipitation. J. Biomater. Sci. Polym. Ed. 2011, 22, 753–771.10.1163/092050610X492093Search in Google Scholar

[28] Khan SA, Schneider M. Improvement of nanoprecipitation technique for preparation of gelatin nanoparticles and potential macromolecular drug loading. Macromol. Biosci. 2013, 13, 455–463.10.1002/mabi.201200382Search in Google Scholar

[29] Couvreur P, Puisieux F. Nano-and microparticles for the delivery of polypeptides and proteins. Adv. Drug Deliv. Rev. 1993, 10, 141–162.10.1016/0169-409X(93)90046-7Search in Google Scholar

[30] Hans M, Lowman A. Biodegradable nanoparticles for drug delivery and targeting. Curr. Opin. Solid State Mater. Sci. 2002, 6, 319–327.10.1016/S1359-0286(02)00117-1Search in Google Scholar

[31] Gaihre B, Khil MS, Lee DR, Kim HY. Gelatin-coated magnetic iron oxide nanoparticles as carrier system: drug loading and in vitro drug release study. Int. J. Pharm. 2009, 365, 180–189.10.1016/j.ijpharm.2008.08.020Search in Google Scholar

[32] Soppimath KS, Aminabhavi TM, Kulkarni AR, Rudzinski WE. Biodegradable polymeric nanoparticles as drug delivery devices. J. Control. Release 2001, 70, 1–20.10.1016/S0168-3659(00)00339-4Search in Google Scholar

[33] Santoro M, Tatara AM, Mikos AG. Gelatin carriers for drug and cell delivery in tissue engineering. J. Control. Release 2014, 190, 210–218.10.1016/j.jconrel.2014.04.014Search in Google Scholar PubMed PubMed Central

[34] Sahoo N, Sahoo RK, Biswas N, Guha A, Kuotsu K. Recent advancement of gelatin nanoparticles in drug and vaccine delivery. Int. J. Biol. Macromol. 2015, 81, 317–331.10.1016/j.ijbiomac.2015.08.006Search in Google Scholar PubMed

[35] Nahar M, Mishra D, Dubey V, Jain NK. Development, characterization, and toxicity evaluation of amphotericin B–loaded gelatin nanoparticles. Nanomed. Nanotechnol. Biol. Med. 2008, 4, 252–261.10.1016/j.nano.2008.03.007Search in Google Scholar PubMed

[36] van Vlerken LE, Amiji MM. Multi-functional polymeric nanoparticles for tumour-targeted drug delivery. Expert Opin. Drug Deliv. 2006, 3, 205–16.10.1517/17425247.3.2.205Search in Google Scholar PubMed

[37] Liu Y, Miyoshi H, Nakamura M. Nanomedicine for drug delivery and imaging: a promising avenue for cancer therapy and diagnosis using targeted functional nanoparticles. Int. J. Cancer 2007, 120, 2527–2537.10.1002/ijc.22709Search in Google Scholar

[38] Vasir JK, Labhasetwar V. Biodegradable nanoparticles for cytosolic delivery of therapeutics. Adv. Drug Deliv. Rev. 2007, 59, 718–728.10.1016/j.addr.2007.06.003Search in Google Scholar

[39] Takakura Y, Hashida M. Macromolecular carrier systems for targeted drug delivery: pharmacokinetic considerations on biodistribution. Pharm. Res. 1996, 13, 820–831.10.1023/A:1016084508097Search in Google Scholar

[40] Buzdar AU, Robertson JF, Eiermann W, Nabholtz JM. An overview of the pharmacology and pharmacokinetics of the newer generation aromatase inhibitors anastrozole, letrozole, and exemestane. Cancer 2002, 95, 2006–2016.10.1002/cncr.10908Search in Google Scholar

[41] Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J. Control. Release 2000, 65, 271–284.10.1016/S0168-3659(99)00248-5Search in Google Scholar

[42] McCormick F. Cancer gene therapy: fringe or cutting edge? Nat. Rev. Cancer 2001, 1, 130–141.10.1038/35101008Search in Google Scholar PubMed

[43] Brown R, Links M. Clinical relevance of the molecular mechanisms of resistance to anti-cancer drugs. Expert Rev. Mol. Med. 1999, 1, 1–21.10.1017/S1462399499001099XSearch in Google Scholar PubMed

[44] Lu Z, Yeh T-K, Tsai M, Au JL-S, Wientjes MG. Paclitaxel-loaded gelatin nanoparticles for intravesical bladder cancer therapy. Clin. Cancer Res. 2004, 10, 7677–7684.10.1158/1078-0432.CCR-04-1443Search in Google Scholar PubMed

[45] Li Z, Percival SS, Bonard S, Gu L. Fabrication of nanoparticles using partially purified pomegranate ellagitannins and gelatin and their apoptotic effects. Mol. Nutr. Food Res. 2011, 55, 1096–1103.10.1002/mnfr.201000528Search in Google Scholar PubMed

[46] Dinauer N, Balthasar S, Weber C, Kreuter J, Langer K, von Briesen H. Selective targeting of antibody-conjugated nanoparticles to leukemic cells and primary T-lymphocytes. Biomaterials 2005, 26, 5898–5906.10.1016/j.biomaterials.2005.02.038Search in Google Scholar PubMed

[47] Blanco M, Trigo R, Teijón C, Gómez C, Teijón J. Slow releasing of ara-C from poly (2-hydroxyethyl methacrylate) and poly (2-hydroxyethyl methacrylate-co-N-vinyl-2-pyrrolidone) hydrogels implanted subcutaneously in the back of rats. Biomaterials 1998, 19, 861–869.10.1016/S0142-9612(97)00247-0Search in Google Scholar

[48] Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ. Cancer statistics, 2008. CA Cancer J. Clin. 2008, 58, 71–96.10.3322/CA.2007.0010Search in Google Scholar PubMed

[49] Brown J, Eraut D, Trask C, Davison A. Age and the treatment of lung cancer. Thorax 1996, 51, 564–568.10.1136/thx.51.6.564Search in Google Scholar PubMed PubMed Central

[50] Long J-T, Cheang T-y, Zhuo S-Y, Zeng R-F, Dai Q-S, Li H-P, Fang S. Anticancer drug-loaded multifunctional nanoparticles to enhance the chemotherapeutic efficacy in lung cancer metastasis. J. Nanobiotechnol. 2014, 12, 37.10.1186/s12951-014-0037-5Search in Google Scholar PubMed PubMed Central

[51] Signorelli P, Ghidoni R. Resveratrol as an anticancer nutrient: molecular basis, open questions and promises. J. Nutr. Biochem. 2005, 16, 449–466.10.1016/j.jnutbio.2005.01.017Search in Google Scholar PubMed

[52] Kang O-H, Jang H-J, Chae H-S, Oh Y-C, Choi J-G, Lee Y-S, Kim J-H, Kim YC, Sohn DH, Park H. Anti-inflammatory mechanisms of resveratrol in activated HMC-1 cells: pivotal roles of NF-κB and MAPK. Pharmacol. Res. 2009, 59, 330–337.10.1016/j.phrs.2009.01.009Search in Google Scholar PubMed

[53] Karthikeyan S, Prasad NR, Ganamani A, Balamurugan E. Anticancer activity of resveratrol-loaded gelatin nanoparticles on NCI-H460 non-small cell lung cancer cells. Biomed. Prev. Nutr. 2013, 3, 64–73.10.1016/j.bionut.2012.10.009Search in Google Scholar

[54] Kim EJ, Lee Y-J, Shin H-K, Park JHY. Induction of apoptosis by the aqueous extract of Rubus coreanum in HT-29 human colon cancer cells. Nutrition 2005, 21, 1141–1148.10.1016/j.nut.2005.02.012Search in Google Scholar PubMed

[55] Ju HK, Cho EJ, Jang MH, Lee YY, Hong SS, Park JH, Kwon SW. Characterization of increased phenolic compounds from fermented Bokbunja (Rubus coreanus Miq.) and related antioxidant activity. J. Pharm. Biomed. Anal. 2009, 49, 820–827.10.1016/j.jpba.2008.12.024Search in Google Scholar PubMed

[56] Seo YC, Choi WY, Lee CG, Cha SW, Kim YO, Kim J-C, Drummen GP, Lee HY. Enhanced immunomodulatory activity of gelatin-encapsulated Rubus coreanus Miquel nanoparticles. Int. J. Mol. Sci. 2011, 12, 9031–9056.10.3390/ijms12129031Search in Google Scholar PubMed PubMed Central

[57] El-Shabouri M. Positively charged nanoparticles for improving the oral bioavailability of cyclosporin-A. Int. J. Pharm. 2002, 249, 101–108.10.1016/S0378-5173(02)00461-1Search in Google Scholar

[58] Modi G, Pillay V, Choonara YE, Ndesendo VM, du Toit LC, Naidoo D. Nanotechnological applications for the treatment of neurodegenerative disorders. Prog. Neurobiol. 2009, 88, 272–285.10.1016/j.pneurobio.2009.05.002Search in Google Scholar PubMed

[59] Ulbrich K, Hekmatara T, Herbert E, Kreuter J. Transferrin-and transferrin-receptor-antibody-modified nanoparticles enable drug delivery across the blood–brain barrier (BBB). Eur. J. Pharm. Biopharm. 2009, 71, 251–256.10.1016/j.ejpb.2008.08.021Search in Google Scholar PubMed

[60] Tian X-h, Lin X-N, Wei F, Feng W, Huang Z-C, Wang P, Ren L, Diao Y. Enhanced brain targeting of temozolomide in polysorbate-80 coated polybutylcyanoacrylate nanoparticles. Int. J. Nanomed. 2011, 6, 445–452.10.2147/IJN.S16570Search in Google Scholar PubMed PubMed Central

[61] Kreuter J. Nanoparticles – a historical perspective. Int. J. Pharm. 2007, 331, 1–10.10.1016/j.ijpharm.2006.10.021Search in Google Scholar PubMed

[62] Koo Y-EL, Reddy GR, Bhojani M, Schneider R, Philbert MA, Rehemtulla A, Ross BD, Kopelman R. Brain cancer diagnosis and therapy with nanoplatforms. Adv. Drug Deliv. Rev. 2006, 58, 1556–1577.10.1016/j.addr.2006.09.012Search in Google Scholar PubMed

[63] Moghimi SM, Hunter AC, Murray JC. Nanomedicine: current status and future prospects. FASEB J. 2005,19, 311–330.10.1096/fj.04-2747revSearch in Google Scholar PubMed

[64] Tian X-h, Wei F, Wang T-x, Wang P, Lin X-n, Wang J, Wang D, Ren L. In vitro and in vivo studies on gelatin-siloxane nanoparticles conjugated with SynB peptide to increase drug delivery to the brain. Int. J. Nanomed. 2012, 7, 1031.10.2147/IJN.S26541Search in Google Scholar PubMed PubMed Central

[65] Burnett JC, Rossi JJ. RNA-based therapeutics: current progress and future prospects. Chem. Biol. 2012, 19, 60–71.10.1016/j.chembiol.2011.12.008Search in Google Scholar PubMed PubMed Central

[66] Glebova K, Marakhonov A, Baranova A, Skoblov MY. Therapeutic siRNAs and nonviral systems for their delivery. Mol. Biol. 2012, 46, 335–348.10.1134/S0026893312020069Search in Google Scholar

[67] Ishikawa H, Nakamura Y, Jo J-i, Tabata Y. Gelatin nanospheres incorporating siRNA for controlled intracellular release. Biomaterials 2012, 33, 9097–9104.10.1016/j.biomaterials.2012.08.032Search in Google Scholar

[68] Kim I-D, Sawicki E, Lee H-K, Lee E-H, Park HJ, Han P-L, Kim KK, Choi H, Lee J-K. Robust neuroprotective effects of intranasally delivered iNOS siRNA encapsulated in gelatin nanoparticles in the postischemic brain. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 1219–1229.10.1016/j.nano.2016.01.002Search in Google Scholar

[69] Kommareddy S, Amiji M. Poly (ethylene glycol)–modified thiolated gelatin nanoparticles for glutathione-responsive intracellular DNA delivery. Nanomed. Nanotechnol. Biol. Med. 2007, 3, 32–42.10.1016/j.nano.2006.11.005Search in Google Scholar

[70] Jain SK, Gupta Y, Jain A, Saxena AR, Khare P, Jain A. Mannosylated gelatin nanoparticles bearing an anti-HIV drug didanosine for site-specific delivery. Nanomed. Nanotechnol. Biol. Med. 2008, 4, 41–48.10.1016/j.nano.2007.11.004Search in Google Scholar

[71] Nahar M, Dubey V, Mishra D, Mishra PK, Dube A, Jain NK. In vitro evaluation of surface functionalized gelatin nanoparticles for macrophage targeting in the therapy of visceral leishmaniasis. J. Drug Target. 2010, 18, 93–105.10.3109/10611860903115290Search in Google Scholar

[72] Weber C, Coester C, Kreuter J, Langer K. Desolvation process and surface characterisation of protein nanoparticles. Int. J. Pharm. 2000, 194, 91–102.10.1016/S0378-5173(99)00370-1Search in Google Scholar

[73] Dwivedi P, Kansal S, Sharma M, Shukla R, Verma A, Shukla P, Tripathi P, Gupta P, Saini D, Khandelwal K. Exploiting 4-sulphate N-acetyl galactosamine decorated gelatin nanoparticles for effective targeting to professional phagocytes in vitro and in vivo. J. Drug Target. 2012, 20, 883–896.10.3109/1061186X.2012.725169Search in Google Scholar

[74] Mahajan S, Prashant C, Koul V, Choudhary V, K Dinda A. Receptor specific macrophage targeting by mannose-conjugated gelatin nanoparticles-an in vitro and in vivo study. Curr. Nanosci. 2010, 6, 413–421.10.2174/157341310791658928Search in Google Scholar

[75] Kaur A, Jain S, Tiwary A. Mannan-coated gelatin nanoparticles for sustained and targeted delivery of didanosine: in vitro and in vivo evaluation. Acta Pharm. 2008, 58, 61–74.10.2478/v10007-007-0045-1Search in Google Scholar

[76] Torchilin VP, Trubetskoy VS. Which polymers can make nanoparticulate drug carriers long-circulating? Adv. Drug Deliv. Rev. 1995, 16, 141–155.10.1016/0169-409X(95)00022-YSearch in Google Scholar

[77] Truong-Le VL, Walsh SM, Schweibert E, Mao H-Q, Guggino WB, August JT, Leong KW. Gene transfer by DNA–gelatin nanospheres. Arch. Biochem Biophys. 1999, 361, 47–56.10.1006/abbi.1998.0975Search in Google Scholar PubMed

[78] Truong-Le VL, August JT, Leong KW. Controlled gene delivery by DNA-gelatin nanospheres. Hum. Gene Ther. 1998, 9, 1709–1717.10.1089/hum.1998.9.12-1709Search in Google Scholar PubMed

[79] Leong YS, Candau F. Inverse microemulsion polymerization. J. Phys. Chem. 1982, 86, 2269–2271.10.1021/j100210a001Search in Google Scholar

[80] Kaul G, Amiji M. Long-circulating poly (ethylene glycol)-modified gelatin nanoparticles for intracellular delivery. Pharm. Res. 2002, 19, 1061–1067.10.1023/A:1016486910719Search in Google Scholar

[81] Kommareddy S, Amiji M. Biodistribution and pharmacokinetic analysis of long-circulating thiolated gelatin nanoparticles following systemic administration in breast cancer-bearing mice. J. Pharm. Sci. 2007, 96, 397–407.10.1002/jps.20813Search in Google Scholar PubMed

[82] Kaul G, Amiji M. Cellular interactions and in vitro DNA transfection studies with poly (ethylene glycol)-modified gelatin nanoparticles. J. Pharm. Sci. 2005, 94, 184–198.10.1002/jps.20216Search in Google Scholar PubMed

[83] Kaul G, Amiji M. Tumor-targeted gene delivery using poly (ethylene glycol)-modified gelatin nanoparticles: in vitro and in vivo studies. Pharm. Res. 2005, 22, 951–961.10.1007/s11095-005-4590-3Search in Google Scholar PubMed PubMed Central

[84] Barton C, Staddon S, Hughes C, Hall P, O’sullivan C, Klöppel G, Theis B, Russell R, Neoptolemos J, Williamson R. Abnormalities of the p53 tumour suppressor gene in human pancreatic cancer. Br. J. Cancer 1991, 64, 1076.10.1038/bjc.1991.467Search in Google Scholar PubMed PubMed Central

[85] Fridman JS, Lowe SW. Control of apoptosis by p53. Oncogene 2003, 22, 9030–9040.10.1038/sj.onc.1207116Search in Google Scholar PubMed

[86] Green DR, Kroemer G. Cytoplasmic functions of the tumour suppressor p53. Nature 2009, 458, 1127–1130.10.1038/nature07986Search in Google Scholar PubMed PubMed Central

[87] Xu J, Singh A, Amiji MM. Redox-responsive targeted gelatin nanoparticles for delivery of combination wt-p53 expressing plasmid DNA and gemcitabine in the treatment of pancreatic cancer. BMC Cancer 2014, 14, 75.10.1186/1471-2407-14-75Search in Google Scholar PubMed PubMed Central

[88] Bardeesy N, DePinho RA. Pancreatic cancer biology and genetics. Nat. Rev. Cancer 2002, 2, 897–909.10.1038/nrc949Search in Google Scholar PubMed

[89] Xu J, Amiji M. Therapeutic gene delivery and transfection in human pancreatic cancer cells using epidermal growth factor receptor-targeted gelatin nanoparticles. J. Vis. Exp. 2012, 59, e3612.Search in Google Scholar

[90] Satoh M, Perkins E, Kimura H, Tang J, Chun Y, Heistad DD, Zhang JH. Posttreatment with adenovirus-mediated gene transfer of calcitonin gene-related peptide to reverse cerebral vasospasm in dogs. J. Neurosurg. 2002, 97, 136–142.10.3171/jns.2002.97.1.0136Search in Google Scholar PubMed

[91] Toyoda K, Faraci FM, Watanabe Y, Ueda T, Andresen JJ, Chu Y, Otake S, Heistad DD. Gene transfer of calcitonin gene–related peptide prevents vasoconstriction after subarachnoid hemorrhage. Circ. Res. 2000, 87, 818–824.10.1161/01.RES.87.9.818Search in Google Scholar

[92] Tian X-H, Wang Z-G, Meng H, Wang Y-H, Feng W, Wei F, Huang Z-C, Lin X-N, Ren L. Tat peptide-decorated gelatin-siloxane nanoparticles for delivery of CGRP transgene in treatment of cerebral vasospasm. Int. J. Nanomed. 2013, 8, 865.10.2147/IJN.S39951Search in Google Scholar PubMed PubMed Central

[93] Wang Z-Y, Zhao Y, Ren L, Jin L-H, Sun L-P, Yin P, Zhang Y-F, Zhang Q-Q. Novel gelatin–siloxane nanoparticles decorated by Tat peptide as vectors for gene therapy. Nanotechnology 2008, 19, 445103.10.1088/0957-4484/19/44/445103Search in Google Scholar PubMed

[94] Yin P, Wang J, Ren L, Wang Z-Y, Wang T-X, Wang D, Tian M-M, Tian X-H. Conjugation of membrane-destabilizing peptide onto gelatin–siloxane nanoparticles for efficient gene expression. Mater. Sci. Engng C 2010, 30, 1260–1265.10.1016/j.msec.2010.07.006Search in Google Scholar

[95] Link BK, Ballas ZK, Weisdorf D, Wooldridge JE, Bossler AD, Shannon M, Rasmussen WL, Krieg AM, Weiner GJ. Oligodeoxynucleotide CpG 7909 delivered as intravenous infusion demonstrates immunologic modulation in patients with previously treated non-Hodgkin lymphoma. J. Immunother. 2006, 29, 558–568.10.1097/01.cji.0000211304.60126.8fSearch in Google Scholar PubMed

[96] Bourquin C, Anz D, Zwiorek K, Lanz A-L, Fuchs S, Weigel S, Wurzenberger C, von der Borch P, Golic M, Moder S. Targeting CpG oligonucleotides to the lymph node by nanoparticles elicits efficient antitumoral immunity. J. Immunol. 2008, 181, 2990–2998.10.4049/jimmunol.181.5.2990Search in Google Scholar PubMed

[97] Du Toit LC, Pillay V, Danckwerts MP. Tuberculosis chemotherapy: current drug delivery approaches. Respir. Res. 2006, 7, 118.10.1186/1465-9921-7-118Search in Google Scholar PubMed PubMed Central

[98] Gelperina S, Kisich K, Iseman MD, Heifets L. The potential advantages of nanoparticle drug delivery systems in chemotherapy of tuberculosis. Am. J. Respir. Crit. Care Med. 2005, 172, 1487–1490.10.1164/rccm.200504-613PPSearch in Google Scholar

[99] Saraogi GK, Gupta P, Gupta U, Jain N, Agrawal G. Gelatin nanocarriers as potential vectors for effective management of tuberculosis. Int. J. Pharm. 2010, 385, 143–149.10.1016/j.ijpharm.2009.10.004Search in Google Scholar

[100] Carter K, Dolan T, Alexander J, Baillie A, McColgan C. Visceral leishmaniasis: drug carrier system characteristics and the ability to clear parasites from the liver, spleen and bone marrow in Leishmania donovani infected BALB/c mice. J. Pharm. Pharmacol. 1989, 41, 87–91.10.1111/j.2042-7158.1989.tb06399.xSearch in Google Scholar

[101] Kleinberg M. What is the current and future status of conventional amphotericin B? Int. J. Antimicrob. Agents 2006, 27, 12–16.10.1016/j.ijantimicag.2006.03.013Search in Google Scholar

[102] Khatik R, Dwivedi P, Khare P, Kansal S, Dube A, Mishra PR, Dwivedi AK. Development of targeted 1, 2-diacyl-sn-glycero-3-phospho-l-serine-coated gelatin nanoparticles loaded with amphotericin B for improved in vitro and in vivo effect in leishmaniasis. Expert Opin. Drug Deliv. 2014, 11, 633–646.10.1517/17425247.2014.889678Search in Google Scholar

[103] Klasen H. A historical review of the use of silver in the treatment of burns. II. Renewed interest for silver. Burns 2000, 26, 131–138.10.1016/S0305-4179(99)00116-3Search in Google Scholar

[104] Rujitanaroj P-O, Pimpha N, Supaphol P. Wound-dressing materials with antibacterial activity from electrospun gelatin fiber mats containing silver nanoparticles. Polymer 2008, 49, 4723–4732.10.1016/j.polymer.2008.08.021Search in Google Scholar

[105] Rattanaruengsrikul V, Pimpha N, Supaphol P. In vitro efficacy and toxicology evaluation of silver nanoparticle-loaded gelatin hydrogel pads as antibacterial wound dressings. J. Appl. Polym. Sci. 2012, 124,1668–1682.10.1002/app.35195Search in Google Scholar

[106] Kim SC, Kim SM, Yoon GJ, Nam SW, Lee SY, Kim JW. Gelatin-based sponge with Ag nanoparticles prepared by solution plasma: Fabrication, characteristics, and their bactericidal effect. Curr. Appl. Phys. 2014, 14, S172–S179.10.1016/j.cap.2013.12.032Search in Google Scholar

[107] Nakaoka R, Tabata Y, Ikada Y. Potentiality of gelatin microsphere as immunological adjuvant. Vaccine 1995, 13, 653–661.10.1016/0264-410X(94)00024-HSearch in Google Scholar

[108] Janero DR, Ewing JF. Nitric oxide and postangioplasty restenosis: pathological correlates and therapeutic potential. Free Radical Biol. Med. 2000, 29, 1199–1221.10.1016/S0891-5849(00)00434-2Search in Google Scholar

[109] Weintraub WS. The pathophysiology and burden of restenosis. Am. J. Cardiol. 2007, 100, S3–S9.10.1016/j.amjcard.2007.06.002Search in Google Scholar PubMed

[110] Costa MA, Simon DI. Molecular basis of restenosis and drug-eluting stents. Circulation 2005, 111, 2257–2273.10.1161/01.CIR.0000163587.36485.A7Search in Google Scholar PubMed

[111] Zhang QY, Wang ZY, Wen F, Ren L, Li J, Teoh SH, Thian ES. Gelatin–siloxane nanoparticles to deliver nitric oxide for vascular cell regulation: synthesis, cytocompatibility, and cellular responses. J. Biomed. Mater. Res. Part A 2015, 103, 929–938.10.1002/jbm.a.35239Search in Google Scholar PubMed

[112] Shea LD, Wang D, Franceschi RT, Mooney DJ. Engineered bone development from a pre-osteoblast cell line on three-dimensional scaffolds. Tissue Engng 2000, 6, 605–617.10.1089/10763270050199550Search in Google Scholar PubMed

[113] Maquet V, Boccaccini AR, Pravata L, Notingher I, Jérôme R. Porous poly (α-hydroxyacid)/Bioglass® composite scaffolds for bone tissue engineering. I: preparation and in vitro characterisation. Biomaterials 2004, 25, 4185–4194.10.1016/j.biomaterials.2003.10.082Search in Google Scholar PubMed

[114] Liu X, Smith LA, Hu J, Ma PX. Biomimetic nanofibrous gelatin/apatite composite scaffolds for bone tissue engineering. Biomaterials 2009, 30, 2252–2258.10.1016/j.biomaterials.2008.12.068Search in Google Scholar PubMed PubMed Central

[115] Xu X, Capito RM, Spector M. Delivery of plasmid IGF-1 to chondrocytes via cationized gelatin nanoparticles. J. Biomed. Mater. Res. Part A 2008, 84, 73–83.10.1002/jbm.a.31372Search in Google Scholar PubMed

[116] Venugopal JR, Low S, Choon AT, Kumar AB, Ramakrishna S. Nanobioengineered electrospun composite nanofibers and osteoblasts for bone regeneration. Artif. Organs 2008, 32, 388–397.10.1111/j.1525-1594.2008.00557.xSearch in Google Scholar PubMed

[117] Li X, Xie J, Yuan X, Xia Y. Coating electrospun poly (ε-caprolactone) fibers with gelatin and calcium phosphate and their use as biomimetic scaffolds for bone tissue engineering. Langmuir 2008, 24, 14145–14150.10.1021/la802984aSearch in Google Scholar PubMed

[118] Ward AG, Courts A. Science and Technology of Gelatin, Academic Press: New York, 1977.Search in Google Scholar

[119] Hench LL, Wilson J. Clinical Performance of Skeletal Prostheses, Springer: Berlin, Germany, 1996.10.1007/978-94-011-0541-5Search in Google Scholar

[120] Hench LL, Wilson J. An Introduction to Bioceramics, World Scientific: Singapore, 1993.10.1142/2028Search in Google Scholar

[121] Li SH, De Wijn JR, Layrolle P, De Groot K. Synthesis of macroporous hydroxyapatite scaffolds for bone tissue engineering. J. Biomed. Mater. Res. 2002, 61, 109–120.10.1002/jbm.10163Search in Google Scholar PubMed

[122] Mozafari M, Moztarzadeh F, Rabiee M, Azami M, Maleknia S, Tahriri M, Moztarzadeh Z, Nezafati N. Development of macroporous nanocomposite scaffolds of gelatin/bioactive glass prepared through layer solvent casting combined with lamination technique for bone tissue engineering. Ceramics Int. 2010, 36, 2431–2439.10.1016/j.ceramint.2010.07.010Search in Google Scholar

[123] Armitage GC. Periodontal diagnoses and classification of periodontal diseases. Periodontology 2000. 2004, 34, 9–21.10.1046/j.0906-6713.2002.003421.xSearch in Google Scholar PubMed

[124] Hench LL. Bioceramics: from concept to clinic. J. Am. Ceramic Soc. 1991, 74, 1487–1510.10.1111/j.1151-2916.1991.tb07132.xSearch in Google Scholar

[125] Peter M, Binulal N, Nair S, Selvamurugan N, Tamura H, Jayakumar R. Novel biodegradable chitosan–gelatin/nano-bioactive glass ceramic composite scaffolds for alveolar bone tissue engineering. Chem. Engng J. 2010, 158, 353–361.10.1016/j.cej.2010.02.003Search in Google Scholar

[126] Dressler M, Dombrowski F, Simon U, Börnstein J, Hodoroaba V, Feigl M, Grunow S, Gildenhaar R, Neumann M. Influence of gelatin coatings on compressive strength of porous hydroxyapatite c eramics. J. Eur. Ceramic Soc. 2011, 31, 523–529.10.1016/j.jeurceramsoc.2010.11.004Search in Google Scholar

[127] Kim HW, Knowles JC, Kim HE. Porous scaffolds of gelatin–hydroxyapatite nanocomposites obtained by biomimetic approach: characterization and antibiotic drug release. J. Biomed. Mater. Res. Part B Appl. Biomater. 2005, 74, 686–698.10.1002/jbm.b.30236Search in Google Scholar PubMed

[128] Habraken W, De Jonge L, Wolke J, Yubao L, Mikos A, Jansen J. Introduction of gelatin microspheres into an injectable calcium phosphate cement. J. Biomed. Mater. Res. Part A 2008, 87, 643–655.10.1002/jbm.a.31703Search in Google Scholar PubMed

[129] Habraken W, Boerman O, Wolke J, Mikos A, Jansen J. In vitro growth factor release from injectable calcium phosphate cements containing gelatin microspheres. J. Biomed. Mater. Res. Part A 2009, 91, 614–622.10.1002/jbm.a.32263Search in Google Scholar

[130] Shu XZ, Liu Y, Palumbo F, Prestwich GD. Disulfide-crosslinked hyaluronan-gelatin hydrogel films: a covalent mimic of the extracellular matrix for in vitro cell growth. Biomaterials 2003, 24, 3825–3834.10.1016/S0142-9612(03)00267-9Search in Google Scholar

[131] Peattie RA, Pike DB, Yu B, Cai S, Shu XZ, Prestwich GD, Firpo MA, Fisher RJ. Effect of gelatin on heparin regulation of cytokine release from hyaluronan-based hydrogels. Drug Deliv. 2008, 15, 389–397.10.1080/10717540802035442Search in Google Scholar

[132] Hosack LW, Firpo MA, Scott JA, Prestwich GD, Peattie RA. Microvascular maturity elicited in tissue treated with cytokine-loaded hyaluronan-based hydrogels. Biomaterials 2008, 29, 2336–2347.10.1016/j.biomaterials.2008.01.033Search in Google Scholar

[133] Zhao F, Yin Y, Lu WW, Leong JC, Zhang W, Zhang J, Zhang M, Yao K. Preparation and histological evaluation of biomimetic three-dimensional hydroxyapatite/chitosan-gelatin network composite scaffolds. Biomaterials 2002, 23, 3227–3234.10.1016/S0142-9612(02)00077-7Search in Google Scholar

[134] Zhao F, Grayson WL, Ma T, Bunnell B, Lu WW. Effects of hydroxyapatite in 3-D chitosan–gelatin polymer network on human mesenchymal stem cell construct development. Biomaterials 2006, 27, 1859–1867.10.1016/j.biomaterials.2005.09.031Search in Google Scholar PubMed

[135] Liu H, Fan H, Cui Y, Chen Y, Yao K, Goh JC. Effects of the controlled-released basic fibroblast growth factor from chitosan-gelatin microspheres on human fibroblasts cultured on a chitosan-gelatin scaffold. Biomacromolecules 2007, 8, 1446–1455.10.1021/bm061025eSearch in Google Scholar PubMed

[136] Mandal BB, Mann JK, Kundu S. Silk fibroin/gelatin multilayered films as a model system for controlled drug release. Eur. J. Pharm. Sci. 2009, 37, 160–171.10.1016/j.ejps.2009.02.005Search in Google Scholar PubMed

[137] Gil ES, Frankowski DJ, Spontak RJ, Hudson SM. Swelling behavior and morphological evolution of mixed gelatin/silk fibroin hydrogels. Biomacromolecules 2005, 6, 3079–3087.10.1021/bm050396cSearch in Google Scholar PubMed

[138] Meng Z, Xu X, Zheng W, Zhou H, Li L, Zheng Y, Lou X. Preparation and characterization of electrospun PLGA/gelatin nanofibers as a potential drug delivery system. Colloids Surf. B Biointerf. 2011, 84, 97–102.10.1016/j.colsurfb.2010.12.022Search in Google Scholar PubMed

[139] Meng Z, Wang Y, Ma C, Zheng W, Li L, Zheng Y. Electrospinning of PLGA/gelatin randomly-oriented and aligned nanofibers as potential scaffold in tissue engineering. Mater. Sci. Engng C 2010, 30, 1204–1210.10.1016/j.msec.2010.06.018Search in Google Scholar

[140] Kasper FK, Kushibiki T, Kimura Y, Mikos AG, Tabata Y. In vivo release of plasmid DNA from composites of oligo (poly (ethylene glycol) fumarate) and cationized gelatin microspheres. J. Control. Release 2005, 107, 547–561.10.1016/j.jconrel.2005.07.005Search in Google Scholar PubMed

[141] Nejadnik MR, Mikos AG, Jansen JA, Leeuwenburgh SC. Facilitating the mineralization of oligo (poly (ethylene glycol) fumarate) hydrogel by incorporation of hydroxyapatite nanoparticles. J. Biomed. Mater. Res. Part A 2012, 100, 1316–1323.10.1002/jbm.a.34071Search in Google Scholar PubMed

[142] Holland TA, Tabata Y, Mikos AG. Dual growth factor delivery from degradable oligo (poly (ethylene glycol) fumarate) hydrogel scaffolds for cartilage tissue engineering. J. Control. Release 2005, 101, 111–125.10.1016/j.jconrel.2004.07.004Search in Google Scholar PubMed

[143] Kim K, Lam J, Lu S, Spicer PP, Lueckgen A, Tabata Y, Wong ME, Jansen JA, Mikos AG, Kasper FK. Osteochondral tissue regeneration using a bilayered composite hydrogel with modulating dual growth factor release kinetics in a rabbit model. J. Control. Release 2013, 168, 166–178.10.1016/j.jconrel.2013.03.013Search in Google Scholar PubMed PubMed Central

[144] Kempen DH, Lu L, Heijink A, Hefferan TE, Creemers LB, Maran A, Yaszemski MJ, Dhert WJ. Effect of local sequential VEGF and BMP-2 delivery on ectopic and orthotopic bone regeneration. Biomaterials 2009, 30, 2816–2825.10.1016/j.biomaterials.2009.01.031Search in Google Scholar PubMed

[145] Drbohlavova J, Adam V, Kizek R, Hubalek J. Quantum dots – characterization, preparation and usage in biological systems. Int. J. Mol. Sci. 2009, 10, 656–673.10.3390/ijms10020656Search in Google Scholar PubMed PubMed Central

[146] Yong K-T, Ding H, Roy I, Law W-C, Bergey EJ, Maitra A, Prasad PN. Imaging pancreatic cancer using bioconjugated InP quantum dots. ACS Nano 2009, 3, 502–510.10.1021/nn8008933Search in Google Scholar PubMed PubMed Central

[147] Choi HS, Liu W, Liu F, Nasr K, Misra P, Bawendi MG, Frangioni JV. Design considerations for tumour-targeted nanoparticles. Nat. Nanotechnol. 2010, 5, 42–47.10.1038/nnano.2009.314Search in Google Scholar PubMed PubMed Central

[148] Lovrić J, Bazzi HS, Cuie Y, Fortin GR, Winnik FM, Maysinger D. Differences in subcellular distribution and toxicity of green and red emitting CdTe quantum dots. J. Mol. Med. 2005, 83, 377–385.10.1007/s00109-004-0629-xSearch in Google Scholar PubMed

[149] Li K, Chen J, Bai S, Wen X, Song S, Yu Q, Li J, Wang Y. Intracellular oxidative stress and cadmium ions release induce cytotoxicity of unmodified cadmium sulfide quantum dots. Toxicol. In Vitro 2009, 23, 1007–1013.10.1016/j.tiv.2009.06.020Search in Google Scholar PubMed

[150] Byrne SJ, Williams Y, Davies A, Corr SA, Rakovich A, Gun’ko YK, Rakovich YP, Donegan JF, Volkov Y. “Jelly dots”: synthesis and cytotoxicity studies of CdTe quantum dot–gelatin nanocomposites. Small 2007, 3, 1152–1156.10.1002/smll.200700090Search in Google Scholar PubMed

[151] Prasad BR, Nikolskaya N, Connolly D, Smith TJ, Byrne SJ, Gérard VA, Gun’ko YK, Rochev Y. Long-term exposure of CdTe quantum dots on PC12 cellular activity and the determination of optimum non-toxic concentrations for biological use. J. Nanobiotechnol. 2010, 8, 7.10.1186/1477-3155-8-7Search in Google Scholar PubMed PubMed Central

[152] Gérard VA, Maguire CM, Bazou D, Gun’ko YK. Folic acid modified gelatine coated quantum dots as potential reagents for in vitro cancer diagnostics. J. Nanobiotechnol. 2011, 9, 50.10.1186/1477-3155-9-50Search in Google Scholar PubMed PubMed Central

[153] Wong C, Stylianopoulos T, Cui J, Martin J, Chauhan VP, Jiang W, Popović Z, Jain RK, Bawendi MG, Fukumura D. Multistage nanoparticle delivery system for deep penetration into tumor tissue. Proc. Natl Acad. Sci. 2011, 108, 2426–2431.10.1073/pnas.1018382108Search in Google Scholar PubMed PubMed Central

[154] Jiang S, Eltoukhy AA, Love KT, Langer R, Anderson DG. Lipidoid-coated iron oxide nanoparticles for efficient DNA and siRNA delivery. Nano Lett. 2013, 13, 1059–1064.10.1021/nl304287aSearch in Google Scholar PubMed PubMed Central

[155] Tse WH, Gyenis L, Litchfield DW, Zhang J, Engineering large gelatin nanospheres coated with quantum dots for targeted delivery of human osteosarcoma with enhanced cellular internalization. In 14th IEEE International Conference on Nanotechnology 2014 Aug 18. IEEE. pp. 672–67710.1109/NANO.2014.6967978Search in Google Scholar

[156] Won Y-W, Kim Y-H. Preparation and cytotoxicity comparison of type A gelatin nanoparticles with recombinant human gelatin nanoparticles. Macromol. Res. 2009, 17, 464–468.10.1007/BF03218893Search in Google Scholar

Received: 2016-1-13
Accepted: 2016-6-24
Published Online: 2016-9-26
Published in Print: 2017-4-1

©2017 Walter de Gruyter GmbH, Berlin/Boston

This article is distributed under the terms of the Creative Commons Attribution Non-Commercial License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Downloaded on 10.5.2024 from https://www.degruyter.com/document/doi/10.1515/ntrev-2016-0009/html
Scroll to top button