Next Article in Journal
Tunable Ag Nanocavity Enhanced Green Electroluminescence from SiNx:O Light-Emitting Diode
Previous Article in Journal
Effect of Titanium Dioxide Particles on the Thermal Stability of Silica Aerogels
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cancer Treatment Using Nanofibers: A Review

by
Muhammad Qamar Khan
1,*,
Muhammad Abbas Alvi
1,
Hafiza Hifza Nawaz
2 and
Muhammad Umar
2,*
1
Department of Textile Engineering, School of Engineering and Technology, National Textile University, Faisalabad 37610, Pakistan
2
Department of Materials, The University of Manchester, Manchester M13 9PL, UK
*
Authors to whom correspondence should be addressed.
Nanomaterials 2024, 14(15), 1305; https://doi.org/10.3390/nano14151305
Submission received: 19 June 2024 / Revised: 22 July 2024 / Accepted: 1 August 2024 / Published: 2 August 2024

Abstract

:
Currently, the number of patients with cancer is expanding consistently because of a low quality of life. For this reason, the therapies used to treat cancer have received a lot of consideration from specialists. Numerous anticancer medications have been utilized to treat patients with cancer. However, the immediate utilization of anticancer medicines leads to unpleasant side effects for patients and there are many restrictions to applying these treatments. A number of polymers like cellulose, chitosan, Polyvinyl Alcohol (PVA), Polyacrylonitrile (PAN), peptides and Poly (hydroxy alkanoate) have good properties for the treatment of cancer, but the nanofibers-based target and controlled drug delivery system produced by the co-axial electrospinning technique have extraordinary properties like favorable mechanical characteristics, an excellent release profile, a high surface area, and a high sponginess and are harmless, bio-renewable, biofriendly, highly degradable, and can be produced very conveniently on an industrial scale. Thus, nanofibers produced through coaxial electrospinning can be designed to target specific cancer cells or tissues. By modifying the composition and properties of the nanofibers, researchers can control the release kinetics of the therapeutic agent and enhance its accumulation at the tumor site while minimizing systemic toxicity. The core–shell structure of coaxial electrospun nanofibers allows for a controlled and sustained release of therapeutic agents over time. This controlled release profile can improve the efficacy of cancer treatment by maintaining therapeutic drug concentrations within the tumor microenvironment for an extended period.

1. Introduction

Cancer is the second leading cause of death in the world, surpassed only by heart diseases. In the United States alone, it results in approximately 1620 deaths daily, highlighting its significant impact on public health. This statistic underscores the urgent need for continued research, enhanced prevention strategies, and the development of more effective treatments. Cancer has emerged as a formidable global health challenge, with its burden escalating over the years. The incidence and mortality rates associated with this disease have shown a steady upward trend. For instance, the global number of new cancer cases surged from approximately 14 million in 2010 to 19.3 million in 2020, reflecting a substantial increase of 37.8%. Similarly, cancer-related deaths climbed from 8.2 million in 2010 to 9.9 million in 2020, underscoring the disease’s devastating impact. These figures not only highlight the growing prevalence of cancer but also emphasize the urgent need for comprehensive prevention, early detection, and effective treatment strategies to combat this global health crisis [1].
Cancer encompasses a diverse group of diseases, with prevalent types receiving global attention including but not limited to brain, breast, kidney, melanoma, and liver cancers as mentioned in Figure 1. Each type of cancer originates in specific tissues and exhibits unique characteristics, affecting various organs. Understanding these cancer types is crucial for effective diagnosis, treatment, and research efforts to improve outcomes and reduce the global burden of cancer. A comprehensive awareness of and research in these areas contribute to advancements in oncology, fostering better strategies for prevention, early detection, and personalized therapies [2].
The higher death rates for cancer observed in high-income countries can be linked to advanced healthcare infrastructures that enable more thorough diagnosis and reporting, alongside factors like increased life expectancy, and lifestyle factors like diet and tobacco use. Cancer is a complex disease influenced by a multitude of factors. Environmental factors play a significant role, with tobacco smoke, radiation, pollution, and occupational hazards being primary culprits. Exposure to these elements can increase the risk of developing various cancer types. Genetic factors also contribute, as inherited mutations can predispose individuals to certain cancers. While not all cancers are hereditary, family history remains a crucial consideration. Lifestyle factors significantly impact cancer risk, with diet, physical activity, obesity, alcohol consumption, and hormone replacement therapy being key influences. A diet rich in fruits and vegetables combined with regular exercise can help mitigate the risk, while unhealthy habits can increase it. Other factors such as age, immune system function, and infectious agents also play a part in cancer development. It is essential to note that the interplay of these factors varies across different cancer types, and while genetics can influence susceptibility, lifestyle modifications and environmental protections remain crucial in cancer prevention. In contrast, lower-income countries often face challenges in accessing quality healthcare, resulting in underdiagnosis and underreporting, contributing to apparent lower cancer death rates [3].
Nanofiber-based drug delivery systems offer a promising approach for achieving controlled and sustained drug release. However, the successful translation of these systems from bench to bedside necessitates a comprehensive understanding of their degradation profile and pharmacokinetic/pharmacodynamic (PK/PD) behavior. The rate and mechanism of nanofiber degradation significantly influence drug release kinetics, with biodegradable polymers offering the advantages of controlled release and the eventual elimination of the carrier system. Concurrently, elucidating the absorption, distribution, metabolism, and excretion of the encapsulated drug is crucial for optimizing dosage regimens and predicting therapeutic outcomes. By meticulously characterizing these parameters, researchers can design nanofiber-based systems that effectively deliver therapeutic agents while minimizing adverse effects.
Metastatic cancer poses a significant threat as it can spread from its original site to other parts of the body, forming secondary tumors. The majority of cancer-related mortality is often attributed to metastasis, as it makes treatment more challenging and increases the complexity of managing the disease. Effective strategies for early detection and intervention are crucial in addressing metastatic cancer and improving patient outcomes [4].

2. Current Cancer Treatments and Their Limitations

For effective treatment of malignant tissues, comprehensive knowledge of the affected tissue, its condition, and treatment methodologies is essential. Various techniques, including hyperthermia/ultrasound, hyperthermia/chemotherapy, hyperthermia/radiotherapy, and hyperthermia/microwave, are utilized in cancer treatment (Figure 2) [5]. However, these machine-driven strategies may induce toxicity in specific tissues [5].
To date, seventy types of conventional anticancer drugs have been utilized as a therapy for cancer affected people. The utilization of these synthetic anticancer drugs can lead to harmful effects like hair loss, blood disorders, and nervous system issues [6,7]. While functionalized magnetic nanoparticles hold significant promise in biomedical applications, it is essential to acknowledge their limitations. The inability of these nanoparticles to traverse the blood–brain barrier poses a considerable challenge for targeting central nervous system diseases. Additionally, concerns regarding potential liver toxicity due to nanoparticle accumulation cannot be overlooked. Rigorous safety assessments and careful dose selection are crucial in mitigating these risks. To fully harness the potential of these nanoparticles, future research should prioritize developing strategies to enhance biodistribution and address safety concerns.
Several products extracted from plants like vincristine [8], vinblastine [9,10], paclitaxel [11,12], docetaxel [13,14], topotecan [15,16], irinotecan [17,18], flavopiridol, acronyciline, bruceantin, and thalicarpin [19,20,21,22] have been explored as natural anticancer agents. These anticancer agents are useful but also have numerous limitations such as low solubility, poisonousness, a shorter halftime, the dynamic release of anti-cancer drugs, and to the possibility of harming healthy cells [23].
The effective targeting of anticancer agents specifically to diseased tissues necessitates their navigation through various physiological barriers, including cellular membranes, extracellular spaces, bloodstreams, and specific organs. The unintended effects of these agents on healthy tissues due to non-specific distribution can lead to adverse side effects and escalate the cost of cancer treatment. This underscores the need for precision in drug delivery to maximize therapeutic efficacy while minimizing harm and financial burden.
Anticancer drug delivery systems have drawn attention due to their ability to (1) adjust the quantity of drug loaded [24], (2) enable precise, localized release of drugs, reducing cytotoxic effects on healthy tissues [25], (3) incorporate multiple therapeutic agents [26], (4) prevent premature burst release in targeted tissues [25], (5) ensure consistent release of drug payloads in targeted areas [27], and (6) offer biodegradable properties, enhancing safety and effectiveness.
Several considerations are crucial when selecting drug delivery carriers, including their structure, surface characteristics, and chemical composition. Currently, a diverse range of carriers are used for drug delivery, such as solid lipid nanoparticles, liposomes [28,29,30], compounds of silicate [31], compounds of magnetite [32,33], compounds of natural and synthetic polymers [34], quantum dots [35], carbon compounds [36,37], and metallic nanoparticles [38,39]. Additionally, various forms like patches [40], intravaginal rings [41], fibers [42], film [43], tablets [44], hydrogels [45], cervical caps [46], and sponges [47] are employed. While some materials primarily serve as carriers, others offer additional benefits in cancer treatment, underscoring the importance of selecting the appropriate carrier based on specific therapeutic needs [31,48,49].
The various types of carriers demonstrate different degrees of effectiveness in drug delivery as mentioned in Figure 3. However, for both natural and biomedical applications, carriers must exhibit essential characteristics: (1) the manufacturing process should be straightforward and cost-effective; (2) the materials used for the carrier’s synthesis, including solvents and precursors, must be non-toxic and affordably priced; and (3) carriers must be biocompatible, renewable, and biodegradable, aligning with safety and environmental standards.

3. Importance of Nanofibers in Cancer Treatment

Nanomaterials have received much more attention as potential carriers in clinical and biomedical applications [50,51,52,53,54,55,56,57,58,59] due to their outstanding properties like the convenient processes used for their production at large scales, their enhanced mechanical characteristics, extremely permeable structure, high surface to volume ratio, analog to extra cellular model, and adjustable combination procedure [60,61,62]. Nanofibers are versatile materials with a broad spectrum of applications in the biomedical field. As illustrated in Figure 4, they excel in drug delivery, tissue engineering, and wound healing. Moreover, nanofibers’ high surface area makes them ideal candidates for biosensor development. By immobilizing biomolecules on their surfaces, these biosensors can detect cancer biomarkers with enhanced sensitivity, facilitating early-stage cancer diagnosis. Nanofibers are also efficiently used for medical applications as mentioned in the Figure 4 below.
These applications include bone tissue engineering [63], medicinal compositions [64], scaffolds and artificial organs [65], wound dressing [50,66,67,68], drug delivery and medical engineering [69], antifungal [70] and anticancer [71] agents, antibacterial dressings [72], antibiotics [66], antimicrobials [73], medical prostheses [74], and postoperative implants [75,76].
Compounds of one dimension with thicknesses of 50 to 500 nm and length–width proportions of more than 1:20 that are composed of melts or polymer solutions are called nanofibers. Nanofibers [77,78,79,80,81,82,83,84,85,86] have permeable walls, are multi-layer and have ribbon, multi-channel, single layer constructions, necklace-like and weblike structures and core–shell structures [87,88,89,90,91,92]. The composition and breadth of nanofibers can be changed by changing numerous parameters like the polymer solution, viscosity, and method of preparation. Nanomaterials are the only material that can encapsulate more of a drug due to their specific size and structure [93]. Nonetheless, by using nanofibers with a single layer as medication transporters, the primary burst deliverance of medications may occur when a treatment is started [94]. On the other hand, the core shell and multi-layer nanofibers never show the phenomenon of self-releasing drugs [95,96,97] because, in core–shell nanofibers and multi-layer nanofibers, drugs are packed in the internal core or layers of fibers [98,99].
Nanofibers are also a cause of delay in drug delivery or release in core–shell nanofibers or multi-layer nanofibers due to their hydrophobic characteristics when they are organized in multi shell layers [100,101].
The restorative viability of anticancer medicinal nanofibers classification is expanded by (A) planned multi-drug/nanofibers preparation [102,103], (B) the preparation of nanofibers from such polymers that can sense both temperature and pH [55,67,86,104,105,106,107,108,109], (C) the fabrication of magnetic nanomaterials for the utilization of drug delivery and hyperthermia treatment [32,110]. The death rate due to cancer is increasing due to manufacturing/designing these carriers.

4. Commonly Used Polymers for Making Nanofibers for Cancer Treatment

Various kinds of polymers, as mentioned in Figure 5, have been accounted for in this review that can capture and convey cancer drugs to the particular area with continuous release. The decision of which drug, producing strategy, polymer, and post-modifications should be used are vital to consider when one needs to manage cancer-affected cells. Currently, the following polymers are being utilized in nanofibers-based disease treatment frameworks and systems.

4.1. PLGA (Poly(lactic-co-glycolic Acid))

Biodegradable and biocompatible, PLGA is a copolymer that is frequently employed in the manufacturing of nanofibers for the treatment of cancer. It is a well-known contender for medication delivery systems due to its capacity for controlled release. PLGA can be used in a variety of biomedical applications since it breaks down in the body into non-toxic metabolites [111].

4.2. PVA (Polyvinyl Alcohol)

PVA is a synthetic polymer that dissolves in water and is frequently used in electrospinning procedures to create nanofibers that are used in cancer therapy applications. Its advantageous qualities make it biocompatible and an efficient medication delivery system carrier [112].

4.3. PEG (Polyethylene Glycol)

Water-soluble and multipurpose, PEG is renowned for its superior biocompatibility. PEG improves the stability and biocompatibility of nanofibers and is frequently employed in conjunction with other polymers, which makes it appropriate for use in cancer treatment applications [113].

4.4. PU (Polyurethane)

PU is a biocompatible, flexible polymer that is used to make nanofibers for tissue engineering and medication delivery. Its application in the treatment of cancer is facilitated by its adaptability and drug compatibility [114].

4.5. Gelatin

Collagen is the natural source of the protein polymer known as gelatin. It is frequently mixed with other polymers to improve the mechanical characteristics and biocompatibility of nanofibers used in tissue engineering and cancer treatment [115].

4.6. PCL (Polycaprolactone)

PCL is a biodegradable polyester that is frequently utilized in cancer therapy nanofiber applications. PCL is appropriate for tissue engineering and sustained release medication delivery systems due to its slow rate of degradation [116].

4.7. Cellulose Acetate

In the presence of sulfuric acid, cellulose acetate can be achieved through the chemical reaction of cellulose with acetic anhydride and CH3COOH. A solution of cellulose acetate is needed for the manufacturing of fibers, which totally depends on degree of substitution in the cellulose acetate solution. A solution of cellulose acetate with the degree of substitution 2 or 2.5 is frequently soluble in different solution like dioxane, methyl acetate, and acetone and if the solution of cellulose acetate contains a degree of substitution greater than, 2.5 then it should be easily soluble in dichloromethane [117]. Cellulose and its mixes are often utilized to make nanofibers which need high absorbency [118]. Cellulose-based nanofibers have great advantages in the medical field for wound dressing and drug distribution owing to their enhanced characteristics like ecological and biological [119]. Suwantong et al. showed curcumin-loaded nanofiber mats made of cellulose acetate. When they placed these curcumin drug-based nanofibers on pig skin, these nanofibers displayed a slower discharge of curcumin. After checking the test result, it was found that 90 to 95% drug was discharged. Cellulose and its derivatives are valued for their exceptional absorbency, making them suitable for numerous applications. At the nanoscale, cellulose demonstrates remarkable mechanical properties, including a high tensile strength, stiffness, and flexibility. These attributes are particularly noteworthy as they persist across a wide range of moisture conditions, rendering cellulose nanomaterials highly versatile and desirable for developing advanced materials with tailored functionalities [120]. Cellulose acetate-based nanofibers displayed nontoxic nature against human dermal fibroblast [121].

4.8. Poly (Vinyl Alcohol)

Poly (viny alcohol) is utilized as a drug delivery agent and is known to be a biodegradable and biocompatible polymer [122]. The analysis of cancer in the initial phases is very crucial. Zhao et al. demonstrated that nanofibers which have diameters 460 nm like polyethyleneimine and PVA nanofibers which were controlled by hyaluronic acid can effectively analyze and capture cancer cells. The generated nanofibers were smooth and displayed an excellent efficiency [123]. Fan et al. used folic acid-based receptors to analyze the detection of cancer cells. They synthesized nanofibers like polyethyleneimine and poly vinyl alcohol and folic acid to check for the early stages of cancer [124]. Poly vinyl alcohol and core–sheath nanofibers loaded with DOX are utilized for the treatment of cancer cells in ovaries with a sustained discharge of drugs. These nanofibers were tested against SKOV3 disease cells and presented great outcomes [125].

4.9. Poly (Styrene)

Polystyrene is an engineered polymer and has an exceptionally low biodegradation rate. Iron oxide nanoparticles-based poly styrene nanofibers can be successfully utilized for the hyperthermic treatment of cancer cells [126]. Hyperthermia treatment is carried out to kill the dangerous cancer cells with the help of heat in a localized area. Iron oxide nanoparticles-based polystyrene nanofibers produce heat when they were uncovered to another magnetic field and destroy human ovarian cells completely in 10 min by becoming attached to their exterior part. Poly styrene nanofibers contain collagen on their surface which helps in capturing more cancer cells to these nanofibers. Against SKOV3 different results have been checked but the most considerable method for results verification was the alternating heating produced method. MDR Leukemia 562 cells have been treated with nanofibers (poly (N-isopropylacrylamide)-co PS) loaded with Daunorubicin; these nanofibers efficiently decreased the cells MDR and also increased the acceptance of this drug [127].

4.10. Poly (Hydroxyalkanoate)

Poly (hydroxyalkanoate) are green materials which can be formed by many microbes [128]. Because of their outstanding characteristics, these nanofibers are used in medical applications [129]. Poly (hydroxyalkanoate) has been shown to be acceptable for clinical purposes, and they are excellent for use in cell multiplication and tissue recovery without the danger of producing cancer cells [130]. The discharge rate of a drug is totally depended on the parameters of electrospinning like size and structure [131]. Due to the unevenness of the surface, poly (hydroxyalkanoate) nanofibers were found to display a reduced crystallinity in comparison with bulk film but to express an amplified angle of contact when checked against bulk film [132].
Cationic peptides have shown the greatest anticancer possibility but in the result of depolymerization of poly (hyroxyalkanoate)-conjugated R10 and R-3- hydroxy decanoic acid R10 are obtained with cationic peptides to increase the efficiency of peptides against cancer. Extra cytotoxic effects have been observed due to the conjugated peptides compared to unconjugated peptides in the contradiction of T cells leukemia, human pancreatic carcinoma, lung carcinoma, human glioma, and colorectal cancer [133]. Poly (hydroxyalkanoate) has been used as a nanocarrier for cellular treatment [134]. Five different forms of drugs have been incorporated into nanofibers (collagen peptides, Poly(3-hydroxybutyrate-co-3-hydroxyvalerate) used against gastric cancer cells [135]. Therefore, these nanofibers are mostly used in the medical field due to their green nature and ecofriendly behavior. But, the commercialization of these nanofibers is very difficult because these polymers are very costly due their preparation from microbes [136,137,138].

4.11. Peptides

Peptides are small chain monomers that are interconnected with amide bonds. Zhang et al. [139] made RADA16-I peptide electrospun nanofibers that exhibited excellent characteristics for three dissimilar categories of ovarian cells. The oral path is more challenging for the delivery of intact peptides because of the acidic pH in the digestive system (stomach). Arginine–glycine-aspartic acid relates to peptide nanofibers that can effectively transfer (curcumin) to the targeted area. Embryonic kidney, breast cancer cells and liver carcinoma cells are three diverse types of cancer cells which have been efficiently analyzed with the help of curcumin-based loaded peptides nanofibers [140]. L-Peptides and D-Peptides’ stability was checked by Yang et al. both in vitro and in vivo, and different behaviors were shown when they were used to vaccinate mice which was likely due to the different bio supply [141].

4.12. Chitosan

Chitosan can be prepared from many materials like nanobeads [142], sponges [143], membranes [144,145], microparticles [146], hydrogels [147], scaffolds [148], nanoparticles [149], and nanofibers [150]. Chitosan is a widely studied naturally occurring polymer broadly used for medical applications, medicine transfer, and tissue engineering [151]. Nanofibers of chitosan are mostly used in clinical applications [152,153]. Ardeshirzadeh et al. demonstrated the feasibility of DOX drug-based Chitosan, polyethylene oxide, and graphene oxide nanofibers for the efficient treatment of cancer. However, a π-π marking between DOX and graphene oxide caused a reduced medicine loading proficiency in Chitosan, polyethylene oxide, and in graphene oxide fibers than Chitosan/polyethylene oxide electro spun drug loaded filaments. These nanofibers displayed pH-dependent release of a drug, and when the pH was 7.4, constant discharge of drug was noticed. An in vitro cytotoxicity examination exhibited an improved resistance to the growth of A549 cells after 72 h in comparison with a free DOX drug which could be credited to the slow and maintained discharge of the medicine [154]. However, anti-cancer and other different characteristics of Chitosan have been demonstrated well at this point, but the important thing is the continuous discharge of drug for long time period as Chitosan is water loving and its expanding performance permits a simple diffusion of medication from filaments. It has additionally been realized that chitosan cannot be electrospun effectively without any secondary polymers and the mostly of them are water loving like poly vinyl alcohol and polyethylene oxide, etc. Therefore, the continuous discharge of a drug for long time period is still under investigation through post modifications or alterations to further polymers.
Their also some drugs used for the cancer treatments as mentioned in Table 1.

5. Methods for Producing Nanofibers

Nanofibers can be prepared by various methods such as force spinning [155], interfacial polymerization [156], phase separation [157], self-assembly [158], wet spinning [159], drawing [160], template melt extrusion [161], thermal induced phase separation [162], melt blowing [162], template synthesis [163], chemical vapors deposition [164], and electrospinning [165]. The multi-needle and multiple-jet needless spinnerets techniques of electrospinning are most commonly utilized in the research work and industrial services for the production of nano and microfibers at small and large scale [166,167,168].
In 1861–1903 Morton and Cooley developed the advanced electrohydrodynamic shower strategy to scatter liquids by electrostatic power [169]. Until 1990, this strategy did not achieve much consideration by experts. On the other hand, between 1934 and 1944 and furthermore between 1855 and 1944, there were a few authorizations on the electrospinning arrangement for the creation of polymeric strands [170,171,172,173].

Production of Nanofibers through Electrospinning

In the system used for electrospinning, polymer solutions are softened under high electric power and the supply is squeezed out from a jet and then it dries and is collected on drum as shown in Figure 6. Different parameters impact the electrospinning system, like the polymer solution and their characteristics, electrospinning machine characteristics, environment, and surroundings [174].
There are some parameters while discussing nanofiber production through electrospinning as mentioned in Table 2.

6. Approaches for Controlled Drug Release from Nanofibers

6.1. Control of Drug by Polymer

The polymers electrospun into nanomaterials have large effect on drug discharge and control. Different polymers show different discharge mechanisms for drugs. Hydrophilic drug-loaded nanofibers are the best polymers for acquiring a burst discharge effect. Hydrophilic nanofibers such as PVA have been allowed for the development of a quick dissolving drug conveyance nanofiber to transport caffeine and riboflavin drugs as mentioned in [175]. These nanofibers delivered the medications in a ruptured way because of the high water-loving nature of the polymer. Within 60 s, 100 percent of the caffeine and nearly 40 percent of the riboflavin was discharged [175]. Likewise, nanofibers of polyvinylpyrrolidone have shown an 84.9 percent ibuprofen discharge in just 20 s for a quick dissolving oral medication conveyance system [176]. Due to the nature of the fibers, PVP nanofibers loaded with Loratadine showed a high burst discharge [177]. A very strong burst effect has also been observed in the case of a burn wound when using nanofibers for a rapid pain-relieving action [178]. Water-repellent polymers can be utilized when a maintained discharge of medicines is expected from nanofibers. Hydrophobic co-polymers such as poly (lactic-co-glycolic acid) have exhibited sustained anticancer drug discharge for 60 days [179]. Nanofibers like PCL, poly lactic acid, and PVA express diverse phenomena in drug discharge because of the character/structure of the polymers [180]. Consequently, the selection of an appropriate polymer for favorable drug discharge is crucial. The mechanism of controlling release of drugs by polymers is illustrated in Figure 7.

6.2. Control of Drug by Structure

Changing the electrospinning parameters (drug loading quantity, mixing/blend, nanofibers layers, size of fibers and affinity of drug with polymers) also changes the structure of fibers. A small modification to the structure of nanofibers can alter the rate of medicine release. Two different structures of fibers loaded with the same amount of drug show two different releasing phenomenon because of a change in the dissemination path. For example, the rate drug release by flat nanofibers is higher than that of rounded nanofibers because there is less distance between the drug and the edges. Recently, round and flat nanofibers have been found to show both slow and fast releases of drugs when loaded with diclofenac sodium [181]. The pore size of nanofibers is another significant boundary to control the medicine discharge. A bigger pore size leads to burst discharge because of the ease of medication dissemination. Polyhydroxyalkanotes nanofibers filled with paclitaxel anticancer drug showed a quicker discharge when the pore size was bigger [131]. Nanofibers with a larger diameter show more diffusion of drugs in comparison to nanofibers with small diameters. Various diameters of nanofibers can be achieved by altering the settings of electrospinning. The speed of the discharge of a drug from nanofibers of poly l-lactic acid show faster and slower discharges from greater to smaller diameters [182]. The development of core–sheath nanofibers is another way to reduce the amount of a drug released by nanofibers. When maintaining all the other parameters (structure, pore size, porosity and diameters), polycaprolactone nanofibers presented less discharge of drugs in comparison to blended nanofibers. Core–sheath fibers showed a drug release of 34% while the blended nanofibers showed a 60% drug discharge in the same time frame [183]. Therefore, by changing the electrospinning conditions, drug release can be controlled as per the requirements by modifying the structure of nanofibers as mentioned above. The difference between controlled and uncontrolled drug delivery mechanism is illustrated in Figure 8.

6.3. Blend-Dependent Release

Mixing one or more polymers together can be used as a compelling tool to make different drug discharge profiles. In an investigation, just a 1.5% discharge of ciprofloxacin has been observed in 40 days by poly (methyl methacrylate) nanofibers. The drug-releasing phenomenon of ciprofloxacin was changed by blending some water-loving polymers like chitosan, PVA, and poly (ethylene oxide). The mixing of 10 percent chitosan with poly (methyl methacrylate) showed a constant discharge of ciprofloxacin, the mixing of PMMA with poly (ethylene oxide) exhibited burst discharge of ciprofloxacin, and mixing with PVA depicted a combination of both constant and burst discharge of ciprofloxacin [177]. The mixing of different polymers has been proven to be very useful in the phenomenon of drug discharge.

6.4. Drug-Dependent Release

The discharge of medicine from different nanofibers relies on the different drug characteristics. A hydrophobic polymer is more suitable for use with a hydrophobic drug while a hydrophilic drug is more suitable for use with a hydrophilic polymer. Several issues arise like the drug becoming attached to the outer layer of fibers which are enclosed and burst discharge due to mixing a hydrophilic polymer or drug with a hydrophobic polymer or drug [184,185].
The final discharge of medicines/drug highly depends on the quantity of drug loaded in the nanofibers. Nanofibers loaded with a large mount of a drug are needed for early burst discharge [186]. The expansion of molecular mass and collaboration among polymers and drug maintains the steady discharge of a drug [187]. To achieve both constant and burst discharge simultaneously, a combination of hydrophilic and hydrophobic drugs are useful; water-repellent drug will discharge slower and water-loving drugs will discharge quickly, and nanofibers composed of gelatin and PLGA nanofibers loaded with effective anticancer drugs (camptothecin and doxorubicin hydrochloride), which are the combination of both hydrophilic and hydrophobic anticancer drugs, have shown a high-level anticancer effect against HepG-2 cancer cells because of the constant discharge of doxorubicin hydrochloride and the burst discharge of camptothecin [188]. Thus, the hydrophobic and hydrophilic character of these drugs plays a vital role in the discharge of drug and also the polymer to which these drugs are attached.

6.5. Post Modification Release of Drug

The continuous discharge of medication can be accomplished by the post adjustment of the composed nanofibers. There is a possible solution to controlling the drug discharge. Chemical and plasma alterations to nanomaterial can initiate the medication discharge [189,190]. Different post-change methods have been used by analysts to alter the medication discharge from nanofibers, like interconnecting them with synthetic compounds or particles through heat action and ions. These techniques are helpful to give permanency to altered nanofibers against water which decreases the medication discharge. For instance, nanofibers of PVA adjusted using the interconnected method with methanol or post-heat treatment have been displayed to lessen the disintegration in water, thus physical firmness can be attained [191]. PVA nanofibers loaded with non-flammable drug dexapanthenol, link up with ammonium peroxydisulphate have been observed to encourage continuous discharge of drug [192]. Likewise, the burst discharge of PVA nanofibers loaded with ketoprofen was shown to decrease when they were linked with methanol [193]. In this manner, an appropriate assortment of post modification treatment can be utilized to alter the medicine discharge from nanofibers.
Controlled drug release from nanofibers is critical for optimizing therapeutic efficacy. Various strategies can be employed to modulate drug release kinetics, including polymer selection, fiber morphology, and drug loading. To achieve targeted drug delivery, stimuli-responsive nanofibers have emerged as a promising approach. By incorporating pH-sensitive or thermoresponsive polymers into the nanofiber matrix, drug release can be triggered by changes in the physiological environment. For instance, pH-sensitive nanofibers can be designed to release their therapeutic payload in the acidic tumor microenvironment, enhancing drug concentration at the target site. Similarly, thermoresponsive nanofibers can be formulated to respond to temperature variations within the body, enabling drug release in specific tissues or organs. These advanced strategies offer the potential to improve drug delivery efficiency, reduce systemic toxicity, and enhance therapeutic outcomes [194].

7. Future Directions

Electrospinning is a process used to create ultrafine fibers by applying a high-voltage electric field to a polymer solution or melt as illustrated in Figure 9. In coaxial electrospinning, two or more solutions are spun simultaneously through separate concentric spinnerets, resulting in core–shell or multilayered fibers shown below.
In coaxial electrospinning for cancer treatment, the core–shell structure allows for the encapsulation of therapeutic agents, such as chemotherapeutic drugs, proteins, or nanoparticles, within the core of the nanofibers. The shell, typically made of a biocompatible polymer, provides protection and allows for the controlled release of the therapeutic agent.
Coaxial electrospinning offers several potential advantages for cancer treatment, including an enhanced drug loading capacity, the improved stability and bioavailability of therapeutic agents, and the ability to tailor the properties of the nanofibers for specific applications.
Overall, coaxial electrospinning holds promise as a novel approach for the development of advanced drug delivery systems for cancer treatment, offering the potential for improved therapeutic outcomes and reduced side effects compared to conventional treatment modalities. However, further research and development is needed to optimize the design and fabrication of coaxial electrospun nanofibers for clinical applications.

8. Conclusions

The traditional method of conveying a drug to the affected area relies on the control and continuous drug discharge of the drug. The clinical sciences have been supported by utilizing nanotechnology-based frameworks. The designated and continuous drug discharge achieved with these nano-transporters shows that it is a promising methodology. Nanofibers show supported drug discharge compared to free drugs and, consequently, are more effective against cancer cells. The choice of the drug and polymer is vital in deciding on the right blend for supported drug conveyance. Nanofibers can carry a high quantity of various anticancer medications; naturally extracted materials are helpful treating cancer-affected cells and magnetic nanoparticles have also proved their worth in blends with other cancer-removing drugs. The conveyance of a hydrophilic and hydrophobic drug like curcumin can generally be controlled with the assistance of nanofibers. Therefore, nanofibers have mind blowing benefits in protecting humans against cancer diseases.

Author Contributions

M.Q.K.: Conceptualization, methodology, data curation, visualization, formal analysis, investigation, supervision, project administration funding acquisition and writing-original draft. M.A.A.: Conceptualization, methodology, data curation, software, validation, formal analysis, visualization, investigation, and writing-original draft. H.H.N.: Writing-review and editing, investigation, and formal analysis. M.U.: Conceptualization, methodology, writing-review and editing, visualization, resources, investigation, supervision, project administration and funding acquisition. All authors have read and approved the final version.

Funding

This research work was supported by a grant from Global Challenge Research Fund (GCRF), UK Research Innovation and the Henry Royce Institute for Advanced Materials, funded through EPSRC grants EP/R00661X/1, EP/P025021/1, and EP/P025498/1.

Data Availability Statement

Not applicable.

Acknowledgments

We are grateful to National Textile University, Faisalabad, Pakistan for their support.

Conflicts of Interest

The authors declare no conflict of interest. All authors agreed to submit the article in this journal.

References

  1. Bugshan, A.; Kassolis, J.; Basile, J. Primary diffuse large B-cell lymphoma of the mandible: Case report and review of the literature. Case Rep. Oncol. 2015, 8, 451–455. [Google Scholar] [CrossRef]
  2. van der Zee, J. Heating the patient: A promising approach? Ann. Oncol. 2002, 13, 1173–1184. [Google Scholar] [CrossRef]
  3. Dagenais, G.R.; Leong, D.P.; Rangarajan, S.; Lanas, F.; Lopez-Jaramillo, P.; Gupta, R.; Diaz, R.; Avezum, A.; Oliveira, G.B.; Wielgosz, A.; et al. Variations in common diseases, hospital admissions, and deaths in middle-aged adults in 21 countries from five continents (PURE): A prospective cohort study. Lancet 2020, 395, 785–794. [Google Scholar] [CrossRef]
  4. Mehlen, P.; Puisieux, A. Metastasis: A question of life or death. Nat. Rev. Cancer 2006, 6, 449–458. [Google Scholar] [CrossRef]
  5. Glazer, E.S.; Curley, S.A. The Ongoing History of Thermal Therapy for Cancer. Surg. Oncol. Clin. NA 2011, 20, 229–235. [Google Scholar] [CrossRef]
  6. Chen, Q.; Zheng, J.; Yuan, X.; Wang, J.; Zhang, L. Folic acid grafted and tertiary amino based pH-responsive pentablock polymeric micelles for targeting anticancer drug delivery. Mater. Sci. Eng. C 2018, 82, 1–9. [Google Scholar] [CrossRef]
  7. Maeda, H. Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity. Adv. Drug Deliv. Rev. 2015, 91, 3–6. [Google Scholar] [CrossRef]
  8. Škubn, J.; Rimpelov, S. Vincristine in Combination Therapy of Cancer: Emerging Trends in Clinics. Biology 2021, 10, 849. [Google Scholar] [CrossRef]
  9. Kumar, A. Vincristine and Vinblastine: A Review. Int. J. Med. Pharm. Sci. 2016, 6, 23–30. [Google Scholar]
  10. Ameyaw, B.; Jolicoeur, M. Missing enzymes in the biosynthesis of the anticancer drug vinblastine in Madagascar periwinkle. Science 2018, 360, 1235–1239. [Google Scholar]
  11. Stankovic, M.Z.; Marinkovic, V.D. Paclitaxel as an anticancer agent: Isolation, activity, synthesis and stability. Open Med. 2011, 6, 527–536. [Google Scholar] [CrossRef]
  12. Barbuti, A.M.; Chen, Z. Paclitaxel Through the Ages of Anticancer Therapy: Exploring Its Role in Chemo-resistance and Radiation Therapy. Cancers 2015, 7, 2360–2371. [Google Scholar] [CrossRef]
  13. Zhao, P.; Astruc, D. Docetaxel Nanotechnology in Anticancer Therapy. ChemMedChem 2012, 7, 952–972. [Google Scholar] [CrossRef] [PubMed]
  14. Herbst, R.S.; Khuri, F.R. Mode of action of docetaxel—A basis for combination with novel anticancer agents. Cancer Treat. Rev. 2003, 29, 407–415. [Google Scholar] [CrossRef]
  15. Ciardiello, F.; Bianco, R.; Damiano, V.; De Lorenzo, S.; Pepe, S.; De Placido, S.; Fan, Z.; Mendelsohn, J.; Bianco, A.R.; Tortora, G. Antitumor activity of sequential treatment with topotecan and anti-epidermal growth factor receptor monoclonal antibody C225. Clin. Cancer Res. 1999, 5, 909–916. [Google Scholar]
  16. O’Brien, M.; Eckardt, J.; Ramlau, R. Recent Advances with Topotecan in the Treatment of Lung Cancer. Oncologist 2007, 12, 1194–1204. [Google Scholar] [CrossRef] [PubMed]
  17. Darji, P.; Patel, J.; Patel, B.; Chudasama, A.; Fnu, P.I.J.; Nalla, S. A comprehensive review on anticancer natural drugs. World J. Pharm. Pharm. Sci. 2024, 13, 717–734. [Google Scholar]
  18. Picher, E.A.; Muhammad, W.; Barth, S.; Chisholm, J.; Shipley, J.; Pors, K. The Capacity of Drug-Metabolising Enzymes in Modulating the Therapeutic Efficacy of Drugs to Treat Rhabdomyosarcoma. Cancers 2024, 16, 1012. [Google Scholar] [CrossRef]
  19. Luapattarakasem, P. Role of Natural Products on Cancer Prevention and Treatment. Srinagarind Med. J. 2005, 20, 180–189. [Google Scholar]
  20. Cragg, G.M.; Newman, D.J. Plants as a source of anti-cancer agents. J. Ethnopharmacol. 2005, 100, 72–79. [Google Scholar] [CrossRef]
  21. Bello, O.A.; Ayanda, O.I.; Aworunse, O.S.; Olukanmi, B.I. Solanecio biafrae: An underutilized nutraceutically-important African indigenous vegetable. Pharmacogn. Rev. 2018, 1, 94–102. [Google Scholar]
  22. Patel, V.; Senderowicz, A.M.; Pinto, D., Jr.; Igishi, T.; Raffeld, M.; Quintanilla-Martinez, L.; Ensley, J.F.; Sausviille, E.A.; Gutkind, J.S. Flavopiridol, a novel cyclin-dependent kinase inhibitor, suppresses the growth of head and neck squamous cell carcinomas by inducing apoptosis. J. Clin. Investig. 1998, 102, 1674–1681. [Google Scholar] [CrossRef] [PubMed]
  23. King, M.R.; Mohamed, Z.J. Dual nanoparticle drug delivery: The future of anticancer therapies? Nanomedicine 2017, 12, 95–98. [Google Scholar] [CrossRef] [PubMed]
  24. Banihashem, S.; Nezhati, M.N.; Panahi, H.A.; Shakeri-Zadeh, A. Synthesis of novel chitosan-g-PNVCL nanofibers coated with gold-gold sulfide nanoparticles for controlled release of cisplatin and treatment of MCF-7 breast cancer. Int. J. Polym. Mater. Polym. Biomater. 2020, 69, 1197–1208. [Google Scholar] [CrossRef]
  25. Zamani, M.; Prabhakaran, M.P.; Ramakrishna, S. Advances in drug delivery via electrospun and electrosprayed nanomaterials. Int. J. Nanomed. 2013, 8, 2997–3017. [Google Scholar] [CrossRef]
  26. Okuda, T.; Tominaga, K.; Kidoaki, S. Time-programmed dual release formulation by multilayered drug-loaded nanofiber meshes. J. Control. Release 2010, 143, 258–264. [Google Scholar] [CrossRef] [PubMed]
  27. Pant, B.; Park, M.; Park, S.J. Drug delivery applications of core-sheath nanofibers prepared by coaxial electrospinning: A review. Pharmaceutics 2019, 11, 305. [Google Scholar] [CrossRef]
  28. Mishra, V.; Bansal, K.K.; Verma, A.; Yadav, N.; Thakur, S.; Sudhakar, K.; Rosenholm, J.M. Solid lipid nanoparticles: Emerging colloidal nano drug delivery systems. Pharmaceutics 2018, 10, 191. [Google Scholar] [CrossRef]
  29. Eskandar, N.G.; Simovic, S.; Prestidge, C.A. Chemical stability and phase distribution of all-trans-retinol in nanoparticle-coated emulsions. Int. J. Pharm. 2009, 376, 186–194. [Google Scholar] [CrossRef]
  30. Mu, H.; Holm, R. Solid lipid nanocarriers in drug delivery: Characterization and design. Expert Opin. Drug Deliv. 2018, 15, 771–785. [Google Scholar] [CrossRef]
  31. Zhao, X.; Zhoo, J.; Lin, Z.Y.; Pan, G.; Zhu, Y.; Cheng, Y.; Cui, W. Self-coated interfacial layer at organic/inorganic phase for temporally controlling dual-drug delivery from electrospun fibers. Colloids Surf. B Biointerfaces 2015, 130, 1–9. [Google Scholar] [CrossRef] [PubMed]
  32. Lin, T.C.; Lin, F.H.; Lin, J.C. In vitro feasibility study of the use of a magnetic electrospun chitosan nanofiber composite for hyperthermia treatment of tumor cells. Acta Biomater. 2012, 8, 2704–2711. [Google Scholar] [CrossRef] [PubMed]
  33. Sun, L.; Wu, Y.; Fan, Z.; Ding, X.; Huang, Y.; Paisley, J. A deep error correction network for compressed sensing MRI. BMC Biomed. Eng. 2020, 2, 4. [Google Scholar] [CrossRef] [PubMed]
  34. Schmaljohann, D. Thermo- and pH-responsive polymers in drug delivery. Adv. Drug Deliv. Rev. 2006, 58, 1655–1670. [Google Scholar] [CrossRef] [PubMed]
  35. Dey, N.; Rao, M. Quantum Dot: Novel Carrier for Drug Delivery. Int. J. Res. Pharma-Ceutical Biomed. Sci. 2011, 2, 448–458. [Google Scholar]
  36. Xu, Q.; Yang, Y.; Hou, J.; Chen, T.; Fei, Y.; Wang, Q.; Zhou, Q.; Li, W.; Ren, J.; Li, Y.-G. A carbon nanotubes based in situ multifunctional power assist system for restoring failed heart function. BMC Biomed. Eng. 2021, 3, 5. [Google Scholar] [CrossRef] [PubMed]
  37. Kaur, J.; Gill, G.S.; Jeet, K. Applications of Carbon Nanotubes in Drug Delivery: A Comprehensive Review. In Characterization and Biology of Nanomaterials for Drug Delivery; Elsevier Inc.: Amsterdam, The Netherlands, 2018. [Google Scholar] [CrossRef]
  38. Vazquez-Munoz, R.; Arellano-Jimenez, M.J.; Lopez-Ribot, J.L. Bismuth nanoparticles obtained by a facile synthesis method exhibit antimicrobial activity against Staphylococcus aureus and Candida albicans. BMC Biomed. Eng. 2020, 2, 1–12. [Google Scholar] [CrossRef]
  39. Zhang, L.; Rean, S.R.; Li, X.; Sack, T.; Wang, Z.; Ahmed, S.; Chan, G.; Das, H.; Zaragoza, A.; Sargent, E.H.; et al. Programmable Metal/Semiconductor Nanostructures for mRNA-Modulated Molecular Delivery. Nano Lett. 2018, 18, 6222–6228. [Google Scholar] [CrossRef] [PubMed]
  40. Woolfson, A.D.; McCafferty, D.F.; McCarron, P.A.; Price, J.H. A bioadhesive patch cervical drug delivery system for the administration of 5-fluorouracil to cervical tissue. J. Control. Release 1995, 35, 49–58. [Google Scholar] [CrossRef]
  41. Ouattara, L.A.; Barnable, P.; Mawson, P.; Seidor, S.; Zydowsky, T.; Kizima, K.; Rodriguez, A. Miv-150-containing intravaginal rings protect macaque vaginal explants against shiv-rt infection. Antimicrob. Agents Chemother. 2014, 58, 2841–2848. [Google Scholar] [CrossRef]
  42. Xi, H.; Zhao, H. Silk fibroin coaxial bead-on-string fiber materials and their drug release behaviors in different pH. J. Mater. Sci. 2019, 54, 4246–4258. [Google Scholar] [CrossRef]
  43. Fan, M.; Ferguson, L.; Rohan, L.; Meyn, L.; Hillier, S. P2-S9.06 Vaginal film microbicides for HIV prevention: A mixed methods study of women’s preferences. Sex. Transm. Infect. 2011, 87, A263. [Google Scholar] [CrossRef]
  44. Cevher, E.; Açma, A.; Sinani, G.; Aksu, B.; Zloh, M.; Mülazimoǧlu, L. Bioadhesive tablets containing cyclodextrin complex of itraconazole for the treatment of vaginal candidiasis. Int. J. Biol. Macromol. 2014, 69, 124–136. [Google Scholar] [CrossRef] [PubMed]
  45. Şenyiǧit, Z.A.; Karavana, S.Y.; Eraç, B.; Gürsel, Ö.; Limoncu, M.H.; Baloǧlu, E. Evaluation of chitosan based vaginal bioadhesive gel formulations for antifungal drugs. Acta Pharm. 2014, 64, 139–156. [Google Scholar] [CrossRef] [PubMed]
  46. Graham, V.; Surwit, E.S.; Weiner, S.; Meyskens, F.L. Phase II trial of β-all-trans-retinoic acid for cervical intraepithelial neoplasia delivered via a collagen sponge and cervical cap. West. J. Med. 1986, 145, 192–195. [Google Scholar] [PubMed]
  47. Kirwan, P.; Naftalin, N.J. Topical 5-fluorouracil in the treatment of vaginal intraepithelial neoplasia. BJOG Int. J. Obstet. Gynaecol. 1985, 92, 287–291. [Google Scholar] [CrossRef] [PubMed]
  48. Zhang, H.; Ba, S.; Yang, Z.; Wang, T.; Lee, J.Y.; Li, T.; Shao, F. Graphene Quantum Dot-Based Nanocomposites for Diagnosing Cancer Biomarker APE1 in Living Cells. ACS Appl. Mater. Interfaces 2020, 12, 13634–13643. [Google Scholar] [CrossRef] [PubMed]
  49. Mora-Huertas, C.E.; Fessi, H.; Elaissari, A. Polymer-based nanocapsules for drug delivery. Int. J. Pharm. 2010, 385, 113–142. [Google Scholar] [CrossRef] [PubMed]
  50. Qadir, M.B.; Jalalah, M.; Shoukat, M.U.; Ahmad, A.; Khaliq, Z.; Nazir, A.; Anjum, M.N.; Rahman, A.; Khan, M.Q.; Tahir, R.; et al. Nonwoven/Nanomembrane Composite Functional Sweat Pads. Membranes 2022, 12, 1230. [Google Scholar] [CrossRef]
  51. Sarwar, M.N.; Ullah, A.; Haider, M.K.; Hussain, N.; Ullah, S.; Hashmi, M.; Khan, M.Q.; Kim, I.S. Evaluating antibacterial efficacy and biocompatibility of pan nanofibers loaded with diclofenac sodium salt. Polymers 2021, 13, 510. [Google Scholar] [CrossRef]
  52. Phan, D.-N.; Khan, M.Q.; Van, C.N.; Vu, M.-H.; Ullah, A.; Khatri, M.; Kim, I.-S. The Inuence of Orange Essential Oil on Mechanical, Chemical, and Antibacterial Properties of Cellulose Nanobers Containing Silver Nanoparticles. Res. Sq. 2021. preprint. [Google Scholar] [CrossRef]
  53. Khan, M.A.; Ahmad, A.; Arshad, S.N.; Nasir, A.; Ahmad, S.; Khan, M.Q.; Shahzad, A.; Satti, A.N.; Qadir, M.; Khaliq, Z. Development of optimized triaxially electrospun titania nanofiber-in-nanotube core-shell structure. J. Appl. Polym. Sci. 2021, 138, 50562. [Google Scholar] [CrossRef]
  54. Munir, M.U.; Mikucioniene, D.; Khanzada, H.; Khan, M.Q. Development of Eco-Friendly Nanomembranes of Aloe vera/PVA/ZnO for Potential Applications in Medical Devices. Polymers 2022, 14, 1029. [Google Scholar] [CrossRef] [PubMed]
  55. Ullah, A.; Sun, L.; Wang, F.-F.; Nawaz, H.; Yamashita, K.; Cai, Y.; Anwar, F.; Khan, M.Q.; Mayakrishnan, G.; Kim, I.-S. Eco-friendly bioactive β-caryophyllene/halloysite nanotubes loaded nanofibrous sheets for active food packaging. Food Packag. Shelf Life 2023, 35, 101028. [Google Scholar] [CrossRef]
  56. Hassan, T.; Jamshaid, H.; Mishra, R.; Khan, M.Q.; Petru, M.; Novak, J.; Choteborsky, R.; Hromasova, M. Acoustic, mechanical and thermal properties of green composites reinforced with natural fiberswaste. Polymers 2020, 12, 654. [Google Scholar] [CrossRef] [PubMed]
  57. Shafiq, F.; Siddique, A.; Pervez, M.N.; Hassan, M.M.; Naddeo, V.; Cai, Y.; Hou, A.; Xie, K.; Khan, M.Q.; Kim, I.-S. Extraction of natural dye from aerial parts of argy wormwood based on optimized taguchi approach and functional finishing of cotton fabric. Materials 2021, 14, 5850. [Google Scholar] [CrossRef] [PubMed]
  58. Imran, M.A.; Khan, M.Q.; Salam, A.; Ahmad, A. Cotton in Nonwoven Products. In Cotton Science and Processing Technology: Gene, Ginning, Garment and Green Recycling; Springer: Singapore, 2020; pp. 305–332. [Google Scholar] [CrossRef]
  59. Khanzada, H.; Salam, A.; Qadir, M.B.; Phan, D.-N.; Hassan, T.; Munir, M.U.; Pasha, K.; Hassan, N.; Khan, M.Q.; Kim, I.S. Fabrication of promising antimicrobial aloe vera/PVA electrospun nanofibers for protective clothing. Materials 2020, 13, 3884. [Google Scholar] [CrossRef] [PubMed]
  60. Semnani, D.; Naghashzargar, E.; Hadjianfar, M.; Manshadi, F.D.; Mohammadi, S.; Karbasi, S.; Efafty, F. Evaluation of PCL/chitosan electrospun nanofibers for liver tissue engineering. Int. J. Polym. Mater. Polym. Biomater. 2017, 66, 149–157. [Google Scholar] [CrossRef]
  61. He, D.; Hu, B.; Yao, Q.; Wang, K.; Yu, S. Large-Scale Synthesis of Flexible Free- Sensitivity: Electrospun PVA Nanofibers of Silver Nanoparticles. ACS Nano 2009, 3, 3993–4002. [Google Scholar] [CrossRef]
  62. Zhang, Y.Z.; Su, B.; Ramakrishna, S.; Lim, C.T. Chitosan nanofibers from an easily electrospinnable UHMWPEO-doped chitosan solution system. Biomacromolecules 2008, 9, 136–141. [Google Scholar] [CrossRef]
  63. Niu, X.; Zhao, L.; Yin, M.; Huang, D.; Wang, N.; Wei, Y.; Hu, Y.; Lian, X.; Chen, W. Mineralized Polyamide66/Calcium Chloride Nanofibers for Bone Tissue Engineering. Tissue Eng. Part C Methods 2020, 26, 352–363. [Google Scholar] [CrossRef] [PubMed]
  64. SantosMiranda, M.E.I. The role of N-carboxymethylation of chitosan in the thermal stability and dynamic. Polym. Int. 2006, 55, 961–969. [Google Scholar]
  65. Ramezani, M.R.; Ansari-Asl, Z.; Hoveizi, E.; Kiasat, A.R. Fabrication and Characterization of Fe(III) Met-al-organic Frameworks Incorporating Polycaprolactone Nanofibers: Potential Scaffolds for Tissue Engineering. Fibers Polym. 2020, 21, 1013–1022. [Google Scholar] [CrossRef]
  66. Zarghami, A.; Irani, M.; Mostafazadeh, A.; Golpour, M.; Heidarinasab, A.; Haririan, I. Fabrication of PEO/chitosan/PCL/olive oil nanofibrous scaffolds for wound dressing applications. Fibers Polym. 2015, 16, 1201–1212. [Google Scholar] [CrossRef]
  67. Kharaghani, D.; Kurniwan, E.B.; Khan, M.Q.; Yoshiko, Y. MiRNA-nanofiber, the next generation of bioactive scaffolds for bone regeneration: A review. Micromachines 2021, 12, 1472. [Google Scholar] [CrossRef]
  68. Waqas, M. Silver sulfadiazine loaded nanofibers for burn infections. Int. J. Polym. Mater. Polym. Biomater. 2023, 72, 517–523. [Google Scholar] [CrossRef]
  69. Ghorbani, M.; Nezhad-Mokhtari, P.; Sohrabi, H.; Roshangar, L. Electrospun chitosan/nanocrystalline cellu-lose-graft-poly (N-vinylcaprolactam) nanofibers as the reinforced scaffold for tissue engineering. J. Mater. Sci. 2020, 55, 2176–2185. [Google Scholar] [CrossRef]
  70. Moeini, A.; Cimmino, A.; Masi, M.; Evidente, A.; Van Reenen, A. The incorporation and release of ungeremine, an antifungal Amaryllidaceae alkaloid, in poly.(lactic acid)/poly.(ethylene glycol) nanofibers. J. Appl. Polym. Sci. 2020, 137, 49098. [Google Scholar] [CrossRef]
  71. Nouri, A.; Dizaji, B.F.; Kianinejad, N.; Rad, A.J.; Rahimi, S.; Irani, M.; Jazi, F.S. Simultaneous linear release of folic acid and doxorubicin from ethyl cellu-lose/chitosan/g-C3N4/MoS2 core-shell nanofibers and its anticancer properties. J. Biomed. Mater. Res. A 2021, 109, 903–914. [Google Scholar] [CrossRef]
  72. Homaeigohar, S.; Boccaccini, A.R. Antibacterial biohybrid nanofibers for wound dressings. Acta Biomater. 2020, 107, 25–49. [Google Scholar] [CrossRef]
  73. Boda, S.K.; Fischer, N.G.; Ye, Z.; Aparicio, C. Dual Oral Tissue Adhesive Nanofiber Membranes for pH-Responsive Delivery of Antimicrobial Peptides. Biomacromolecules 2020, 21, 4945–4961. [Google Scholar] [CrossRef] [PubMed]
  74. Agrawal, S.; Srivastava, R. Osteoinductive and osteoconductive biomaterials. In Racing for the Surface: Antimicrobial and Interface Tissue Engineering; Springer: Cham, Switzerland, 2020. [Google Scholar] [CrossRef]
  75. Grierson, I.; Minckler, D.; Rippy, M.K.; Marshall, A.J.; Collignon, N.; Bianco, J.; Detry, B.; Jonstone, M. A novel suprachoroidal microinvasive glaucoma implant: In vivo biocompatibility and biointegration. BMC Biomed. Eng. 2020, 2, 10. [Google Scholar] [CrossRef] [PubMed]
  76. Shokrollahi, M.; Bahrami, S.H.; Nazarpak, M.H.; Solouk, A. Biomimetic double-sided polypropylene mesh modified by DOPA and ofloxacin loaded carboxyethyl chitosan/polyvinyl alcohol-polycaprolactone nanofibers for potential hernia repair applications. Int J. Biol. Macromol. 2020, 165, 902–917. [Google Scholar] [CrossRef] [PubMed]
  77. Jalalah, M.; Ahmad, A.; Saleem, A.; Qadir, M.B.; Khaliq, Z.; Khan, M.Q.; Nazir, A.; Faisal, M.; Alsaiari, M.; Irfan, M.; et al. Electrospun Nanofiber/Textile Supported Composite Membranes with Improved Mechanical Performance for Biomedical Applications. Membranes 2022, 12, 1158. [Google Scholar] [CrossRef] [PubMed]
  78. Phan, D.-N.; Khan, M.Q.; Nguyen, V.-C.; Vu-Manh, H.; Dao, A.-T.; Thanh Thao, P.; Nguyen, N.-M.; Le, V.-T.; Ullah, A.; Khatri, M.; et al. Investigation of Mechanical, Chemical, and Antibacterial Properties of Electrospun Cellu-lose-Based Scaffolds Containing Orange Essential Oil and Silver Nanoparticles. Polymers 2022, 14, 85. [Google Scholar] [CrossRef]
  79. Salam, A.; Hassan, T.; Jabri, T.; Riaz, S.; Khan, A.; Iqbal, K.M.; Khan, S.u.; Wasim, M.; Shah, M.R.; Khan, M.Q.; et al. Electrospun nanofiber-based viroblock/ZnO/PAN hybrid antiviral nanocomposite for personal protective applications. Nanomaterials 2021, 11, 2208. [Google Scholar] [CrossRef] [PubMed]
  80. Hussain, M.; Salam, A.; Arain, M.F.; Ullah, A.; Dao, A.-T.; Vu-Manh, H.; Phan, D.-N.; Ansari, A.S.; Khan, M.Q.; Javed, Z.; et al. Polyacrylonitrile nanofibers containing viroblock as promising material for protective clothing. Appl. Sci. 2021, 11, 11469. [Google Scholar] [CrossRef]
  81. Kim, M.O.; Khan, M.Q.; Ullah, A.; Phan, D.; Zhu, C.; Lee, J.-S.; Kim, I.S. Fabrication and characterization of colorimetric polymer based novel nanofibers for sensing and blocking of bacterial. Mater. Res. Express 2020, 7, 085405. [Google Scholar] [CrossRef]
  82. Salam, A.; Khan, M.Q.; Hassan, T.; Hassan, N.; Nazis, A.; Hussain, T.; Azeem, M.; Kim, I.S. In-vitro assessment of appropriate hydrophilic scaffolds by co-electrospinning of poly (1,4 cyclohexane isosorbide terephthalate)/polyvinyl alcohol. Sci. Rep. 2020, 10, 19751. [Google Scholar] [CrossRef]
  83. Bie, X.; Khan, M.Q.; Ullah, A.; Ullah, S.; Karaghani, D.; Phan, D.; Tamada, Y.; Kim, I.S. Fabrication and characterization of wound dressings containing gentamicin/silver for wounds in diabetes mellitus patients. Mater. Res. Express 2020, 7, 045004. [Google Scholar] [CrossRef]
  84. Hassan, T.; Khan, M.Q.; Salam, A.; Hassan, N.; Raza, A.; Bukhsh, N.; Javed, Z.; Kim, I.S. The assessment of finishing properties on the mass per unit area, pilling, bursting strength, and wicking behavior of polyester weft-knitted jersey fabric. Coatings 2020, 10, 723. [Google Scholar] [CrossRef]
  85. Ayaz, M.; Salam, A.; Khan, S.U.; Khan, M.Q.; Hussain, T.; Kim, I.S. Bioactive Silvadur loaded polyacrylonitrile nanofibrous membranes for wound dressing applications. Mater. Res. Express 2022, 9, 115305. [Google Scholar] [CrossRef]
  86. Khan, M.Q.; Kharagani, D.; Sanaullah, D.; Shahzad, A.; Duy, N.P.; Hasegawa, Y.; Azeemullah, D. Fabrication of Antibacterial Nanofibers Composites by Functionalizing the Surface of Cellulose Acetate Nanofibers. ChemistrySelect 2020, 5, 1315–1321. [Google Scholar] [CrossRef]
  87. Su, Z.; Ding, J.; Wei, G. Electrospinning: A facile technique for fabricating polymeric nanofibers doped with carbon nanotubes and metallic nanoparticles for sensor applications. RSC Adv. 2014, 4, 52598–52610. [Google Scholar] [CrossRef]
  88. Jin, Y.; Yang, D.; Kang, D.; Xiang, X. Fabrication of necklace-like structures via electrospinning. Langmuir 2010, 26, 1186–1190. [Google Scholar] [CrossRef] [PubMed]
  89. Nirmala, R.; Navamathavan, R.; Park, S.J.; Kim, H.Y. Recent Progress on the Fabrication of Ultrafine Polyamide-6 Based Nanofibers Via Electrospinning: A Topical Review. Nano-Micro Lett. 2014, 6, 89–107. [Google Scholar] [CrossRef]
  90. Fathi-Azarbayjani, A.; Chan, S.Y. Single and multi-layered nanofibers for rapid and controlled drug delivery. Chem. Pharm. Bull. 2010, 58, 143–146. [Google Scholar] [CrossRef]
  91. Zhu, L.F.; Zheng, Y.; Fan, J.; Yao, Y.; Ahmad, Z.; Chang, M.W. A novel core-shell nanofiber drug delivery system intended for the synergistic treatment of melanoma. Eur. J. Pharm. Sci. 2019, 137, 105002. [Google Scholar] [CrossRef] [PubMed]
  92. Lu, L.; Cao, X.; Shen, Z.; Li, L.; Huo, J.; Chen, W.; Liu, C.; Liu, H. Electrospun nitrogen-doped carbon nanofibers for electrocatalysis. Sustain. Mater. Technol. 2020, 26, e00221. [Google Scholar] [CrossRef]
  93. Akhgari, A.; Shakib, Z.; Sanati, S. A review on electrospun nanofibers for oral drug delivery. Nanomed. J. 2017, 4, 197–207. [Google Scholar] [CrossRef]
  94. Anaraki, N.A.; Rad, L.R.; Irani, M.; Haririan, I. Fabrication of PLA/PEG/MWCNT electrospun nanofibrous scaffolds for anticancer drug delivery. J. Appl. Polym. Sci. 2015, 132, 1–9. [Google Scholar] [CrossRef]
  95. Vernosfaderani, F.R.; Semnani, D. Manufacturing and Optimization the Nanofibres Tissue of Poly (N-vinyl-2-pyrrolidone)-Poly (e-caprolactone) Shell/Poly (N-vinyl-2-pyrrolidone)-Amphotericin B Core for Controlled Drug Release System. Fibers Polym. 2018, 19, 620–626. [Google Scholar] [CrossRef]
  96. Acevedo, F.; Hermosilla, J.; Sanhueza, C.; Mora-Lagos, B.; Fuentes, I.; Rubilar, M.; Concheiro, A.; Lorenzo, C. Gallic acid loaded PEO-core/zein-shell nanofibers for chemopreventive action on gallbladder cancer cells. Eur. J. Pharm. Sci. 2018, 119, 49–61. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, J.; Li, S.; Ju, D.-D.; Li, X.; Zhang, J.-C.; Yan, X.; Long, Y.-Z.; Song, F. Flexible inorganic core-shell nanofibers endowed with tunable multicolor upconversion fluores-cence for simultaneous monitoring dual drug delivery. Chem. Eng. J. 2018, 349, 554–561. [Google Scholar] [CrossRef]
  98. Irani, M.; Sadeghi, G.M.M.; Haririan, I. Electrospun biocompatible poly (ε-caprolactonediol)-based polyurethane core/shell nanofibrous scaffold for controlled release of temozolomide. Int. J. Polym. Mater. Polym. Biomater. 2018, 67, 361–366. [Google Scholar] [CrossRef]
  99. Han, D.; Serar, R.; Gorelick, N.; Fatima, Y.; Eberhart, C.G.; Brem, H.; Tyler, B.; Steckl, A. Multi-layered core-sheath fiber membranes for controlled drug release in the local treatment of brain tumor. Sci. Rep. 2019, 9, 17936. [Google Scholar] [CrossRef] [PubMed]
  100. Alghoraibi, I.; Alomari, S.; Nanofibers, H.O. Handbook of Nanofibers; Springer Nature: Dordrecht, The Netherlands, 2020. [Google Scholar] [CrossRef]
  101. Aytac, Z.; Uyar, T. Applications of core-shell nanofibers: Drug and biomolecules release and gene therapy. In Core-Shell Nanostructures for Drug Delivery and Theranostics; Woodhead Publishing: Sawston, UK, 2018; pp. 375–404. [Google Scholar]
  102. Jouybari, M.H.; Hosseini, S.; Mahboobnia, K.; Boloursaz, L.A.; Moradi, M.; Irani, M. Simultaneous controlled release of 5-FU, DOX and PTX from chitosan/PLA/5-FU/g-C3N4-DOX/g-C3N4-PTX triaxial nanofibers for breast cancer treatment in vitro. Colloids Surf. B Biointerfaces 2019, 179, 495–504. [Google Scholar] [CrossRef] [PubMed]
  103. Bazzazzadeh, A.; Dizaji, B.F.; Kianinejad, N.; Nouri, A.; Irani, M. Fabrication of poly (acrylic acid) graft-ed-chitosan/polyurethane/magnetic MIL-53 metal organic framework composite core-shell nanofibers for co-delivery of temozolomide and paclitaxel against glioblastoma cancer cells. Int. J. Pharm. 2020, 587, 119674. [Google Scholar] [CrossRef]
  104. Salehi, R.; Irani, M.; Eskandani, M.; Nowruzi, K.; Davaran, S.; Haririan, I. Interaction, controlled release, and antitumor activity of doxorubicin hydrochloride from pH-sensitive P (NIPAAm-MAA-VP) nanofibrous scaffolds prepared by green electrospinning. Int. J. Polym. Mater. Polym. Biomater. 2014, 63, 609–619. [Google Scholar] [CrossRef]
  105. Yuan, H.; Li, B.; Liang, K.; Lou, X.; Zhang, Y. Regulating drug release from pH- and temperature-responsive electrospun CTS-g-PNIPAAm/poly (ethylene oxide) hydrogel nanofibers. Biomed. Mater. 2014, 9, 055001. [Google Scholar] [CrossRef]
  106. Liu, L.; Bai, S.; Yang, H.; Li, S.; Quan, J.; Zhu, L.; Nie, H. Controlled release from thermo-sensitive PNVCL-co-MAA electrospun nanofibers: The effects of hydrophilicity/hydrophobicity of a drug. Mater. Sci. Eng. C 2016, 67, 581–589. [Google Scholar] [CrossRef] [PubMed]
  107. Ullah, S.; Hashimi, M.; Kharagani, D.; Khan, M.Q.; Saito, Y.; Yamamoto, T.; Lee, J.; Kim, S.I. Antibacterial properties of in situ and surface functionalized impregnation of silver sulfadiazine in polyacrylonitrile nanofiber mats. Int. J. Nanomed. 2019, 14, 2693–2703. [Google Scholar] [CrossRef] [PubMed]
  108. Hassan, T.; Salam, A.; Khan, A.; Khan, S.Y.; Khanzada, H.; Wasim, M.; Khan, M.Q.; Kim, I.S. Functional nanocomposites and their potential applications: A review. J. Polym. Res. 2021, 28, 36. [Google Scholar] [CrossRef]
  109. Ullah, S.; Hashimi, M.; Khan, M.Q.; Kharagani, D.; Saito, Y.; Yamamoto, T.; Kim, I.S. Silver sulfadiazine loaded zein nanofiber mats as a novel wound dressing. RSC Adv. 2019, 9, 268–277. [Google Scholar] [CrossRef] [PubMed]
  110. Hadjianfar, M.; Semnani, D.; Varshosaz, J. An investigation on polycaprolactone/chitosan/Fe3O4 nanofibrous composite used for hyperthermia. Polym. Adv. Technol. 2019, 30, 2729–2741. [Google Scholar] [CrossRef]
  111. Makadia, H.K.; Siegel, S.J. Poly Lactic-co-Glycolic Acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers 2011, 3, 1377–1397. [Google Scholar] [CrossRef] [PubMed]
  112. DeMerlis, C.C. Review of the oral toxicity of polyvinyl alcohol. Food Chem. Toxicol. 2003, 41, 319–326. [Google Scholar] [CrossRef] [PubMed]
  113. Wolosewick, J.J. The application of polyethylene glycol (PEG) to electron microscopy. J. Cell Biol. 1980, 86, 675. [Google Scholar] [CrossRef]
  114. Zafar, F. Polyurethane; InTechOpen: London, UK, 2012. [Google Scholar] [CrossRef]
  115. Alipal, J.; Pu’ad, N.A.S.M.; Lee, T.C.; Nayan, N.H.M.; Sahari, N.; Basri, H.; Idris, M.I.; Abdullah, H.Z. A review of gelatin: Properties, sources, process, applications, and commercialization. Mater. Today Proc. 2019, 42, 240–250. [Google Scholar] [CrossRef]
  116. Labet, M.; Thielemans, W. Synthesis of polycaprolactone: A review. Chem. Soc. Rev. 2009, 38, 3484–3504. [Google Scholar] [CrossRef]
  117. Fischer, S.; Thümmler, K.; Volkert, B.; Hettrich, K.; Schmidt, I.; Fischer, K. Properties and applications of cellulose acetate. Macromol. Symp. 2008, 262, 89–96. [Google Scholar] [CrossRef]
  118. Ohkawa, K. Nanofibers of cellulose and its derivatives fabricated using direct electrospinning. Molecules 2015, 20, 9139–9154. [Google Scholar] [CrossRef]
  119. Liu, X. Antimicrobial electrospun nanofibers of cellulose acetate and polyester urethane composite for wound dressing. J. Biomed. Mater. Res. B Appl. Biomater. 2012, 100, 1556–1565. [Google Scholar] [CrossRef] [PubMed]
  120. Marcuello, C.; Foulon, L.; Chabbert, B.; Aguié-Béghin, V.; Molinari, M. Atomic force microscopy reveals how relative humidity impacts the Young’s modulus of lignocellulosic polymers and their adhesion with cellulose nano-crystals at the nanoscale. Int. J. Biol. Macromol. 2020, 147, 1064–1075. [Google Scholar] [CrossRef]
  121. Suwantong, O.; Opanasopit, P.; Ruktanonchai, U.; Supaphol, P. Electrospun cellulose acetate fiber mats containing curcumin and release characteristic of the herbal substance. Polymer 2007, 48, 7546–7557. [Google Scholar] [CrossRef]
  122. Abid, S.; Raza, Z.A.; Rehman, A. Synthesis of poly (3-hydroxybutyrate) nanospheres and deposition thereof into porous thin film. Mater. Res. Express 2016, 3, 105042. [Google Scholar] [CrossRef]
  123. Zhao, Y.; Fan, Z.; Shen, M.; Shi, X. Hyaluronic Acid-Functionalized Electrospun Polyvinyl Alco-hol/Polyethyleneimine Nanofibers for Cancer Cell Capture Applications. Adv. Mater. Interfaces 2015, 2, 1500256. [Google Scholar] [CrossRef]
  124. Fan, Z.Y.; Zhao, Y.L.; Zhu, X.Y.; Luo, Y.; Shen, M.W.; Shi, X.Y. Folic acid modified electrospun poly (vinyl alcohol)/polyethyleneimine nanofibers for cancer cell capture applications. Chin. J. Polym. Sci. 2016, 34, 755–765. [Google Scholar] [CrossRef]
  125. Yan, E.; Fan, Y.; Sun, Z.; Gao, J.; Hao, X.; Pei, S.; Wang, C.; Sun, L.; Zhang, D. Biocompatible core-shell electrospun nanofibers as potential application for chemotherapy against ovary cancer. Mater. Sci. Eng. C 2014, 41, 217–223. [Google Scholar] [CrossRef]
  126. Huang, C.; Soenan, S.J.; Rejman, J.; Trekker, J.; Chengxun, L.; Lagae, K.; Ceelen, W.; Wilhelm, C.; Dermeester, J.; De Smedt, S.C. Magnetic electrospun fibers for cancer therapy. Adv. Funct. Mater. 2012, 22, 2479–2486. [Google Scholar] [CrossRef]
  127. Song, M.; Guo, D.; Pan, C.; Jiang, H.; Chen, C.; Zhang, R.; Gu, Z.; Wang, X. The application of poly (N-isopropylacrylamide)-co-polystyrene nanofibers as an additive agent to facilitate the cellular uptake of an anticancer drug. Nanotechnology 2008, 19, 165102. [Google Scholar] [CrossRef]
  128. Akaraonye, E.; Keshavarz, T.; Roy, I. Production of polyhydroxyalkanoates: The future green materials of choice. J. Chem. Technol. Biotechnol. 2010, 85, 732–743. [Google Scholar] [CrossRef]
  129. Loo, C.; Sudesh, K. Polyhydroxyalkanoates: Bio-based microbial plastics and their properties. Malays. Polym. J. 2007, 2, 31–57. [Google Scholar]
  130. Peng, S.W.; Guoo, X.-Y.; Shang, G.-G.; Li, J.; Xu, X.-Y.; You, M.-L.; Li, P.; Chen, G. An assessment of the risks of carcinogenicity associated with polyhydroxyalkanoates through an analysis of DNA aneuploid and telomerase activity. Biomaterials 2011, 32, 2546–2555. [Google Scholar] [CrossRef] [PubMed]
  131. Sudesh, K.; Lee, Y.-F.; Sridewi, N.; Ramanathan, S. The Influence of Electrospinning Parameters and Drug Loading on Polyhydroxyalkanoate (PHA) Nanofibers for Drug Delivery. Int. J. Biotechnol. Wellness Ind. 2016, 4, 103–113. [Google Scholar] [CrossRef]
  132. Mottin, A.C.; Ayres, E.; Oréfice, R.L.; Câmara, J.J.D. What Changes in Poly(3-Hydroxybutyrate) (PHB) When Processed as Electrospun Nanofibers or Thermo-Compression Molded Film? Mater. Res. 2016, 19, 57–66. [Google Scholar] [CrossRef]
  133. O’Connor, S.; Szwej, E.; Nikodinovic-Runic, J.; O’Connor, A.; Byrne, A.T.; Devocelle, M.; O’Donovan, N.; Gallagher, W.M. The anti-cancer activity of a cationic anti-microbial peptide derived from monomers of polyhydroxyalkanoate. Biomaterials 2013, 34, 2710–2718. [Google Scholar] [CrossRef]
  134. Lu, X.Y.; Li, M.C.; Zhu, X.L.; Fan, F.; Wang, L.L.; Ma, J.G. Microbial synthesized biodegradable PHBHHxPEG hybrid copolymer as an efficient intracellular delivery nanocarrier for kinase inhibitor. BMC Biotechnol. 2014, 14, 4. [Google Scholar] [CrossRef]
  135. Kim, Y.J.; Bae, H.I.; Kwon, O.K.; Choi, M.S. Three-dimensional gastric cancer cell culture using nanofiber scaffold for chemosensitivity test. Int. J. Biol. Macromol. 2009, 45, 65–71. [Google Scholar] [CrossRef]
  136. Abid, S.; Raza, Z.A.; Hussain, T. Production kinetics of polyhydroxyalkanoates by using Pseudomonas aeruginosa gamma ray mutant strain EBN-8 cultured on soybean oil. 3 Biotech 2016, 6, 142. [Google Scholar] [CrossRef]
  137. Raza, Z.A.; Abid, S.; Banat, I.M. Polyhydroxyalkanoates: Characteristics, production, recent developments and applications. Int. Biodeterior. Biodegradation 2018, 126, 45–56. [Google Scholar] [CrossRef]
  138. Raza, Z.A.; Abid, S.; Rehman, A.; Hussain, T. Synthesis kinetics of poly(3-hydroxybutyrate) by using a Pseudomonas aeruginosa mutant strain grown on hexadecane. Int. Biodeterior. Biodegradation 2016, 115, 171–178. [Google Scholar] [CrossRef]
  139. Yang, Z.; Zhao, X. A 3D model of ovarian cancer cell lines on peptide nanofiber scaffold to explore the cell-scaffold interaction and chemotherapeutic resistance of anticancer drugs. Int. J. Nanomed. 2011, 6, 303–310. [Google Scholar] [CrossRef] [PubMed]
  140. Liu, J.; Jinjian, L.; Hongyan, X.; Yumin, Z.; Liping, C.; Qingfen, L.; Naling, S.; Cuihong, H. Novel tumor-targeting, self-assembling peptide nanofiber as a carrier for effective curcumin delivery. Int. J. Nanomedicine 2014, 197–207. [Google Scholar] [CrossRef] [PubMed]
  141. Yang, C.; Chu, L.; Zhang, Y.; Shi, Y.; Liu, J.; Fan, S.; Yang, Z.; Ding, D.; Kong, D.; Liu, J. Dynamic biostability, biodistribution, and toxicity of l/d -peptide-based supramolecular nanofibers. ACS Appl. Mater. Interfaces 2015, 7, 2735–2744. [Google Scholar] [CrossRef] [PubMed]
  142. Mokhtar, A.; Abdelkrim, S.; Djelod, A.; Sardi, A.; Boukoussa, B.; Sassi, M.; Bengueddach, A. Adsorption behavior of cationic and anionic dyes on magadiite-chitosan composite beads. Carbohydr. Polym. 2020, 229, 115399. [Google Scholar] [CrossRef]
  143. Sukul, M.; Sahariah, P.; Lauzon, H.L.; Borges, J.; Masson, M.; Mano, J.F.; Haugen, H.J.; Reseland, J.E. In vitro biological response of human osteoblasts in 3D chitosan sponges with controlled degree of deacetylation and molecular weight. Carbohydr. Polym. 2021, 254, 117434. [Google Scholar] [CrossRef] [PubMed]
  144. Silvestre, W.P.; Baldasso, C.; Tessaro, I.C. Potential of chitosan-based membranes for the separation of essential oil components by target-organophilic pervaporation. Carbohydr. Polym. 2020, 247, 116676. [Google Scholar] [CrossRef] [PubMed]
  145. Dudek, G.; Turczyn, R.; Konieczny, K. Robust poly (vinyl alcohol) membranes containing chitosan/chitosan derivatives microparticles for pervaporative dehydration of ethanol. Sep. Purif. Technol. 2020, 234, 116094. [Google Scholar] [CrossRef]
  146. Barrera-Martínez, C.L.; Padilla-Vaca, F.; Liakos, I.; Meléndez-Ortiz, H.I.; Cortez-Mazatan, G.Y.; Peral-ta-Rodríguez, R.D. Chitosan microparticles as entrapment system for trans- cinnamaldehyde: Synthesis, drug loading, and in vitro cytotoxicity evaluation. Int. J. Biol. Macromol. 2020, 166, 322–332. [Google Scholar] [CrossRef]
  147. Michalik, R.; Wandzik, I. A mini-review on chitosan-based hydrogels with potential for sustainable agricultural applications. Polymers 2020, 12, 2425. [Google Scholar] [CrossRef] [PubMed]
  148. Garakani, S.S.; Khanmohammadi, M.; Atoufi, Z.; Kamrava, S.K.; Setayeshmehr, M.;; Alizadeh, R.; Faghihi, F.; Bagher, Z.; Davachi, S.M.; Abbaspourrad, A. Fabrication of chitosan/agarose scaffolds containing extracellular matrix for tissue engineering applications. Int. J. Biol. Macromol. 2020, 143, 533–545. [Google Scholar] [CrossRef]
  149. Garavand, F.; Caciotti, I.; Vahedikia, N.; Rehman, A.; Tarhan, O.; Akbari-Alavjeh, S. A comprehensive review on the nanocomposites loaded with chitosan nanoparticles for food packaging. Crit. Rev. Food Sci. Nutr. 2022, 1383–1416. [Google Scholar] [CrossRef] [PubMed]
  150. Saudi, S.; Bhattarai, S.R.; Adhikari, Y.; Khanal, S.; Sankar, J.; Aravamudhan, S.; Bhattarai, N. Nanonet-nano fiber electrospun mesh of PCL-chitosan for controlled and extended release of diclofenac sodium. Nanoscale 2020, 12, 23556–23569. [Google Scholar] [CrossRef]
  151. Khor, E.; Lim, L.Y. Implantable applications of chitin and chitosan. Biomaterials 2003, 24, 2339–2349. [Google Scholar] [CrossRef] [PubMed]
  152. Ignatova, M.; Manolova, N.; Rashkov, I. Novel antibacterial fibers of quaternized chitosan and poly (vinyl pyrrolidone) prepared by electrospinning. Eur. Polym. J. 2007, 43, 1112–1122. [Google Scholar] [CrossRef]
  153. Jayakumar, R.; Prabaharan, M.; Nair, S.V.; Tamura, H. Novel chitin and chitosan nanofibers in biomedical applications. Biotechnol. Adv. 2010, 28, 142–150. [Google Scholar] [CrossRef]
  154. Ardeshirzadeh, B.; Anaraki, N.A.; Irani, M.; Rad, L.R.; Shamshiri, S. Controlled release of doxorubicin from electrospun PEO/chitosan/graphene oxide nanocomposite nanofibrous scaffolds. Mater. Sci. Eng. C 2015, 48, 384–390. [Google Scholar] [CrossRef]
  155. Padron, S.; Fuentes, A.; Caruntu, D.; Lozano, K. Experimental study of nanofiber production through forcespinning. J. Appl. Phys. 2013, 113, 024318. [Google Scholar] [CrossRef]
  156. Zhang, X.; Chan-Yu-King, R.; Jose, A.; Manohar, S.K. Nanofibers of polyaniline synthesized by interfacial polymerization. Synth. Met. 2004, 145, 23–29. [Google Scholar] [CrossRef]
  157. Zhao, J.; Han, W.; Chen, H.; Tu, M.; Zeng, R.; Shi, Y.; Cha, Z.; Zhou, C. Preparation, structure and crystallinity of chitosan nano-fibers by a solid-liquid phase separation technique. Carbohydr. Polym. 2011, 83, 1541–1546. [Google Scholar] [CrossRef]
  158. Hartgerink, J.D.; Beniash, E.; Stupp, S.I. Self-assembly and mineralization of peptide-amphiphile nanofibers. Science 2001, 294, 1684–1688. [Google Scholar] [CrossRef] [PubMed]
  159. Meier, C.; Welland, M.E. Wet-spinning of amyloid protein nanofibers into multifunctional high-performance biofibers. Biomacromolecules 2011, 12, 3453–3459. [Google Scholar] [CrossRef] [PubMed]
  160. Bajakova, J.; Chaloupek, J.; Lukaš, D.; Lacarin, M. “Drawing”—The production of individual nanofibers by experimental method. In Proceedings of the NANOCON 2011—Conference Proceedings, 3rd International Conference, Brno, Czech Republic, 21–23 September 2011; pp. 322–326. [Google Scholar]
  161. Li, H.; Ke, Y.; Hu, Y. Polymer nanofibers prepared by template melt extrusion. J. Appl. Polym. Sci. 2006, 99, 1018–1023. [Google Scholar] [CrossRef]
  162. Shao, J.; Chen, C.; Wang, Y.; Chen, X.; Du, C. Reactive & Functional Polymers Early stage evolution of structure and nanoscale property of nanofibers in thermally induced phase separation process. React. Funct. Polym. 2012, 72, 765–772. [Google Scholar] [CrossRef]
  163. Zelenski, C.M.; Dorhout, P.K. Template synthesis of near-monodisperse microscale nanofibers and nano-tubules of MoS2. J. Am. Chem. Soc. 1998, 120, 734–742. [Google Scholar] [CrossRef]
  164. He, Z.; Liu, L.; Gao, C.; Zhou, Z.; Liang, X.; Lei, T.; He, Z.; Liu, S. Carbon nanofibers grown on the surface of graphite felt by chemical vapour deposition for vanadium redox flow batteries. RSC Adv. 2013, 3, 19774–19777. [Google Scholar] [CrossRef]
  165. Subbiah, T.; Bhat, G.S.; Tock, R.W.; Parameswaran, S.; Ramkumar, S.S. Electrospinning of nanofibers. J. Appl. Polym. Sci. 2005, 96, 557–569. [Google Scholar] [CrossRef]
  166. Gugulothu, D.; Barhoum, A.; Nerella, R.; Ajmer, R.; Bechelany, M.; Nanofibers, F.O.; Techniques, N.-E. Fabrication of nanofibers: Electrospinning and non-electrospinning techniques. In Handbook of Nanofibers; Springer: Cham, Switzerland, 2019. [Google Scholar] [CrossRef]
  167. Wang, X.; Lin, T.; Wang, X. Scaling up the production rate of nanofibers by needleless electrospinning from multiple ring. Fibers Polym. 2014, 15, 961–965. [Google Scholar] [CrossRef]
  168. Wei, L.; Liu, C.; Mao, X.; Dong, J.; Fan, W.; Zhi, C.; Qin, X.; Sun, R. Multiple-jet needleless electrospinning approach via a linear flume spinneret. Polymers 2019, 11, 2052. [Google Scholar] [CrossRef]
  169. Molnár, K.; Vas, L.M. Electrospun Composite Nanofibers and Polymer Composites. In Synthetic Polymer-Polymer Composites, 1st ed.; Carl Hanser Verlag: Munich, Germany, 2012. [Google Scholar] [CrossRef]
  170. Sydenstricker, V.P.; Cleckley, H.M. The effect of nicotinic acid in stupor, lethargy and various other psychiatric disorders. Am. J. Psychiatry 1941, 98, 83–92. [Google Scholar] [CrossRef]
  171. Yong, N.; Mullins; Bhattacharyya. United States: United States. Eur. J. Political Res. Political Data Yearb. 2021, 2–4. [Google Scholar]
  172. United States. Index of Patents Issued from the United States Patent Office; Government Printing Office: Washington, DC, USA, 1943.
  173. Formhals, A. Artificial Thread and Method of Producing Same. U.S. Patent No. 2,187,306, 16 January 1940. [Google Scholar]
  174. Akhgari, A.; Dezfuli, A.G.; Rezaei, M.; Kiarsi, M.; Abbaspour, M. The design and evaluation of a fast-dissolving drug delivery system for loratadine using the electrospinning method. Jundishapur J. Nat. Pharm. Prod. 2016, 11. [Google Scholar] [CrossRef]
  175. Li, X.; Kanjwal, M.A.; Lin, L.; Chronakis, I.S. Electrospun polyvinyl-alcohol nanofibers as oral fast-dissolving delivery system of caffeine and riboflavin. Colloids Surfaces B Biointerfaces 2013, 103, 182–188. [Google Scholar] [CrossRef] [PubMed]
  176. Yu, D.-G.; Shen, X.-X.; Branford-White, C.; White, K.; Zhu, L.-M.; Bligh, S.W.A. Oral fast-dissolving drug delivery membranes prepared from electrospun polyvinylpyrrolidone ultrafine fibers. Nanotechnology 2009, 20, 055104. [Google Scholar] [CrossRef] [PubMed]
  177. Zupančič, Š.; Sinha-Ray, S.; Sinha-Ray, S.; Kristl, J.; Yarin, A.L. Long-Term Sustained Ciprofloxacin Release from PMMA and Hydrophilic Polymer Blended Nanofibers. Mol. Pharm. 2016, 13, 295–305. [Google Scholar] [CrossRef] [PubMed]
  178. Abid, S.; Hussain, T.; Nazir, A.; Zahir, A.; Khenoussi, N. Acetaminophen loaded nanofibers as a potential contact layer for pain management in Burn wounds. Mater. Res. Express 2018, 5, 085017. [Google Scholar] [CrossRef]
  179. Xie, J.; Wang, C.H. Electrospun micro- and nanofibers for sustained delivery of paclitaxel to treat C6 glioma in vitro. Pharm. Res. 2006, 23, 1817–1826. [Google Scholar] [CrossRef] [PubMed]
  180. Hrib, J.; Sirc, J.; Hobzova, R.; Hampejsova, Z.; Bosakova, Z.; Munzarova, M.; Michalek, J. Nanofibers for drug delivery—Incorporation and release of model molecules, influence of molecular weight and polymer structure. Beilstein J. Nanotechnol. 2015, 6, 1939–1945. [Google Scholar] [CrossRef]
  181. Yu, D.G.; Xu, Y.; Li, Z.; Du, L.P.; Zhao, B.G.; Wang, X. Coaxial electrospinning with mixed solvents: From flat to round eudragit l100 nanofibers for better colon-targeted sustained drug release profiles. J. Nanomater. 2014, 2014, 967295. [Google Scholar] [CrossRef]
  182. Xie, Z.; Buschle-Diller, G. Electrospun poly (D,L-lactide) fibers for drug delivery: The influence of cosolvent and the mechanism of drug release. J. Appl. Polym. Sci. 2010, 115, 1–8. [Google Scholar] [CrossRef]
  183. Repanas, A.; Wolkers, W.F. Coaxial Electrospinning as a Process to Engineer Biodegradable Polymeric Scaffolds as Drug Delivery Systems for Anti-Inflammatory and Anti-Thrombotic Pharmaceutical Agents. Clin. Exp. Pharmacol. 2015, 5, 1–8. [Google Scholar] [CrossRef]
  184. Zeng, J.; Yang, L.; Liang, Q.; Zhang, X.; Guan, H.; Xu, X.; Chen, X.; Jing, X. Influence of the drug compatibility with polymer solution on the release kinetics of electrospun fiber formulation. J. Control. Release 2005, 105, 43–51. [Google Scholar] [CrossRef] [PubMed]
  185. Wang, Y.; Qiao, W.; Wang, B.; Zhang, Y.; Shao, P.; Yin, T. Electrospun composite nanofibers containing nanoparticles for the programmable release of dual drugs. Polym. J. 2011, 43, 478–483. [Google Scholar] [CrossRef]
  186. Natu, M.V.; de Sousa, H.C.; Gil, M.H. Effects of drug solubility, state and loading on controlled release in bicomponent electrospun fibers. Int. J. Pharm. 2010, 397, 50–58. [Google Scholar] [CrossRef]
  187. Taepaiboon, P.; Rungsardthong, U.; Supaphol, P. Drug-loaded electrospun mats of poly(vinyl alcohol) fibres and their release characteristics of four model drugs. Nanotechnology 2006, 17, 2317–2329. [Google Scholar] [CrossRef]
  188. Wei, J.; Hu, J.; Li, M.; Chen, Y.; Chen, Y. Multiple drug-loaded electrospun PLGA/gelatin composite nanofibers encapsulated with mesoporous ZnO nanospheres for potential postsurgical cancer treatment. RSC Adv. 2014, 4, 28011–28019. [Google Scholar] [CrossRef]
  189. Meinel, A.J.; Germershaus, O.; Luhmann, T.; Merkle, H.P.; Meinel, L. Electrospun matrices for localized drug delivery: Current technologies and selected biomedical applications. Eur. J. Pharm. Biopharm. 2012, 81, 1–13. [Google Scholar] [CrossRef]
  190. Yoo, H.S.; Kim, T.G.; Park, T.G. Surface-functionalized electrospun nanofibers for tissue engineering and drug delivery. Adv. Drug Deliv. Rev. 2009, 61, 1033–1042. [Google Scholar] [CrossRef]
  191. Miraftab, M.; Saifullah, A.N.; Çay, A. Physical stabilisation of electrospun poly(vinyl alcohol) nanofibres: Comparative study on methanol and heat-based crosslinking. J. Mater. Sci. 2015, 50, 1943–1957. [Google Scholar] [CrossRef]
  192. Tamizi, E.; Azizi, M.; Dorraji, M.S.S.; Dusti, Z.; Panahi-Azar, V. Stabilized core/shell PVA/SA nanofibers as an efficient drug delivery system for dexpanthenol. Polym. Bull. 2018, 75, 547–560. [Google Scholar] [CrossRef]
  193. Kenawy, E.R.; Abdel-Hay, F.I.; El-Newehy, M.H.; Wnek, G.E. Controlled release of ketoprofen from electro-spun poly (vinyl alcohol) nanofibers. Mater. Sci. Eng. A 2007, 459, 390–396. [Google Scholar] [CrossRef]
  194. Alizadeh, H.; Ahmadi, M.; Shayesteh, O.H. On chip synthesis of a pH sensitive gefitinib anticancer drug nanocarrier based on chitosan/alginate natural polymers. Sci. Rep. 2024, 14, 772. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Types of cancer.
Figure 1. Types of cancer.
Nanomaterials 14 01305 g001
Figure 2. Types of cancer treatments.
Figure 2. Types of cancer treatments.
Nanomaterials 14 01305 g002
Figure 3. Drug delivery in human beings.
Figure 3. Drug delivery in human beings.
Nanomaterials 14 01305 g003
Figure 4. Applications of nanofibers in the medical field.
Figure 4. Applications of nanofibers in the medical field.
Nanomaterials 14 01305 g004
Figure 5. Commonly used polymers for nanofibers for cancer treatment.
Figure 5. Commonly used polymers for nanofibers for cancer treatment.
Nanomaterials 14 01305 g005
Figure 6. Production of nanofibers through electrospinning.
Figure 6. Production of nanofibers through electrospinning.
Nanomaterials 14 01305 g006
Figure 7. Control of drugs by polymers.
Figure 7. Control of drugs by polymers.
Nanomaterials 14 01305 g007
Figure 8. Controlled and uncontrolled drug delivery mechanism.
Figure 8. Controlled and uncontrolled drug delivery mechanism.
Nanomaterials 14 01305 g008
Figure 9. Electrospinning method to prepare nanofibers web for cancer treatment.
Figure 9. Electrospinning method to prepare nanofibers web for cancer treatment.
Nanomaterials 14 01305 g009
Table 1. Commonly used drugs in cancer treatment.
Table 1. Commonly used drugs in cancer treatment.
Drug NameApplication(s)
PaclitaxelBreast, ovarian, lung, and pancreatic cancers
ImatinibChronic myeloid leukemia (CML), gastrointestinal stromal tumors (GISTs)
TamoxifenBreast cancer (especially hormone receptor-positive)
RituximabNon-Hodgkin lymphoma, chronic lymphocytic leukemia (CLL)
CisplatinVarious solid tumors, including testicular and ovarian cancers
TrastuzumabHER2-positive breast cancer, gastric cancer
MethotrexateVarious cancers, including leukemia and lymphomas
BevacizumabColorectal, lung, breast, and kidney cancers
Table 2. Parameters for nanofiber production through electrospinning.
Table 2. Parameters for nanofiber production through electrospinning.
ParameterGeneral RangeRole in Electrospinning
Temperature (°C)20–30Affects solvent evaporation rate and solution viscosity. Higher temperatures can dry fibers faster but may also lead to defects.
Viscosity (cP)100–1000Influences jet stability and fiber diameter. Higher viscosity generally leads to thicker fibers.
Flow rate (µL/min)0.1–10Controls the amount of solution pumped and affects fiber diameter and morphology.
Voltage (kV)5–30Determines the electrostatic force on the jet, influencing fiber stretching and diameter. Higher voltage generally leads to thinner fibers.
Needle collector distance (cm)5–20Affects solvent evaporation and fiber alignment. Shorter distance can lead to thicker fibers and slower drying.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Khan, M.Q.; Alvi, M.A.; Nawaz, H.H.; Umar, M. Cancer Treatment Using Nanofibers: A Review. Nanomaterials 2024, 14, 1305. https://doi.org/10.3390/nano14151305

AMA Style

Khan MQ, Alvi MA, Nawaz HH, Umar M. Cancer Treatment Using Nanofibers: A Review. Nanomaterials. 2024; 14(15):1305. https://doi.org/10.3390/nano14151305

Chicago/Turabian Style

Khan, Muhammad Qamar, Muhammad Abbas Alvi, Hafiza Hifza Nawaz, and Muhammad Umar. 2024. "Cancer Treatment Using Nanofibers: A Review" Nanomaterials 14, no. 15: 1305. https://doi.org/10.3390/nano14151305

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop