Abstract
Breast cancer is the most frequently happening cancer and the most typical cancer death among females. Despite the crucial progress in breast cancer therapy by using Chemotherapeutic agents, most anti-tumor drugs are insufficient to destroy exactly the breast cancer cells. The noble method of drug delivery using nanoparticles presents a great promise in treating breast cancer most sufficiently and with the least harm to the patient. Nanoparticles, with their spectacular characteristics, help overcome problems of this kind. Unique features of nanoparticles such as biocompatibility, bioavailability, biodegradability, sustained release, and, most importantly, site-specific targeting enables the Chemotherapeutic agents loaded in nanocarriers to differentiate between healthy tissue and cancer cells, leading to low toxicity and fewer side effects. This review focuses on evaluating and comprehending nanoparticles utilized in breast cancer treatment, including the most recent data related to the drugs they can carry. Also, this review covers all information related to each nanocarrier, such as their significant characteristics, subtypes, advantages, disadvantages, and chemical modification methods with recently published studies. This article discusses over 21 nanoparticles used in breast cancer treatment with possible chemical ligands such as monoclonal antibodies and chemotherapeutic agents binding to these carriers. These different nanoparticles and the unique features of each nanocarrier give the researchers all the data and insight to develop and use the brand-new drug delivery system.
Keywords: Breast cancer, nanoparticles, chemotherapeutic agents, immunotherapy, site-specific tumor targeting, targeted therapy
Introduction
Cancer immunotherapy is the non-natural stimulus of the immune system to cure cancer, boosting the immune system’s natural ability to eradicate ailments, which is common in the treatment of bladder cancer, breast cancer, kidney cancer, cervical cancer, brain cancer, head and neck cancer and colorectal and esophageal cancers and many other types of cancer [1,2]. Cancer immunotherapy has altered the standard for cancer cure; such treatments focus on enhancing anti-tumor immune reflexes with more insignificant off-target impacts than chemotherapies and some other mediators that straightly eliminate cancer cells [3-5]. This method has shown promising results in cancer remedies. Nevertheless, patient response rates continue to vary for reasons that are not well understood; however, there have been some assumptions for this phenomenon, such as immune competency and variety, differing antigen specificity and expression levels, and gut microbiota [6]. The use of cancer immunotherapy to trigger the immune system to identify and eradicate malignancies has provided novel promises for successful cancer treatment [7,8]. Immunotherapies that increase the capability of endogenous T cells to terminate cancer cells have established therapeutic effectiveness in a diversity of human malignancies [9,10]. Nanoparticles have lured significant consideration and display excessive potential in the field of malignancy immunotherapy. NPs used as transporters can carry immune cargo, such as antigens, gene therapeutics, and proteins to the intended position. Compared to specific immunotherapy, NP-based immunotherapy boosts a more brutal immune response and has higher specificity and effectiveness [11,12]. Such nanosized structures are anticipated to participate in imaging, pursuing, and observing abilities with the targeted transfer of mixtures to tumors, cellular purposes and procedures, or precise organs [13-17]. Breast cancer is a severe ailment in women and is the top reason for mortality. This malignancy is a heterogeneous illness, so stratification of tumors is essential to attain better clinical outcomes [18]. Metastasis is primarily responsible for its incurableness [19,20]. Epidemiologic studies have acknowledged the diversity of breast cancer risk factors such as race, ethnicity, family history of cancer, genetic traits, and changeable contacts such as augmented alcohol consumption, physical sedentariness, exogenous hormones, and confident females generative factors [21]. Cancer immunotherapy is an innovative method that promotes the host immune system to identify and eliminate highly selected cancer cells. However, there is no comprehensive assessment of methods used to eradicate tumoral cells. This article tends to deliberate using of nanoparticles as too invasive tumoral cells and the effectiveness of this method. In this review, contemporary findings of nanomaterials used in breast cancer immunotherapy have been widely classified and extensively included through distinct sectors. These classifications are immensely helpful for a comprehensive understanding of used particles and mechanisms of theranostics in this era.
Breast cancer
Breast cancer is the most frequently happening cancer and is the most typical cancer death amole females. His disease is a multifaceted ailment that shows a considerable degree of inter- and intra-tumoral heterogeneousness [18,22-25]. According to the data from WHO, in 2020, there were 2.3 million women identified with breast cancer and 685,000 losses worldwide. As of the end of 2020, 7.8 million females were thriving who were detected with breast cancer in the past five years, making it the world’s most predominant cancer [26] (Figure 1). Breast cancer positions the fifth reason for mortality from malignancy. While it is the most common reason for cancer death in females in less industrialized districts, it is now the second cause of cancer death in more industrialized communities after lung cancer [27,28]. There are numerous risk factors for breast cancer such as aging, genetic mutations, having dense breasts, the unique history of breast cancer or specific non-cancerous breast diseases, family history of breast or ovarian cancer, preceding treatment using radiation therapy, and consumption of diethylstilbestrol drug [29]. Breast cancer’s common symptoms in women are a new lump in the breast or underarm (armpit), thickening or swelling of the breast, irritation of breast skin, redness or flaky skin in the nipple part of the breast, pulling in of the nipple or discomfort in the nipple area, nipple discharge except for breast milk, including blood, any alteration in the size or the shape of the breast, ache in any part of the breast [30]. The experiment of Joan R. Bloom showed that breast cancer exerts physical distresses and emotional distresses. Emotional support is particularly significant during the acute recovery period for one’s mental health, whereas instrumental support gives promising results for women who have poorer physical and psychological healthiness [31].
Figure 1.

Most common cancers among women.
Types of breast cancer
There are different kinds of breast cancer encompassing non-invasive and invasive cancers. Non-invasive breast cancer includes Ductal carcinoma in situ, and Lobular carcinoma in situ and invasive breast cancer contains Invasive ductal carcinoma, invasive lobular carcinoma, Paget’s disease of the nipple, Inflammatory breast cancer, Phyllodes tumors of the breast, locally advanced breast cancer, and metastatic breast cancer [32,33] (Figure 2).
Figure 2.

Classification of breast cancer.
In the past years, traditional classifications encompassing histological valuation and staging were applied to establish the patient’s treatment method. Recently, molecular analysis of breast tumors has opened a gate for further understanding breast cancer biology and classification [34,35]. Also, targeted-gene sequencing has emerged as the most novel and exact approach to determine the class of breast cancer. This next-generation method has helped extensively evaluate the mutations in breast tumor cells [36].
Molecular classification
Through molecular analysis of breast cancers with gene expression profiling, breast cancer could be sub-classified into diverse subtypes. Generally, these subtypes comprise luminal ER (estrogen receptor) positive (luminal A and luminal B), human epidermal growth factor receptor 2 (HER2) enriched, and basal-like or triple-negative [22,37,38]. Luminal A breast cancer is hormone-receptor-positive (estrogen-receptor and progesterone-receptor positive), HER2 negative, and has trivial levels of the protein Ki-67, which helps control how fast cancer cells grow [39]. Luminal B breast cancers are categorized by a minor ER expression, a slight progesterone receptor (PgR), and a high histologic grade [40,41]. HER2-enriched breast cancer is hormone-receptor negative (estrogen-receptor and progesterone-receptor negative) and HER2 positive [42]. Triple-negative/basal-like breast cancer is HER2 negative and hormone-receptor negative (estrogen-receptor and progesterone-receptor negative) [43] (Table 1).
Table 1.
Classification of molecular subtypes
| Molecular subtype | ER-estrogen receptor | human epidermal growth factor receptor 2 | PR-progesterone receptor | Ref. |
|---|---|---|---|---|
| Luminal A | positive | negative | positive | [39] |
| Luminal B | minimum | positive/negative | minimum | [40,41] |
| HER2 | negative | positive | negative | [42] |
| Triple-negative or basal-like | negative | negative | negative | [43] |
Targeted-gene sequencing
Targeted-gene sequencing is a revolutionary DNA sequencing practice aiming at amplicons and particular genes [44]. Comparing and analyzing the Genome of the DNA and gene expression of primary cancer cells and metastases in numerous cases can help estimate the mechanisms that cause metastasis [45]. In addition, targeted-gene sequencing has facilitated the prediction of future recurrence sites [46,47], precise treatment based on molecular features, response to therapy, and survival [48]. Targeted gene sequencing panels are used to determine the exact mutation in a sample. Focused conferences hold the genes or regions suspected to cause breast cancer [49].
Classification of particles
Microparticles used in drug delivery
Microparticles (MPs) are dense or dispersed particles ranging from 1 to 1000 µm [50]. The medication is dissolved, trapped, encapsulated, or connected to a microparticle matrix. Dependent on the technique of composing, microparticles, microspheres, or microcapsules can be produced [51,52]. Like nanoparticles, microparticles are used as drug transporters or as adjuvants for vaccines. The medications or antigens may combine into particles in the shape of a dense dispersion or a solid solution [53]. Also, they may be adjoined to the particle exterior by physical adsorption and chemical attaching [54]. These transportation systems present several benefits contrasted to common dosage forms, which contain enhanced effectiveness, decreased toxicity, and bettered patient convenience [55]. Some critical challenges confronting immunotherapy include inaccurate toxicity and inexact immune activation [56]. To help undertake this problem, nanoparticles and microparticles play an essential role as effective drug transporting structures for immunotherapies to regulate the immune system [57]. N.P./MPs include many kinds; the most important ones are polymeric particles and liposomes. The majority of the typical polymeric particles are composed of chitosan, biodegradable polyesters, altered Dextrans, and Polyketals [58]. Most of the biodegradable polyesters (e.g., poly lactic-co-glycolic acid (PLGA)) are drug delivery tools that are approved by Food and Drug Administration [59].
Nanoparticles used in drug delivery
Nanoparticle drug delivery systems are professional methods that transport medication to the intended cells and control the release of drugs [60]. The recent procedure of a drug delivery method tends to lessen side effects and decrease the dosage. Lately, different NPs, as depicted in (Figure 3), have stimulated interest because of their possible use for efficient drug delivery [61]. Boosting ligand binding effectiveness can reduce dosage and decrease NP. Toxicity Reducing dosage or dosage frequency also lessens the mass of nanoparticles per mass of medication; therefore, attaining more energy [62]. According to the characterization from NNI (National Nanotechnology Initiative), nanoparticles are systems of sizes fluctuating between 1 to 100 nm in as a single minimum dimension [63].
Figure 3.

A summary of the nanoparticle types used in breast cancer drug delivery. In general, organic and inorganic nanoparticles are the two primary nanoparticles used for drug delivery. Also, polymeric NPs are the most frequently used particles that contain nanocapsules as the most valuable object to deliver the drug to the intended specific breast tumor cells.
Nevertheless, the prefix “nano” usually refers to particles up to several hundred nanometers in size. Nanoparticles with enhanced physicochemical and biological features are absorbed in cells more effortlessly than giant molecules [64]. Thus, nanoparticles could be effectively utilized as transporting means for presently accessible bioactive compounds [60]. NPs can load different components at the same time for simultaneous transport, keep the loads from degradation and early release, and inactively or actively target cancer cells by the enhanced permeability and retention (EPR) influence or surface alteration by ligands correspondingly [61] (Figure 4). Also, inorganic nanoparticles could be utilized as a local source of ICD-inducing cure or focus attention on a treatment by external energy fields to diminish harm to normal cells [65,66].
Figure 4.

It indicates nanoparticle-mediated drug delivery to tumor cells, especially breast cells and summarizes how NPs reach the target organ and stimulate the destination cells. NPs have been utilized for effective drug delivery, analytic tools and facilitate an efficient, targeted biomolecular interface to lessen side effects caused during the treatment.
Due to the lack of full clinical application support and intratumorally administration of these nanoparticles, most nanocarriers’ use remains only in vivo and in vitro stage. According to the academic journals, there has been a hope that vascular interventional administration might be a possible solution to utilize NPs by locating the tumor-feeding vessels [67]. A summary in (Table 2). is brought to compare these nanoparticles in detail.
Table 2.
The summary of the advantages and disadvantages of each nano-carriers with their variable size
| Np Type | Size | Advantage | Disadvantage | Ref |
|---|---|---|---|---|
| Polymeric Nanoparticles | Biodegradable, biocompatible, Efficacious distribution of both water-soluble and insoluble medications. | Cytotoxicity, Organizational heterogeneity as reproduced by high polydispersity index. | [229] | |
| Nanospheres | 100-200 nm | Significant external to volume ratio, Measured release of insoluble actives. | Absence of Stability of some actives, High manufacture expenditure. | [230-232] |
| Nanocapsules | 5-1000 nm | The usage of natural polymers such as polysaccharides and proteins can rise bioavailability and biodegradability. | Extensive dispersal of condensed actives, a purification procedure is required after the synthesis of nanocapsules. | [232-234] |
| Dendrimers | 1-100 nm | Functionalization of outlying groups control solubilization and permits targeted delivery of load-Appropriate for combining lipophilic and lipophobic cargo. | Toxicity linked with surface amin groups-Pharmacokinetics, biodistribution, biodegradation, and chronic toxicity of PAMAM is not understood yet. | [235] |
| Micelles | 20--100 nm | Self-assembling, thermodynamic constancy, targeting potent. | Selection of appropriate surfactants. | [236] |
| Polymersomes | 100 nm to a few μm | highly adaptable and biologically steady systems and their overall possessions and drug encapsulation and release competencies can be effortlessly tuned by applying numerous block copolymers that are biodegradable and/or stimuli-responsive. | More clinical studies are vital for its formation as gold standard avenues. | [237] |
| Solid lipid Nanoparticles | 50--100 nm | Progresses solubility in water of hydrophobic cargo, Hydrophilic cargo conceivable, Relatively low-cost manufacture, Biocompatible/biodegradable, Possible production scaling-up. | Recrystallization danger and little encapsulation load, High water content in dispersals (70-99.9%), Premature cargo release during storing. | [238,239] |
| Liposomes | 30 nm to a few μm | Effective delivery of both water-soluble and insoluble drugs, simply tailored size and carrying capacity, Significant construction. | Swift release, Petite shelf lives, Variability, clearance to reticuloendothelial structure. | [229,240,241] |
| Metal Nanoparticles | 1 nm to a few hundreds of nm | Uniformity in scope, shape, and branch length Tuned p harmacokinetics and biodistribution Augmented surface area, enlarged loading Targeting is achieved. | Poisonous effects on the body. | [229,235] |
| Carbon Nanotubes | About 0.7 nm | Multiple roles Chemical alteration Water soluble and biocompatible Efficient cargo. | Poisonousness. | [235] |
| Ceramic Nanoparticles | 1--100 nm | Do not swell or change porosity and are steady at numerous pH and temperatures. | Sluggish biodegradation or non-degradation. | [242,243] |
| Human serum albumin (HSA) Nanoparticles | 66.5 kDa | Low toxicity, biodegradability, reproducibility, manageable release, and numerous drug binding sites. | the potential risk of pathogen contamination (e.g., HIV, hepatitis, CJD), side effects. | [219,221] |
Overall, the benefits overcome the drawbacks; however, the extended use of some nanoparticles is limited due to the toxicity.
Organic nanoparticles
Organic nanoparticles are developed upon organic or synthetic organic molecules. Nature provides many examples of organic NPs like protein combinations, lipid bodies, milk emulsions, or more intricate prepared structures such as viruses [68,69]. The principal value of devising medications into nanoparticles is enhancing particle exterior surface in touch with the dissolution agent, enhancing bioavailability. Numerous drugs have been formulated with this method and are available [70]. By using an innovative technique called SMILE (Stabilized Metal Ion Ligand complex), Scientists have produced a biomimetic nanoparticle formulation of Cu(DDC)2 to overcome drug delivery issues [71]. Cu(DDC)2 metal-organic compound core and surface covered bovine serum albumin (BSA) are integrated to make Metal-organic Nanoparticle (MON), which efficiently prevents the outgrowth of breast cancer cells [72,73].
Polymeric nanoparticles
Polymeric NPs have drawn significant attention over the latest years because of their features due to their tiny scale [74-76]. Possible usage for controlled drug release, the capability to retain medication and other molecules with biological function from the environment, enhance their bioavailability, and therapeutic index are benefits of polymeric nanoparticles in drug delivery [77]. Nanocapsules and Nanospheres are two types of polymeric nanoparticles. Polymer nanoparticles are one of the most advanced non-invasive methods for drug delivery uses [78]. Polymeric nanoparticles are units within the size variation from 1 to 1000 nm and can be filled with effective compounds trapped inside or surface-adsorbed onto the polymeric core [77]. Polymeric nanoparticles can control drug release either by diffusion via polymer matrix or matrix degradation. They have been inspected as drug-delivery systems for the site-specific targeting of cancer cells [79]. The most significant benefits for these particles are supplying controlled release to the preferred site, supply stability to labile molecules (e.g., proteins), and supply capability to alter surfaces with ligands for stealth and targeted drug delivery goals [80]. Due to the poor water-solubility, three of the most critical drugs in breast cancer delivered by polycaprolactone-polyethylene glycol (PCL-PEG) nanoparticles are Cisplatin, Doxorubicin, and 5-fluorouracil. PCL-PEG is a polymeric nanoparticle that increases the stability and solubility of drug molecules to enhance drug delivery systems [81,82]. Bressler and his colleagues have shown that AXT050 is a multi-modal peptide with anti-tumorigenic and antiangiogenic properties by targeting integrin αVβ3 on the surface of cells in culture. It also can target and disturb both cancer cells and endothelial cells; therefore, this technology may be capable of administrations in cancer nanomedicine [83]. Abou-El-Naga’s experiment has represented that cellular uptake of Docetaxel was time-dependent and reached the maximum after conjugating on PLGA NPs and with folic acid combination, which triggered the endocytosis mechanism; therefore, folic acid/PLGA NPs can have a promising drug delivery system for Docetaxel in breast cancer treatment [84]. Soe ZC and her colleagues’ study has shown that transferrin-conjugated polymeric nanoparticle-targeted NP used as a doxorubicin carrier into a drug-resistant cell line has (a transferrin (Tf)-conjugated polymeric nanoparticle composed of poloxamer 407 (F127) and 123 (P123) (Dox/F127&P123-Tf)) improved cellular uptake and tempted prohibition of cell propagation in vitro, not only in doxorubicin-sensitive cells but also in the doxorubicin-resistant in a particular type of breast cancer cells. Hence, transferrin-targeted NPs can be utilized as harmless and effective drug carriers to treat doxorubicin-sensitive and resistant breast tumor cells [85].
(1) PEG
The most helpful polymer for drug delivery is Polyethylene glycol (PEG) because of its unique behavior that prevents early recognition by the immune system (opsonization) [86]. Also, the hydrophilic characteristic of PEG leads to stabilizing nanoparticles by steric and not ionic effects, especially in water [87]. Carrying PEGylated nanoparticles with drugs to epidermal growth factor receptor (EGFR) + Triple-negative breast cancer (TNBC) requires binding PEG engager to polyethylene glycol and EGFR simultaneously. Enhancing the anti-proliferative activity of PEG-liposomal doxorubicin by PEG engagement in EGFR+TNBC cells brings about a reduction in breast tumor cells’ growth and reductions in their number [88-90].
(2) PCL
Poly-ε-caprolactone (PCL) is another frequently used polymer nanocarrier decomposed by hydrolysis of its ester. Versatile nature, ease of fabrication, and biocompatibility are the features that have made PCL the center of attention in nanoparticle drug delivery [91,92]. This nanocarrier is most suitable for long-term delivery up to one year because of its unique characteristic in slow biodegrading compared to other polymers [93]. NPs based on Poly (ε-caprolactone)-Poly (ethylene glycol)-Poly (ε-caprolactone) (PCL-PEG-PCL) can efficiently deliver Doxorubicin (DOX), an anthracycline anti-cancer drug, to the targeted breast tumor cells [94]. A study has shown synthesis of robust docetaxel/aptamer-polydopamine (DTX/Apt-pD)-CA-(PCL-ran-PLA) NPs with star-shaped CA-(PCL-ran-PLA) copolymers, which could be practical as optimistic targeting drug delivery systems for synergistic chemo-photothermal therapy of breast cancer [95].
(3) PEG-PCL
Cuong N-V and colleagues showed that three biodegradable PEG-PCL-PEG triblock copolymers with many PCL blocks were manufactured to transport anti-cancer drugs. The structures of copolymers were characterized by proton nuclear magnetic resonance, Fourier transforms infrared spectroscopy, gel permeation chromatography, differential scanning calorimetry, and X-ray diffraction, correspondingly. These hydrophobic PCL elements contained copolymers that could compress DOX into the micelle cores. The cytotoxicity of DOX-loaded micelles was higher than that of free DOX in specific breast cancer cells. These explanations show that DOX-loaded micelle is a hopeful means of treating multidrug-resistant tumors [96]. Additionally, in a study, mPEG-PCL copolymer was created. The copolymer mPEG-PCL was self-assembled into polymersomes in an aqueous solution in the presence of Methotrexate. Assays showed that methotrexate-loaded mPEG-PCL polymersomes had more prohibition effects on breast cancer cells than free Methotrexate, providing an appropriate and proper system for delivering Methotrexate to the breast cancer cells [97].
(4) Chitosan
Chitosan is a polymeric nanocarrier that is biodegradable, biocompatible, and vastly used in drug delivery with diverse administration methods. Positive surface charge and mucoadhesive feature enable chitosan NPs to connect to mucus membranes and release the loaded drug in a sustained release way [98,99]. Other beneficial characteristics of chitosan are low immunogenicity and high biocompatibility, along with a high cationic charge [100,101]. Additionally, chitosan is used to overcome the drug resistance and increased toxicity of some sufficient anti-cancer medications such as 5-Fluorouracil and doxorubicin [102]. A water-soluble product of chitosan called carboxymethyl chitosan (CMCS) can load 5-Fluorouracil and doxorubicin and facilitate the sustained release of these drugs [103]. Mono-dispersed and pH-sensitive chitosan silica hollow nanospheres (CSeSiO2 HNPs) have shown promising results in breast cancer therapy. Antibody molecule (to ErbB 2) is conjugated with SiO2 HNPs to a pH-sensitive polyelectrolyte layer to make a suitable nano-transporter for targeted TNF drug transport to breast cancer cells [104,105]. In Santos-Carballal B’s study, chitosan oligosaccharide (COS) showed promising results in preventing the aggressive capability of MDA-MB-231 breast tumor cells, which leads to a reduction in the metastasis process [106]. Even in Cancer gene therapy, chitosan is outstandingly valuable through specific nano complexes like chitosan-hsa-miRNA-145 (CS-miRNA), which downregulates the target mRNA proliferation in MCF-7 breast cancer cells [107].
(5) Gelatin
Gelatin is a versatile and natural biopolymer that has numerous significant functions because of its low price, easy accessibility, biodegradable and biocompatible characteristic, non-toxicity, easy adhesion, and adjusting chemically and consequently has a vast capacity to be utilized in drug delivery systems such as nanoparticles which contain protein [108-110]. Amifostine is used to firmly connect a targeting ligand (Herceptin) to amphiphilic gelatin (AG)-iron oxide calcium phosphate (CaP) NP which produces a nanoparticle system with a pH-sensitive CaP shell and degradable AG core, enabling the manageable sustained release of the two medicines [111]. A bioligand and magnetic targeting with the dual-targeting system of AGIO@CaP-CD (HER-AGIO@CaP-CD) led to a considerably high cellular uptake in HER2-overexpressing SKBr3 cells and more effective treatment for breast cancer [112].
(6) Poly-D, L-lactide-co-glycolide (PLGA)
Because of the adjustable and sustained-release characteristics, low toxicity, and biocompatibility with tissue and cells, US Food and Drug Administration have approved Poly PLGA as one of the most efficient biodegradable polymeric NPs to be utilized in drug delivery structures [113]. Hydrolyzed PLGA in the body produces lactic acid and glycolic acid, biodegradable metabolite monomers. The body deals with these two monomers so quickly, leading to the low toxicity of PLGA [114]. Also, in cancer treatment, incorporating Docetaxel into the rod-shaped PLGA improves the efficacy of taxane-resistant triple-negative breast cancer [115].
(7) PLGA-PEG polymeric nanoparticles
In Akbari E’s research, trapoxin/Methotrexate-co-loaded PLGA-PEG NPs were synthesized. The trapoxin/Methotrexate co-loaded PLGA-PEG displayed high growth inhibition against breast cancer cells; consequently, it showed good accomplishment, which supports using the system as a good candidate in the synergistic delivery of antineoplastic agents to treat breast cancer [116]. In another experiment done by Amirsaadat S and colleagues, PLGA-PEG NPs were used to co-deliver two natural anti-cancer agents, Metformin and Silibinin, against breast cancer cells. Outcomes disclosed that the encapsulation of Metformin and Silibinin in PLGA-PEG NPs could efficiently hinder the propagation of breast cancer cells than their pure forms [117]. Jusu SM and colleagues experimented with releasing targeted and untargeted (PGS and PTX) cancer drugs from physical blends of PLGA and PEG microparticles. The outcomes presented that the composite microcapsules allow the prolonged release of cancer drugs (PGS, PTX, PGS-LHRH, PTX-LHRH) over time that could significantly ease the localized treatment of TNBC [118]. Results show that nanochrysin or chrysin-loaded PLGA-PEG used in T47D and MCF7 cell lines apply a repressing effect on the two breast cancer cell lines, further than that of pure chrysin. Based on these outcomes, nanochrysin can be utilized for breast cancer therapy and proposes a novel and effective drug delivery system to contest breast cancer [119]. In Cao D’s work, injectable liposomal doxorubicin-loaded PLGA-PEG-PLGA thermogel was synthesized. Liposomal doxorubicin-loaded thermogel exhibited protracted release of DOX deprived of separate initial burst compared with DOX-Gel. This study suggests that a hybrid medication delivery system containing liposome and hydrogel can sustain and enhance drug release, improving the anti-cancer effectiveness through localized therapy and reducing cytotoxicity [120]. Tabatabaei Mirakabad FS’s study showed that curcumin-loaded PLGA-PEG nanoparticles have an inhibitory effect on the MCF-7 breast cancer cell line more than pure curcumin [121].
(8) Polylactic acid (PLA)
PLA is a thermoplastic polyester nanoparticle that has drawn attention due to its biocompatibility, non-toxicity, and biodegradability features. Poly-lactic acid nanoparticles (PLA-NP) are commonly used as nanomedicines because of their benefits over metallic NP, such as keeping anti-cancer drug loads for sustained periods [122]. The anti-cancer drug paclitaxel (PTX) can be loaded to PLA/hydroxyapatite (HAp, which exhibits pH sensitivity) core-shell nanoparticles and facilitate effective drug delivery via the EPR effect to the breast tumor cells [123].
(9) Nanosphere
Nanospheres must be assumed to the matrix-based structures [124], which are different in size from 10 to 200 nm in diameter and can be crystalline or amorphous. They integrate with drugs, dissolve, encapsulate, or join bioactive to the polymeric matrix compound. These NPs guard bioactive and keep them from degrading chemicals and enzymes [125]. Hollow carbon nanospheres (HCNs) modified with anti-HER2 antibody and loaded with DOX facilitates the drug delivery (Figure 5) to the specific HER2-positive breast cancer cells and considerably restrain the tumor cells in vitro, causing ~a 60% decrease in the size of the HER2-positive tumors in vivo [126]. To overcome some of the typical restrictions of breast cancer treatment like off-targeted drug delivery, fast drug clearance, and drug resistance, porous magnetite nanospheres are used to load Doxo with great effectiveness for targeted drug transport to breast cancer cells depending on pH-sensitive drug release [127].
Figure 5.

It summarizes the two agents working together to fight against breast tumor cells. The first one is the body’s immune system which produces CD8+ T cells based on the antigen of the tumor cells presented by DCs to T cells. The second agent is immune checkpoint inhibitors delivered by nanoparticles to the organ. CD8+ T cells are then capable of attacking cancer cells and eliminating them with the help of immune checkpoint inhibitors carried to the position by nanoparticles like nanosphere.
(10) Nanocapsule
A polymeric nanocapsule is a liquid or solid core with a polymeric shell covering the whole structure, its main difference from the nanosphere (Figure 6) [128]. These nanoparticles can transport enzyme biocatalysts, drugs, and vectors to tumor cells [129]. They have a spherical form; drugs are located in the unfilled center and coated with polymeric shells [130]. The size of these particles differs from 50 to 300 nm. Nanocapsules can transport both hydrophilic and lipophilic drugs [131]. Lipid nanocapsules can efficiently transport hydrophobic drugs like Docetaxel and Thymoquinone to the intended breast tumor cells. These nanoparticles considerably improve the anti-tumor effect of free Docetaxel in breast cancer cells [132]. Utilizing Chitosan Grafted Lipid Nanocapsules loaded with Docetaxel and Thymoquinone is a novel method to enhance chemotherapeutic effectiveness in treating resistant breast tumor cells, which has shown promising results both in vivo and in vitro [133].
Figure 6.

The main differences between nanosphere and nanocapsules. Nanocapsules have vesicular structures that a drug is loaded in the unfilled center and coated with a polymeric shell, while nanospheres matrix-based structures with a drug dispersed in it physically.
(11) Dendrimers
Dendrimers are created of repeating components, like polymers. However, they are significantly different from usual polymers by two key features: dendrimers are never produced by polymerization reactions, but they have a flawlessly devised and vastly reproducible structure and a very branched 3D design because of the usage of at least one kind of branching components as construction blocks for their synthesis [134,135]. The main difference between these two is that a dendron usually has a sole chemically addressa-ble assembly called the focal point or core. Dendrimers are built by adding layers to the branching groups [136]. The construction of dendrimers includes a core molecule with branching groups to which different branching molecules are attached in layers [137]. Because of their low toxicity, a polymeric dendrimer is of significant attention in biomedical uses such as polyamidoamine (PAMAM) dendrimers [138]. In HER2-positive breast cancer, Trastuzumab (TZ)-grafted dendrimers significantly enhance the transport of Docetaxel (DTX) to the tumor cells. These nanoparticles lead to higher antiproliferation activity, cellular internalization, and induction of apoptosis against HER2-positive breast cancer cells [139-141].
(12) Micelles
Polymeric micelles are widely investigated transporters to carry weakly water-soluble medications [142]. Amphiphilic block copolymers shape Polymeric micelles nanosized core/shell systems. The reasons that make these nanoparticles exceptionally well suitable for drug delivery uses are both the natural and adjustable features of polymeric micelles [143]. Polymeric micelles are applied in drug delivery because of their appealing qualities, like biocompatibility, less toxicity, core-shell assembly, micellar association, morphology, nano size, and relatively high steadiness [144]. The fundamental notion in this method is receptor-mediated endocytosis. The ligands coupled to the micelles attach to their exact receptors on the cell membrane, leading to the micelles’ endocytosis [145]. The variation in mass between the micelles and universe enables them for extended circulation in the blood with escaping of renal clearance, yet permits for a means of eliminating the surfactant from the body when no longer needed [143]. IR780 iodide, a near-infrared (NIR) dye delivered to the tumor cells by micelles, has shown magnificent ability in the phototherapy of cancer cells. Herein, amphiphilic 28 micelles based on D-a-tocopheryl polyethylene glycol succinate (TPGS) and D-a-tocopheryl succinate 29 (TOS) can carry IR780 to the targeted malignant breast cells [146-148].
(13) Polymersome
Polymersomes are synthetic vesicles, small empty with a solution. These nanoparticles are made of amphiphilic synthetic block copolymers that shape the vesicle membrane and have sizes fluctuating from 50 nm to 5 µm [149].
Both hydrophilic doxorubicin hydrochloride (DOX·HCl) and hydrophobic doxorubicin base (DOX) can be delivered by polymersomes to the breast tumor cells [150].
Lipid Nanoparticles
(1) Solid lipid nanoparticles
Solid lipid nanoparticles (SLNs) have been produced to transport dissolvable water drugs and corrective dynamic medication efficiently. These NPs are made up of assembled quality polymers, making them capable of advanced drug delivery with less toxicity [151,152]. Doxorubicin encapsulated in lipid nanoparticles (SLN-Dox) goes through the cell membrane via endocytosis and arrives at the cytoplasm. This drug-NP complex overcomes the resistance mechanism in the cell membrane without the support of a chemosensitizer [153,154]. An examination that Bin Lu and Su-Bin Xiong did showed that conjugation of solid lipid nanoparticles with mitoxantrone could controllably release without burst effect and high effectiveness in the targeting of the drug to lymph nodes and a definite affinity for tumor tissues which assistances to improve the therapeutic outcome and diminish the side effects of anti-tumor agents [155]. In another examination that Oliveira da Rocha and Bento da Silva did show that a combination of solid lipid nanoparticles and Docetaxel can inhibit the tumor growth of 4T1 breast cancer; furthermore, it did not have noticeable systemic noxiousness in mice; consequently, it can be used for the treatment of breast cancer [156].
(2) Liposome delivery
Liposomes are spherical systems designed by one or multiple concentrical lipid bilayers surrounding separate aqueous spaces [157]. These NPs are usually manufactured with innately connected phospholipids, chiefly phosphatidylcholine. To amend the membrane’s firmness and enhance stability, cholesterol is often manufacturing liposomes. The molecular load is filled via liposome formation in aqueous solution, solvent substitute mechanisms, or pH gradients techniques [158]. Indeed, liposomes are the most valuable and complex NP class because of their ability to carry numerous biologically effective complexes and macromolecules [159-161]. Liposomes have exclusive features for drug delivery; actually, they can include an extensive diversity of hydrophilic and hydrophobic diagnostic or therapeutic mediators (Figure 7) [162]. In eliminating tumor cells, liposomes have been established to be very practical. The purpose behind that is liposomes decrease the toxic side effects of chemotherapeutic medications and increase their anti-cancer effectiveness [163]. For progressive chemotherapy of tumor cells, size is the critical feature of liposomes; Bigger liposomes are efficiently eliminated from circulation by the reticuloendothelial system (RES) [164]. Liposome-based chemotherapeutics can gather in tumor tissue with this “passive” drug transport [165]. Metformin (MET), an anti-diabetic drug, also has been effective against breast cancer. MET can be delivered to the tumor cells by MET-encapsulating liposome (LP-MET) and Herceptin-conjugated LP-MET (Her-LP-MET), which have shown promising results both in vivo and in vitro [166]. The Zeta potentials of LP-MET and Her-LP-MET and their size efficiently increase penetrability and retaining effects [167]. In the same way, PEGylated DSPC liposomes produced with the microfluidic system containing doxorubicin expose less toxicity because of their sustained release from the liposome [168].
Figure 7.

Liposomes are new drug delivery structures that have vesicular structures consisting of bilayers. These nanoparticles are made of aqueous spaces, which are surrounded by a membrane created of lipid bilayers.
Inorganic nanoparticles and nanocrystals
Inorganic Nanoparticles and Nanocrystals are made of crystalline clusters of atoms (from just a few to numerous tens of thousands). When the size of the resources is decreased to the nanometer scale, their natural features can be extraordinarily altered, and novel physical features can appear [169]. Inert metals such as gold and titanium shape nanospheres; nevertheless, iron oxide nanoparticles similarly turn out to be a choice. Several latest findings, like the nanoparticles’ intracellular degradation or the tight connection amid intracellular localization and local nanoparticle concentration and their cytotoxic consequence [170,171], offer new visions to undertake the influence of nanoparticles on cells. Metal inorganic nanoparticle structures break down into their component metal atoms, and these materials might have contact with biosystems. A significant quantity of these nanoparticles might stay in the body after treatment which can cause toxicity [172]. Inorganic NPs have exclusive properties, which contributes to the lessening of nontoxic photosensitizer (PS) leakage, allow for a high loading capacity of PSs, augment PS passive uptake, and allow for ease of functionalization with numerous ligands to promote active PS absorption and, therefore, allow for the overall improvement of photodynamic treatment of breast cancer treatment [173-175].
Metal nanoparticles
Metal nanoparticles are micron-sized systems containing metals like gold, silver, titanium, platinum, cerium, zinc, thallium, and iron, or their composites like hydroxides, oxides, sulfides, fluorides, chlorides, and phosphates [176]. Noble metal nanoparticles (Ag, Au, Pt) are useful for numerous biomedical uses like anti-cancer, radiotherapy enhancement, drug transfer, thermal ablation, and many others [177]. Ionic covalent bonding and physical absorption can attach gold nanoparticles (AuNPs) surfaces to drugs that are transferred to the tissue and regulate drug release via biological stimuli or light initiation [178]. Noble metal NPs have exclusive features like high surface-to-volume ratio, vast optical components, effortless manufacturing, and trouble-free surface chemistry, making them versatile for cancer treatment [179-182].
(1) Gold nanoparticles
Gold nanoparticles (AuNPs) are small gold particles with a diameter of 1 to 100 nm, which, once distributed in water, are also acknowledged as colloidal gold [183]. Gold nanoparticles can be utilized to enhance the biodistribution of drugs to diseased organs, tissues, or cells to improve target drug delivery [184,185]. Gold nanoparticles can accumulate in breast tumor tissue. Conjugated with drugs such as Trastuzumab, it can also be a drug carrier to augment uptake of the drug to the tumor tissue [186]. Sani A’s studies have shown that AuNPs are not poisonous; nevertheless, many other studies oppose this declaration; consequently, further studies are needed [187]. In Libutti SK’s experiment, a new nanomedicine was conjugated with human tumor necrosis factor-alpha (rhTNF) and thiolated PEG onto the surface of gold nanoparticles (named CYT-6091) [188]. The outcomes presented that it was less poisonous than treatment with only rhTNF. Moreover, the gold nanoparticles had accumulated in the tumor site and mostly avoided healthy tissue [189-191]. Zhang C’s studies represented that by using the aptitude of doxorubicin to interpolate DNA duplexes, the novel dual-drug encompassing DNA-Gold nanoparticles, Dox@affi-F/AuNPs (dual-drug-containing affibody-DNA-Gold nanoparticles), can exhibit outstanding constancy in simulated physiological circumstances, and it could target HER2 overexpressing cancer cells [192]. Studies have revealed a prominent cytotoxicity outcome of prepared chrysin functionalized metal- reduced graphene oxide nanocomposites against breast carcinoma cells than the normal cells via prompting apoptosis, and it holds an optimistic future for cancer chemotherapeutic purposes; however additional studies need to be carried out with different types of cancer cell lines [193].
(2) Silver nanoparticles
Silver nanoparticles are nanoparticles of silver between 1 nm and 100 nm in size. While frequently described as ‘silver’, some are manufactured of a large percentage of silver oxide due to their large surface to bulk silver atoms [194]. Silver nanoparticles transport breast cancer drugs due to their competent agents targeting tumor cells and risk-free, healthy tissues [195]. Swanner and Fahrenholtz’s examination for the first time showed that systemically administered AgNPs effectively reduce the growth of solid, TNBC mammary tumors in mice, which supports the likelihood that AgNPs may be beneficial for the treatment of some human breast cancers [196].
(3) Platinum nanoparticles
Platinum nanoparticles are frequently in the form of a suspension or colloid of nanoparticles of platinum in a fluid, usually water [197]. Examination of Manzoor and Junaid Bashir showed that platinum nanoparticles also revealed anti-metastatic potential on the breast cancer cell. In the cell cycle analysis, platinum nanoparticles stimulated cell cycle arrest at the G0/G1 phase. Platinum nanoparticles also showed antibacterial properties against pathogenic bacteria [198].
Carbon nanotubes
Carbon nanotubes (CNTs) have become widespread and extensively investigated in medical use. They have proven to be effective in the areas of drug transport and biosensing techniques for cancer treatment. This method has been revealed to improve drug transport and biosensing techniques, and therefore, carbon nanotubes have lately drawn attention in cancer treatment [199]. CNTs are round unified cylinders of graphene layers, exhibiting distinctive physical, mechanical, and chemical features that have recently appealed to great attention [200-205]. Some evidence indicates that IGF1 (Insulin-like growth factor-1) and HER2-specific monoclonal antibodies can be attached to single-wall carbon nanotubes, which is a considerable step to overcome breast cancer in the late stages [206]. The field of carbon nanomaterials is developing technology with promising applications in the biomedicine field, especially for detecting, recognizing, and treating breast cancer. Nevertheless, no carbon nanomaterial simultaneously presents the desirable traits for therapeutic Administration in humans, and further studies are needed [207]. A novel treatment of metastatic breast cancer is also given by combining two advanced methods, including selective near-infrared photothermal removal and carbon nanotubes loaded with immune checkpoint inhibitors [208]. More specifically, this treatment uses anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) checkpoint inhibition, resulting in unrestrained T-cell propagation [209].
Ceramic nanoparticles
Ceramic nanoparticles (CNs) are developing as drug delivery systems, primarily because of their small size (<50 nm) and physicochemical features. They include albumin, iron oxide, or silica [210,211]. Aluminum oxide (Al2O3) and titanium dioxide (TiO2) are the most frequently used NPs for the proposal of nanocarriers. These NPs are not susceptible to swelling or variations in porosity with pH. Because of their more unchanging, bioavailable, readily makeable, and acceptable proteins and polypeptide formulation, ceramics nanoparticles are a particular carrier for protein and peptide delivery systems [212-214]. Based on the examination of Mahidhara and K Kanwar, they showed that iron-saturated-bovine lactoferrin-loaded alginate-enclosed chitosan-calcium phosphate (ACSC) ceramic Nano Capsules (Fe-bLf-loaded ACSC NCs) has promising anti-breast cancer efficacy in both in vitro and in vivo studies. The NCs proved to be highly anti-tumorigenic since none of the mice injected with breast cancer cells developed tumors in the xenograft models [215].
Nanoparticle albumin-bound (nab) technology
Paclitaxel (nab-paclitaxel; Abraxane®) is Food and Drug Administration-approved nanoparticle albumin-bound (nab™) for curing metastatic breast cancer. The key feature of the drug is that the formulation has no conventional surfactants in water-based injections [216]. Albumin is a versatile biomaterial for the manufacturing of NPs [217]. The effectiveness of the albumin-based transfer exists in its capability to boost tumor targeting and accumulation. For example, improved tumor accumulation is due to the enhanced uptake passively facilitated by the improved permeability and retaining effect [218].
Human serum albumin (HSA) nanoparticles
Prepared human serum albumin (HSA) can be used as one of the versatile carrier structures for drug delivery [219]. HSA nanoparticles can bind to numerous drug molecules and have excellent stability during storing and in vivo applications. Also, the toxicity and antigenicity of these NPs are very low. Biodegradability, reproducibility, and manageable release are other significant features of HSA nanoparticles. In addition, due to the numerous drug binding sites on the albumin molecule, significant amounts of drugs can be loaded on the matrix of HSA nanoparticles [220,221].
Antibody monoclonal modified-nanoparticles
Monoclonal antibodies (mAbs) are produced by B cells and specifically target antigens [222]. Pertuzumab and Trastuzumab are the most favorable and Food and Drug Administration approved antibodies used in breast cancer treatment, and they target HER2/neu receptors and inhibit cell growth rate [223,224] (Table 3; Figure 8). HER2 is a receptor with tyrosine kinase activity and participates in the epidermal growth factor receptor family [225] (Figure 9). HER2 is highly expressed in around 20-30% of breast cancer tumors, and the outcome of that is a more aggressive ailment, augmented mortality, and higher recurrence rate [226]. NPs containing HSA show a promising approach for targeted drug delivery to tumor cells; consequently, the binding of HSA nanoparticles to the antibody trastuzumab takes advantage of the capability of HER2-positive cells to integrate elements binding to HER2, and it is an appropriate method for treatment [227]. Conjugation of antibodies with nanoparticles such as gold, metal, iron oxide with different sizes and shapes showed promising results in Bryan E. White and Molly K. White’s examination [228].
Table 3.
Monoclonal antibody especially for breast cancer
| Drug | Mechanism | Type | Therapy Group | Side effects | FDA Approval | REF. |
|---|---|---|---|---|---|---|
| Pertuzumab | HER2/neu | Humanized monoclonal antibody | Metastatic HER2-positive breast cancer | Risk of infection, Bruising and bleeding, Anemia, Diarrhea, Loss of appetite, Sore mouth | Yes | [244] |
| Trastuzumab | HER2/neu | Humanized monoclonal antibody | breast cancer and stomach cancer | flu-like symptoms (high temperature, chills, and mild pain), nausea, and diarrhea | Yes | [245] |
Figure 8.

Mechanism of trastuzumab.
Figure 9.

HER-2 receptors send signals to cells and are in charge of cell growth; thus, in breast cancer, it causes cells to grow and divide at an uncontrolled rate, leading to tumor growth. Arrows in the figure illustrate growth signals from HER2 receptors to the nucleus of cells.
Discussion
Breast cancer is one of the foremost causes of death among women; subsequently, immunologists worldwide are trying to treat this problem. One of the best cures for treating this malignancy is nanoparticles and monoclonal antibodies. Her2/neu is a receptor that is in charge of breast cells’ growth every single cell by sending signals to nuclear cells; however, in some tumoral cells, it overexpresses; thus, cells begin to grow uncontrollably. The advent method of utilizing nanoparticles to treat cancer has significantly changed the protocols for eliminating tumor cells and drug delivery. The most significant advantage of these nanoparticles is precise targeting in which the drug does not affect other healthy tissues and just attacks the malignant cells. Also, drugs carried in nanoparticles are more stable than other conventional drugs and are protected from degradation or environmental effect. Biocompatibility, or fewer side-effects, is another advantage of these nanosized particles used as drug transporters. This means that these materials are compatible with living tissue, and we experience fewer secondary adverse effects other than the therapeutic influence.
Due to the fact that breast cancer is one of the invasive types of cancers, especially among women, the classification of nanoparticle usages and their results can assist in remedying the treatment of this malignancy. Countless nanoparticles have been developed to eradicate breast cancer; however, no article clearly classified them. Therefore, this article has tried to gather all valuable data to this extent. The name of nanoparticles, their results, and their future function have been mentioned to assist physicians and scholars remedy this cancer.
All in all, these benefits have made nanoparticles one of the most valuable tools to cure cancer. Every nanoparticle has its exceptional characteristic, but Among NPs, due to their unique features and fewer disadvantages, polymeric NPs and compact lipid nanostructures like phospholipids, including liposomes and micelles, seem to be the best choice in drug transport to breast cancer cells. Liposomes can carry a wide variety of biologically active compounds because of their high firmness and stability that keep these compounds safe. With fewer side effects, greater-sized liposomes are also an excellent choice for drug delivery since they are removed faster through the reticuloendothelial system (RES). Polymeric nanoparticles, specifically PCL, PEG, nanospheres, and nanocapsules, tend to be ideal drug transporters to the targeted breast tumor cells. Again, outstanding stability is why these NPs are widely utilized in drug delivery systems. Various bioactive drugs make them sufficient drug transporters since they guard medications against biodegradation and enzyme effects. For battling these tumoral cells, some drugs can be used to target these receptors, and by blocking those signaling, tumoral cells begin to diminish. Compared with other monoclonal antibodies such as PD-1 inhibitors, Her2/neu Mabs give promising and better outcomes. The FDA-approved monoclonal antibody for this kind of treatment is Pertuzumab and Trastuzumab, which explicitly targets Her2/neu receptors. These two monoclonal antibodies are humanized; subsequently, the body does not battle against them, and it does not assume them as an unknown substance so that they can be used safely; however, they have some adverse effects; therefore, the physician should pay attention to patients’ overall stability for prescribing those for patients’ therapy. Some of those side effects are the risk of infection, bruising and bleeding, Anemia, Loss of appetite, Sore mouth, flu-like symptoms (such as fever, chills, and mild pain), nausea, and diarrhea. Besides, most Nps cannot be administrated in clinical stages, so exploring possible methods to make them clinically applicable is the main challenge for future research. For the first time, this article discussed the most novel method to treat breast cancer in the most effective way possible by gathering recent data from numerous nanoparticles and breast cancer immunotherapy articles.
Conclusion
Nanoparticles are designed to improve the qualities of conventional drugs, especially chemotherapeutic agents, in numerous aspects. These nanocarriers hinder the degradation of the drug, decrease toxicity and adverse side effects and control the sustained release of the medication. Breast cancer is also treated with these versatile nanocarriers, exclusively Polymeric NPs, in the most efficient way possible. Although abundant nanocarriers have been investigated preclinically, only a handful of nano carrier-based drugs such as Doxil (Caelyx) (Liposomal Doxorubicin (PEGylated)), Myocet (Liposomal doxorubicin (non-PEGylated)), Lipusu (Liposomal paclitaxel), and Abraxane (Albumin-particle bound paclitaxel) are approved by Food and Drug Administration (FDA) and available in the pharmaceutical market for treating breast cancer in different stages. However, most of these NPs are still in vivo and in vitro stage and do not have clinical use, so there are no clear clinical records for most of the NPs.This review illustrates that much work must be done to discover new methods to clinically utilize the NPs and manufacture more efficient and less toxic drugs. This article highlights the advantages and drawbacks of all available NPs and opens the gates for further investigations by reviewing the most prominent studies in this field.
Disclosure of conflict of interest
None.
Abbreviations
- NP
Nanoparticles
- ER
Estrogen receptor
- HER2
Human epidermal growth factor receptor 2
- PgR
Progesterone receptor
- MPs
Microparticles
- PLGA
Poly lactic-co-glycolic acid
- NNI
National Nanotechnology Initiative
- EPR
Enhanced permeability and retention
- PEG
Polyethylene glycol
- PCL
Poly-ε-caprolactone
- PCL-PEG
polycaprolactone-polyethylene glycol
- EGFR
Epidermal growth factor receptor
- TNBC
Triple-negative breast cancer
- DOX
Doxorubicin
- CSeSiO2 HNPs
pH-sensitive chitosan silica hollow nanospheres
- Erb2
Erythroblastic oncogene B
- AG
Amphiphilic gelatin
- CaP
Calcium phosphate
- PLA
Polylactic Acid
- PTX
Paclitaxel
- HCN
Hollow carbon nanospheres
- PAMAM
Polyamidoamine
- TZ
Trastuzumab
- DTX
Docetaxel
- NIR
Near-infrared
- TPGS
Tocopheryl polyethylene glycol succinate
- TOS
Tocopheryl succinate
- DOX·HCl
doxorubicin hydrochloride
- SLN
Solid lipid nanoparticles
- RES
Reticuloendothelial system
- PS
Photosensitizer
- CNT
Carbon nanotubes
- IGF1
Insulin-like growth factor-1
- CN
Ceramic nanoparticles
- Fe-bLf-loaded ACSC NCs
Iron-saturated-bovine lactoferrin-loaded alginate-enclosed chitosan-calcium phosphate ceramic Nano Capsules
- HSA
Human serum albumin
- mAbs
Monoclonal antibodies
- Dox/F127&P123-Tf
a transferrin (Tf)-conjugated polymeric nanoparticle composed of poloxamer 407 (F127) and 123(P123)
- DTX/Apt-pD
docetaxel/aptamer-polydopamine
References
- 1.Stanculeanu DL, Daniela Z, Lazescu A, Bunghez R, Anghel R. Development of new immunotherapy treatments in different cancer types. J Med Life. 2016;9:240–248. [PMC free article] [PubMed] [Google Scholar]
- 2.Kodach LL, Peppelenbosch MP. Targeting the myeloid-derived suppressor cell compartment for inducing responsiveness to immune checkpoint blockade is best limited to specific subtypes of gastric cancers. Gastroenterology. 2021;161:727. doi: 10.1053/j.gastro.2021.03.047. [DOI] [PubMed] [Google Scholar]
- 3.Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov. 2019;18:175–196. doi: 10.1038/s41573-018-0006-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ahmed S, Rai KR. Interferon in the treatment of hairy-cell leukemia. Best Pract Res Clin Haematol. 2003;16:69–81. doi: 10.1016/s1521-6926(02)00084-1. [DOI] [PubMed] [Google Scholar]
- 5.Rosenberg SA. IL-2: the first effective immunotherapy for human cancer. J Immunol. 2014;192:5451–5458. doi: 10.4049/jimmunol.1490019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Sambi M, Bagheri L, Szewczuk MR. Current challenges in cancer immunotherapy: multimodal approaches to improve efficacy and patient response rates. J Oncol. 2019;2019:4508794. doi: 10.1155/2019/4508794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ventola CL. Cancer immunotherapy, part 2: efficacy, safety, and other clinical considerations. P T. 2017;42:452–463. [PMC free article] [PubMed] [Google Scholar]
- 8.Weiner LM. Cancer immunology for the clinician. Clin Adv Hematol Oncol. 2015;13:299–306. [PubMed] [Google Scholar]
- 9.Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331:1565–1570. doi: 10.1126/science.1203486. [DOI] [PubMed] [Google Scholar]
- 10.Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74. doi: 10.1126/science.aaa4971. [DOI] [PubMed] [Google Scholar]
- 11.Shao K, Singha S, Clemente-Casares X, Tsai S, Yang Y, Santamaria P. Nanoparticle-based immunotherapy for cancer. ACS Nano. 2015;9:16–30. doi: 10.1021/nn5062029. [DOI] [PubMed] [Google Scholar]
- 12.Xiong Y, Wang Y, Tiruthani K. Tumor immune microenvironment and nano-immunotherapeutics in colorectal cancer. Nanomedicine. 2019;21:102034. doi: 10.1016/j.nano.2019.102034. [DOI] [PubMed] [Google Scholar]
- 13.Wang M, Thanou M. Targeting nanoparticles to cancer. Pharmacol Res. 2010;62:90–99. doi: 10.1016/j.phrs.2010.03.005. [DOI] [PubMed] [Google Scholar]
- 14.Li C. A targeted approach to cancer imaging and therapy. Nat Mater. 2014;13:110–115. doi: 10.1038/nmat3877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Getts DR, Terry RL, Getts MT, Deffrasnes C, Müller M, van Vreden C, Ashhurst TM, Chami B, McCarthy D, Wu H, Ma J, Martin A, Shae LD, Witting P, Kansas GS, Kühn J, Hafezi W, Campbell IL, Reilly D, Say J, Brown L, White MY, Cordwell SJ, Chadban SJ, Thorp EB, Bao S, Miller SD, King NJ. Therapeutic inflammatory monocyte modulation using immune-modifying microparticles. Sci Transl Med. 2014;6:219ra217. doi: 10.1126/scitranslmed.3007563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Gupta AS. Nanomedicine approaches in vascular disease: a review. Nanomedicine. 2011;7:763–779. doi: 10.1016/j.nano.2011.04.001. [DOI] [PubMed] [Google Scholar]
- 17.Gupta C, Prakash D, Gupta S. Cancer treatment with nano-diamonds. Front Biosci (Schol Ed) 2017;9:62–70. doi: 10.2741/s473. [DOI] [PubMed] [Google Scholar]
- 18.Yeo SK, Guan JL. Breast cancer: multiple subtypes within a tumor? Trends Cancer. 2017;3:753–760. doi: 10.1016/j.trecan.2017.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.DeSantis CE, Fedewa SA, Goding Sauer A, Kramer JL, Smith RA, Jemal A. Breast cancer statistics, 2015: convergence of incidence rates between black and white women. CA Cancer J Clin. 2016;66:31–42. doi: 10.3322/caac.21320. [DOI] [PubMed] [Google Scholar]
- 20.Sun YS, Zhao Z, Yang ZN, Xu F, Lu HJ, Zhu ZY, Shi W, Jiang J, Yao PP, Zhu HP. Risk factors and preventions of breast cancer. Int J Biol Sci. 2017;13:1387–1397. doi: 10.7150/ijbs.21635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Coughlin SS. Epidemiology of breast cancer in women. Adv Exp Med Biol. 2019;1152:9–29. doi: 10.1007/978-3-030-20301-6_2. [DOI] [PubMed] [Google Scholar]
- 22.Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lønning PE, Børresen-Dale AL, Brown PO, Botstein D. Molecular portraits of human breast tumours. Nature. 2000;406:747–752. doi: 10.1038/35021093. [DOI] [PubMed] [Google Scholar]
- 23.Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, Speed D, Lynch AG, Samarajiwa S, Yuan Y, Gräf S, Ha G, Haffari G, Bashashati A, Russell R, McKinney S, Langerød A, Green A, Provenzano E, Wishart G, Pinder S, Watson P, Markowetz F, Murphy L, Ellis I, Purushotham A, Børresen-Dale AL, Brenton JD, Tavaré S, Caldas C, Aparicio S. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346–352. doi: 10.1038/nature10983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70. doi: 10.1038/nature11412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Marusyk A, Almendro V, Polyak K. Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer. 2012;12:323–334. doi: 10.1038/nrc3261. [DOI] [PubMed] [Google Scholar]
- 26.Breast cancer [Internet] Address: https://www.who.int/news-room/fact-sheets/detail/breast-cancer. Date of citation: 8 January 2022.
- 27.Goodarzi EML, Sohrabivafa M, Adineh HA. Epidemiology incidence and mortality of breast cancer and its association with the body mass index and human development index in the Asian population. WCRJ. 2019 [Google Scholar]
- 28.2020 Cancer incidence and mortality in EU-27 countries-EU Science Hub-European Commission; [Google Scholar]
- 29.Centers for disease control and prevention. 2022. What are the risk factors for breast cancer? [Internet] [cited 28 January 2022] [Google Scholar]
- 30.Centers for disease control and prevention. 2022. What are the symptoms of breast cancer? [Internet] [cited 28 January 2022] [Google Scholar]
- 31.Bloom JR, Stewart SL, Johnston M, Banks P, Fobair P. Sources of support and the physical and mental well-being of young women with breast cancer. Soc Sci Med. 2001;53:1513–1524. doi: 10.1016/s0277-9536(00)00440-8. [DOI] [PubMed] [Google Scholar]
- 32.Arjaans M, Schröder CP, Oosting SF, Dafni U, Kleibeuker JE, de Vries EG. VEGF pathway targeting agents, vessel normalization and tumor drug uptake: from bench to bedside. Oncotarget. 2016;7:21247–21258. doi: 10.18632/oncotarget.6918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Cho SY, Park SY, Bae YK, Kim JY, Kim EK, Kim WG, Kwon Y, Lee A, Lee HJ, Lee JS, Park JY, Gong G, Yoon HK. Standardized pathology report for breast cancer. J Pathol Transl Med. 2021;55:1–15. doi: 10.4132/jptm.2020.11.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Voduc KD, Cheang MC, Tyldesley S, Gelmon K, Nielsen TO, Kennecke H. Breast cancer subtypes and the risk of local and regional relapse. J. Clin. Oncol. 2010;28:1684–1691. doi: 10.1200/JCO.2009.24.9284. [DOI] [PubMed] [Google Scholar]
- 35.Tsang JYS, Tse GM. Molecular classification of breast cancer. Adv Anat Pathol. 2020;27:27–35. doi: 10.1097/PAP.0000000000000232. [DOI] [PubMed] [Google Scholar]
- 36.Vuong D, Simpson PT, Green B, Cummings MC, Lakhani SR. Molecular classification of breast cancer. Virchows Archiv. 2014;465:1–14. doi: 10.1007/s00428-014-1593-7. [DOI] [PubMed] [Google Scholar]
- 37.Fragomeni SM, Sciallis A, Jeruss JS. Molecular subtypes and local-regional control of breast cancer. Surg Oncol Clin N Am. 2018;27:95–120. doi: 10.1016/j.soc.2017.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sørlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS, Thorsen T, Quist H, Matese JC, Brown PO, Botstein D, Lønning PE, Børresen-Dale AL. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A. 2001;98:10869–10874. doi: 10.1073/pnas.191367098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Gao JJ, Swain SM. Luminal A breast cancer and molecular assays: a review. Oncologist. 2018;23:556–565. doi: 10.1634/theoncologist.2017-0535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Li ZH, Hu PH, Tu JH, Yu NS. Luminal B breast cancer: patterns of recurrence and clinical outcome. Oncotarget. 2016;7:65024–65033. doi: 10.18632/oncotarget.11344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Harbeck N, Thomssen C, Gnant M. St. Gallen 2013: brief preliminary summary of the consensus discussion. Breast Care (Basel) 2013;8:102–109. doi: 10.1159/000351193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Prat A, Pascual T, De Angelis C, Gutierrez C, Llombart-Cussac A, Wang T, Cortés J, Rexer B, Paré L, Forero A, Wolff AC, Morales S, Adamo B, Brasó-Maristany F, Vidal M, Veeraraghavan J, Krop I, Galván P, Pavlick AC, Bermejo B, Izquierdo M, Rodrik-Outmezguine V, Reis-Filho JS, Hilsenbeck SG, Oliveira M, Dieci MV, Griguolo G, Fasani R, Nuciforo P, Parker JS, Conte P, Schiff R, Guarneri V, Osborne CK, Rimawi MF. HER2-enriched subtype and ERBB2 expression in HER2-positive breast cancer treated with dual HER2 blockade. J Natl Cancer Inst. 2019;112:46–54. doi: 10.1093/jnci/djz042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Toft DJ, Cryns VL. Minireview: basal-like breast cancer: from molecular profiles to targeted therapies. Mol Endocrinol. 2011;25:199–211. doi: 10.1210/me.2010-0164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Smith NG, Gyanchandani R, Shah OS, Gurda GT, Lucas PC, Hartmaier RJ, Brufsky AM, Puhalla S, Bahreini A, Kota K, Wald AI, Nikiforov YE, Nikiforova MN, Oesterreich S, Lee AV. Targeted mutation detection in breast cancer using MammaSeq™. Breast Cancer Res. 2019;21:22. doi: 10.1186/s13058-019-1102-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Siegel MB, He X, Hoadley KA, Hoyle A, Pearce JB, Garrett AL, Kumar S, Moylan VJ, Brady CM, Van Swearingen AE, Marron D, Gupta GP, Thorne LB, Kieran N, Livasy C, Mardis ER, Parker JS, Chen M, Anders CK, Carey LA, Perou CM. Integrated RNA and DNA sequencing reveals early drivers of metastatic breast cancer. J Clin Invest. 2018;128:1371–1383. doi: 10.1172/JCI96153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Harrell JC, Prat A, Parker JS, Fan C, He X, Carey L, Anders C, Ewend M, Perou CM. Genomic analysis identifies unique signatures predictive of brain, lung, and liver relapse. Breast Cancer Res Treat. 2012;132:523–535. doi: 10.1007/s10549-011-1619-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Lin NU, Claus E, Sohl J, Razzak AR, Arnaout A, Winer EP. Sites of distant recurrence and clinical outcomes in patients with metastatic triple-negative breast cancer: high incidence of central nervous system metastases. Cancer. 2008;113:2638–2645. doi: 10.1002/cncr.23930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.McCabe MJ, Gauthier MA, Chan CL, Thompson TJ, De Sousa SMC, Puttick C, Grady JP, Gayevskiy V, Tao J, Ying K, Cipponi A, Deng N, Swarbrick A, Thomas ML, Lord RV, Johns AL, Kohonen-Corish M, O’Toole SA, Clark J, Mueller SA, Gupta R, McCormack AI, Dinger ME, Cowley MJ kConFab. Development and validation of a targeted gene sequencing panel for application to disparate cancers. Sci Rep. 2019;9:17052. doi: 10.1038/s41598-019-52000-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bewicke-Copley F, Arjun Kumar E, Palladino G, Korfi K, Wang J. Applications and analysis of targeted genomic sequencing in cancer studies. Comput Struct Biotechnol J. 2019;17:1348–1359. doi: 10.1016/j.csbj.2019.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Langer R, Folkman J. Polymers for the sustained release of proteins and other macromolecules. Nature. 1976;263:797–800. doi: 10.1038/263797a0. [DOI] [PubMed] [Google Scholar]
- 51.Putney SD, Burke PA. Improving protein therapeutics with sustained-release formulations. Nat Biotechnol. 1998;16:153–157. doi: 10.1038/nbt0298-153. [DOI] [PubMed] [Google Scholar]
- 52.Padalkar A, Shahi S, Thube patil M. Micro particles: an approach for betterment of drug delivery system. International Journal of Pharma Research and Development-Online. 2011;3 [Google Scholar]
- 53.Beck P, Kreuter J, Reszka R, Fichtner I. Influence of polybutylcyanoacrylate nanoparticles and liposomes on the efficacy and toxicity of the anticancer drug mitoxantrone in murine tumour models. J Microencapsul. 1993;10:101–114. doi: 10.3109/02652049309015316. [DOI] [PubMed] [Google Scholar]
- 54.Kreuter J. Nanoparticles and microparticles for drug and vaccine delivery. J Anat. 1996;189:503–505. [PMC free article] [PubMed] [Google Scholar]
- 55.Ravi Kumar MN. Nano and microparticles as controlled drug delivery devices. J Pharm Pharm Sci. 2000;3:234–258. [PubMed] [Google Scholar]
- 56.Carson WE, Dierksheide JE, Jabbour S, Anghelina M, Bouchard P, Ku G, Yu H, Baumann H, Shah MH, Cooper MA, Durbin J, Caligiuri MA. Coadministration of interleukin-18 and interleukin-12 induces a fatal inflammatory response in mice: critical role of natural killer cell interferon-gamma production and STAT-mediated signal transduction. Blood. 2000;96:1465–1473. [PubMed] [Google Scholar]
- 57.Bookstaver ML, Tsai SJ, Bromberg JS, Jewell CM. Improving vaccine and immunotherapy design using biomaterials. Trends Immunol. 2018;39:135–150. doi: 10.1016/j.it.2017.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Gregory AE, Titball R, Williamson D. Vaccine delivery using nanoparticles. Front Cell Infect Microbiol. 2013;3:13. doi: 10.3389/fcimb.2013.00013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Batty CJ, Tiet P, Bachelder EM, Ainslie KM. Drug delivery for cancer immunotherapy and Vaccines. Pharm Nanotechnol. 2018;6:232–244. doi: 10.2174/2211738506666180918122337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Suri SS, Fenniri H, Singh B. Nanotechnology-based drug delivery systems. J Occup Med Toxicol. 2007;2:16. doi: 10.1186/1745-6673-2-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wilczewska AZ, Niemirowicz K, Markiewicz KH, Car H. Nanoparticles as drug delivery systems. Pharmacol Rep. 2012;64:1020–1037. doi: 10.1016/s1734-1140(12)70901-5. [DOI] [PubMed] [Google Scholar]
- 62.Hainfeld JF, Lin L, Slatkin DN, Avraham Dilmanian F, Vadas TM, Smilowitz HM. Gold nanoparticle hyperthermia reduces radiotherapy dose. Nanomedicine. 2014;10:1609–1617. doi: 10.1016/j.nano.2014.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Roco MC. The long view of nanotechnology development: the National Nanotechnology Initiative at 10 years. J Nanopart Res. 2011;13:427–445. [Google Scholar]
- 64.Jeevanandam J, Barhoum A, Chan YS, Dufresne A, Danquah MK. Review on nanoparticles and nanostructured materials: history, sources, toxicity and regulations. Beilstein J Nanotechnol. 2018;9:1050–1074. doi: 10.3762/bjnano.9.98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Chen C, Li S, Liu K, Ma G, Yan X. Co-assembly of heparin and polypeptide hybrid nanoparticles for biomimetic delivery and anti-thrombus therapy. Small. 2016;12:4719–4725. doi: 10.1002/smll.201600328. [DOI] [PubMed] [Google Scholar]
- 66.Duan X, Chan C, Lin W. Nanoparticle-mediated immunogenic cell death enables and potentiates cancer immunotherapy. Angew Chem Int Ed Engl. 2019;58:670–680. doi: 10.1002/anie.201804882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Yu Z, Gao L, Chen K, Zhang W, Zhang Q, Li Q, Hu K. Nanoparticles: a new approach to upgrade cancer diagnosis and treatment. Nanoscale Research Letters. 2021;16:88. doi: 10.1186/s11671-021-03489-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wang AZ, Langer R, Farokhzad OC. Nanoparticle delivery of cancer drugs. Annu Rev Med. 2012;63:185–198. doi: 10.1146/annurev-med-040210-162544. [DOI] [PubMed] [Google Scholar]
- 69.Müller RH, Gohla S, Keck CM. State of the art of nanocrystals--special features, production, nanotoxicology aspects and intracellular delivery. Eur J Pharm Biopharm. 2011;78:1–9. doi: 10.1016/j.ejpb.2011.01.007. [DOI] [PubMed] [Google Scholar]
- 70.Gigliobianco MR, Casadidio C, Censi R, Di Martino P. Nanocrystals of poorly soluble drugs: drug bioavailability and physicochemical stability. Pharmaceutics. 2018;10:134. doi: 10.3390/pharmaceutics10030134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Chen W, Yang W, Chen P, Huang Y, Li F. Disulfiram copper nanoparticles prepared with a stabilized metal ion ligand complex method for treating drug-resistant prostate cancers. ACS Appl Mater Interfaces. 2018;10:41118–41128. doi: 10.1021/acsami.8b14940. [DOI] [PubMed] [Google Scholar]
- 72.Li D, Zhang M, Xu F, Chen Y, Chen B, Chang Y, Zhong H, Jin H, Huang Y. Biomimetic albumin-modified gold nanorods for photothermo-chemotherapy and macrophage polarization modulation. Acta Pharm Sin B. 2018;8:74–84. doi: 10.1016/j.apsb.2017.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Chang Y, Jiang J, Chen W, Yang W, Chen L, Chen P, Shen J, Qian S, Zhou T, Wu L, Hong L, Huang Y, Li F. Biomimetic metal-organic nanoparticles prepared with a 3D-printed microfluidic device as a novel formulation for disulfiram-based therapy against breast cancer. Appl Mater Today. 2020;18:100492. doi: 10.1016/j.apmt.2019.100492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Soppimath KS, Aminabhavi TM, Kulkarni AR, Rudzinski WE. Biodegradable polymeric nanoparticles as drug delivery devices. J Control Release. 2001;70:1–20. doi: 10.1016/s0168-3659(00)00339-4. [DOI] [PubMed] [Google Scholar]
- 75.Cano A, Ettcheto M, Chang JH, Barroso E, Espina M, Kühne BA, Barenys M, Auladell C, Folch J, Souto EB, Camins A, Turowski P, García ML. Dual-drug loaded nanoparticles of Epigallocatechin-3-gallate (EGCG)/Ascorbic acid enhance therapeutic efficacy of EGCG in a APPswe/PS1dE9 Alzheimer’s disease mice model. J Control Release. 2019;301:62–75. doi: 10.1016/j.jconrel.2019.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Cano A, Sánchez-López E, Ettcheto M, López-Machado A, Espina M, Souto EB, Galindo R, Camins A, García ML, Turowski P. Current advances in the development of novel polymeric nanoparticles for the treatment of neurodegenerative diseases. Nanomedicine (Lond) 2020;15:1239–1261. doi: 10.2217/nnm-2019-0443. [DOI] [PubMed] [Google Scholar]
- 77.Zielińska A, Carreiró F, Oliveira AM, Neves A, Pires B, Venkatesh DN, Durazzo A, Lucarini M, Eder P, Silva AM, Santini A, Souto EB. Polymeric nanoparticles: production, characterization, toxicology and ecotoxicology. Molecules. 2020;25:3731. doi: 10.3390/molecules25163731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Calzoni E, Cesaretti A, Polchi A, Di Michele A, Tancini B, Emiliani C. Biocompatible polymer nanoparticles for drug delivery applications in cancer and neurodegenerative disorder therapies. J Funct Biomater. 2019;10:4. doi: 10.3390/jfb10010004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kozlowski M. Polymeric Nanomaterials for Nanotherapeutics. Ed. C. Vasile, Elsevier, 1st Edition. 2018. Series Volume Editors: Cornelia Vasile, eBook ISBN: 9780128139332; Paperback ISBN: 9780128139325: Elsevier, Published Date: 2nd November 2018; 558 pages. In Micro and Nano Technologies Series. Polymers (Basel) 2019;11:1452. [Google Scholar]
- 80.Otto DP, de Villiers MM. Physicochemical principles of nanosized drug delivery systems. In: de Villiers MM, Aramwit P, Kwon GS, editors. Nanotechnology in drug delivery. New York, NY: Springer New York; 2009. pp. 3–33. [Google Scholar]
- 81.Rosenberg B. Fundamental studies with cisplatin. Cancer. 1985;55:2303–l6. doi: 10.1002/1097-0142(19850515)55:10<2303::aid-cncr2820551002>3.0.co;2-l. [DOI] [PubMed] [Google Scholar]
- 82.Eatemadi A, Darabi M, Afraidooni L, Zarghami N, Daraee H, Eskandari L, Mellatyar H, Akbarzadeh A. Comparison, synthesis and evaluation of anticancer drug-loaded polymeric nanoparticles on breast cancer cell lines. Artif Cells Nanomed Biotechnol. 2016;44:1008–1017. doi: 10.3109/21691401.2015.1008510. [DOI] [PubMed] [Google Scholar]
- 83.Bressler EM, Kim J, Shmueli RB, Mirando AC, Bazzazi H, Lee E, Popel AS, Pandey NB, Green JJ. Biomimetic peptide display from a polymeric nanoparticle surface for targeting and antitumor activity to human triple-negative breast cancer cells. J Biomed Mater Res A. 2018;106:1753–1764. doi: 10.1002/jbm.a.36360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Abou-El-Naga AM, Mutawa G, El-Sherbiny IM, Mousa SA. Activation of polymeric nanoparticle intracellular targeting overcomes chemodrug resistance in human primary patient breast cancer cells. Int J Nanomedicine. 2018;13:8153–8164. doi: 10.2147/IJN.S182184. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 85.Soe ZC, Kwon JB, Thapa RK, Ou W, Nguyen HT, Gautam M, Oh KT, Choi HG, Ku SK, Yong CS, Kim JO. Transferrin-conjugated polymeric nanoparticle for receptor-mediated delivery of doxorubicin in doxorubicin-resistant breast cancer cells. Pharmaceutics. 2019;11:63. doi: 10.3390/pharmaceutics11020063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Verhoef JJ, Anchordoquy TJ. Questioning the use of PEGylation for drug delivery. Drug Deliv Transl Res. 2013;3:499–503. doi: 10.1007/s13346-013-0176-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Locatelli E, Comes Franchini M. Biodegradable PLGA-b-PEG polymeric nanoparticles: synthesis, properties, and nanomedical applications as drug delivery system. J Nanopart Res. 2012;14:1–17. [Google Scholar]
- 88.Su YC, Burnouf PA, Chuang KH, Chen BM, Cheng TL, Roffler SR. Conditional internalization of PEGylated nanomedicines by PEG engagers for triple negative breast cancer therapy. Nat Commun. 2017;8:15507. doi: 10.1038/ncomms15507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Inoue S, Patil R, Portilla-Arias J, Ding H, Konda B, Espinoza A, Mongayt D, Markman JL, Elramsisy A, Phillips HW. Nanobiopolymer for direct targeting and inhibition of EGFR expression in triple negative breast cancer. PLoS One. 2012;7:e31070. doi: 10.1371/journal.pone.0031070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Franchina T, Adamo B, Ricciardi GRR, Caristi N, Agostino RM, Proto C, Adamo V. Activity of pegylated liposomal doxorubicin in combination with gemcitabine in triple negative breast cancer with skin involvement: two case reports. Cancer Biol Ther. 2012;13:472–476. doi: 10.4161/cbt.19593. [DOI] [PubMed] [Google Scholar]
- 91.Woodruff MA, Hutmacher DW. The return of a forgotten polymer-polycaprolactone in the 21st century. Prog Polym Sci. 2010;35:1217–1256. [Google Scholar]
- 92.Dash TK, Konkimalla VB. Polymeric Modification and its implication in drug delivery: poly-ε-caprolactone (PCL) as a model polymer. Mol Pharm. 2012;9:2365–2379. doi: 10.1021/mp3001952. [DOI] [PubMed] [Google Scholar]
- 93.Merkli A, Tabatabay C, Gurny R, Heller J. Biodegradable polymers for the controlled release of ocular drugs. Prog Polym Sci. 1998;23:563–580. [Google Scholar]
- 94.Zhang L, Chen Z, Wang H, Wu S, Zhao K, Sun H, Kong D, Wang C, Leng X, Zhu D. Preparation and evaluation of PCL-PEG-PCL polymeric nanoparticles for doxorubicin delivery against breast cancer. RSC Adv. 2016;6:54727–54737. [Google Scholar]
- 95.Kong N, Deng M, Sun XN, Chen YD, Sui XB. Polydopamine-functionalized CA-(PCL-ran-PLA) nanoparticles for target delivery of docetaxel and chemo-photothermal therapy of breast cancer. Front Pharmacol. 2018;9:125. doi: 10.3389/fphar.2018.00125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Cuong NV, Hsieh MF, Chen YT, Liau I. Synthesis and characterization of PEG-PCL-PEG triblock copolymers as carriers of doxorubicin for the treatment of breast cancer. J Appl Polym Sci. 2010;117:3694–3703. [Google Scholar]
- 97.Nosrati H, Adinehvand R, Manjili HK, Rostamizadeh K, Danafar H. Synthesis, characterization, and kinetic release study of methotrexate loaded mPEG-PCL polymersomes for inhibition of MCF-7 breast cancer cell line. Pharm Dev Technol. 2019;24:89–98. doi: 10.1080/10837450.2018.1425433. [DOI] [PubMed] [Google Scholar]
- 98.Mohammed MA, Syeda JTM, Wasan KM, Wasan EK. An overview of chitosan nanoparticles and its application in non-parenteral drug delivery. Pharmaceutics. 2017;9:53. doi: 10.3390/pharmaceutics9040053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Nagpal K, Singh SK, Mishra DN. Chitosan nanoparticles: a promising system in novel drug delivery. Chem Pharm Bull (Tokyo) 2010;58:1423–1430. doi: 10.1248/cpb.58.1423. [DOI] [PubMed] [Google Scholar]
- 100.Nimesh S. 9-Chitosan nanoparticles. In: Nimesh S, editor. Gene therapy. Woodhead Publishing; 2013. pp. 163–196. [Google Scholar]
- 101.Shu XZ, Zhu KJ. The influence of multivalent phosphate structure on the properties of ionically cross-linked chitosan films for controlled drug release. Eur J Pharm Biopharm. 2002;54:235–243. doi: 10.1016/s0939-6411(02)00052-8. [DOI] [PubMed] [Google Scholar]
- 102.Niu S, Williams GR, Wu J, Wu J, Zhang X, Zheng H, Li S, Zhu LM. A novel chitosan-based nanomedicine for multi-drug resistant breast cancer therapy. Chemical Engineering Journal. 2019;369:134–149. [Google Scholar]
- 103.E A K N, S B, Martin CA, J RR, A S, V N, B S L, Frank-Kamenetskaya OV, Radhakrishnan S, S NK. A competent bidrug loaded water soluble chitosan derivative for the effective inhibition of breast cancer. Sci Rep. 2020;10:3991. doi: 10.1038/s41598-020-60888-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Yang Q, Wang S, Fan P, Wang L, Di Y, Lin K, Xiao FS. pH-responsive carrier system based on carboxylic acid modified mesoporous silica and polyelectrolyte for drug delivery. Chemistry of Materials. 2005;17:5999–6003. [Google Scholar]
- 105.Deng Z, Zhen Z, Hu X, Wu S, Xu Z, Chu PK. Hollow chitosan-silica nanospheres as pH-sensitive targeted delivery carriers in breast cancer therapy. Biomaterials. 2011;32:4976–4986. doi: 10.1016/j.biomaterials.2011.03.050. [DOI] [PubMed] [Google Scholar]
- 106.Nam KS, Shon YH. Suppression of metastasis of human breast cancer cells by chitosan oligosaccharides. J Microbiol Biotechnol. 2009;19:629–633. doi: 10.4014/jmb.0811.603. [DOI] [PubMed] [Google Scholar]
- 107.Santos-Carballal B, Aaldering LJ, Ritzefeld M, Pereira S, Sewald N, Moerschbacher BM, Götte M, Goycoolea FM. Physicochemical and biological characterization of chitosan-microRNA nanocomplexes for gene delivery to MCF-7 breast cancer cells. Sci Rep. 2015;5:13567. doi: 10.1038/srep13567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Yasmin R, Shah M, Khan SA, Ali R. Gelatin nanoparticles: a potential candidate for medical applications. Nanotechnology Reviews. 2017;6:191–207. [Google Scholar]
- 109.Elzoghby AO. Gelatin-based nanoparticles as drug and gene delivery systems: reviewing three decades of research. J Control Release. 2013;172:1075–1091. doi: 10.1016/j.jconrel.2013.09.019. [DOI] [PubMed] [Google Scholar]
- 110.Azimi B, Nourpanah P, Rabiee M, Arbab S. Producing gelatin nanoparticles as delivery system for bovine serum albumin. Iran Biomed J. 2014;18:34–40. doi: 10.6091/ibj.1242.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Li WM, Chiang CS, Huang WC, Su CW, Chiang MY, Chen JY, Chen SY. Amifostine-conjugated pH-sensitive calcium phosphate-covered magnetic-amphiphilic gelatin nanoparticles for controlled intracellular dual drug release for dual-targeting in HER-2-overexpressing breast cancer. J Control Release. 2015;220:107–118. doi: 10.1016/j.jconrel.2015.10.020. [DOI] [PubMed] [Google Scholar]
- 112.Konecny GE, Pegram MD, Venkatesan N, Finn R, Yang G, Rahmeh M, Untch M, Rusnak DW, Spehar G, Mullin RJ, Keith BR, Gilmer TM, Berger M, Podratz KC, Slamon DJ. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res. 2006;66:1630–1639. doi: 10.1158/0008-5472.CAN-05-1182. [DOI] [PubMed] [Google Scholar]
- 113.Sadat Tabatabaei Mirakabad F, Nejati-Koshki K, Akbarzadeh A, Yamchi MR, Milani M, Zarghami N, Zeighamian V, Rahimzadeh A, Alimohammadi S, Hanifehpour Y, Joo SW. PLGA-based nanoparticles as cancer drug delivery systems. Asian Pac J Cancer Prev. 2014;15:517–535. doi: 10.7314/apjcp.2014.15.2.517. [DOI] [PubMed] [Google Scholar]
- 114.Kumari A, Yadav SK, Yadav SC. Biodegradable polymeric nanoparticles based drug delivery systems. Colloids Surf B Biointerfaces. 2010;75:1–18. doi: 10.1016/j.colsurfb.2009.09.001. [DOI] [PubMed] [Google Scholar]
- 115.Bowerman CJ, Byrne JD, Chu KS, Schorzman AN, Keeler AW, Sherwood CA, Perry JL, Luft JC, Darr DB, Deal AM, Napier ME, Zamboni WC, Sharpless NE, Perou CM, DeSimone JM. Docetaxel-loaded PLGA nanoparticles improve efficacy in taxane-resistant triple-negative breast cancer. Nano Lett. 2017;17:242–248. doi: 10.1021/acs.nanolett.6b03971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Akbari E, Mousazadeh H, Sabet Z, Fattahi T, Dehnad A, Akbarzadeh A, Alizadeh E. Dual drug delivery of trapoxin A and methotrexate from biocompatible PLGA-PEG polymeric nanoparticles enhanced antitumor activity in breast cancer cell line. J Drug Deliv Sci Technol. 2021;61:102294. [Google Scholar]
- 117.Amirsaadat S, Jafari-Gharabaghlou D, Alijani S, Mousazadeh H, Dadashpour M, Zarghami N. Metformin and Silibinin co-loaded PLGA-PEG nanoparticles for effective combination therapy against human breast cancer cells. J Drug Deliv Sci Technol. 2021;61:102107. [Google Scholar]
- 118.Jusu SM, Obayemi JD, Salifu AA, Nwazojie CC, Uzonwanne V, Odusanya OS, Soboyejo WO. Drug-encapsulated blend of PLGA-PEG microspheres: in vitro and in vivo study of the effects of localized/targeted drug delivery on the treatment of triple-negative breast cancer. Sci Rep. 2020;10:14188. doi: 10.1038/s41598-020-71129-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Anari E, Akbarzadeh A, Zarghami N. Chrysin-loaded PLGA-PEG nanoparticles designed for enhanced effect on the breast cancer cell line. Artif Cells Nanomed Biotechnol. 2016;44:1410–1416. doi: 10.3109/21691401.2015.1029633. [DOI] [PubMed] [Google Scholar]
- 120.Cao D, Zhang X, Akabar MD, Luo Y, Wu H, Ke X, Ci T. Liposomal doxorubicin loaded PLGA-PEG-PLGA based thermogel for sustained local drug delivery for the treatment of breast cancer. Artif Cells Nanomed Biotechnol. 2019;47:181–191. doi: 10.1080/21691401.2018.1548470. [DOI] [PubMed] [Google Scholar]
- 121.Tabatabaei Mirakabad FS, Akbarzadeh A, Milani M, Zarghami N, Taheri-Anganeh M, Zeighamian V, Badrzadeh F, Rahmati-Yamchi M. A Comparison between the cytotoxic effects of pure curcumin and curcumin-loaded PLGA-PEG nanoparticles on the MCF-7 human breast cancer cell line. Artif Cells Nanomed Biotechnol. 2016;44:423–430. doi: 10.3109/21691401.2014.955108. [DOI] [PubMed] [Google Scholar]
- 122.da Luz CM, Boyles MSP, Falagan-Lotsch P, Pereira MR, Tutumi HR, de Oliveira Santos E, Martins NB, Himly M, Sommer A, Foissner I, Duschl A, Granjeiro JM, Leite PE. Poly-lactic acid nanoparticles (PLA-NP) promote physiological modifications in lung epithelial cells and are internalized by clathrin-coated pits and lipid rafts. J Nanobiotechnology. 2017;15:11. doi: 10.1186/s12951-016-0238-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Lee S, Miyajima T, Sugawara-Narutaki A, Kato K, Nagata F. Development of paclitaxel-loaded poly(lactic acid)/hydroxyapatite core-shell nanoparticles as a stimuli-responsive drug delivery system. R Soc Open Sci. 2021;8:202030. doi: 10.1098/rsos.202030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Guterres SS, Alves MP, Pohlmann AR. Polymeric nanoparticles, nanospheres and nanocapsules, for cutaneous applications. Drug Target Insights. 2007;2:147–57. [PMC free article] [PubMed] [Google Scholar]
- 125.Awesh K. Yadav UGaRS. Nano drug delivery strategies for the treatment of cancers. 2020 [Google Scholar]
- 126.Zhang L, Wang P, Zheng W, Jiang X. Hollow carbon nanospheres for targeted delivery of chemotherapeutics in breast cancer therapy. J Mater Chem B. 2017;5:6601–6607. doi: 10.1039/c7tb01105a. [DOI] [PubMed] [Google Scholar]
- 127.Sharifi M, Hasan A, Nanakali NMQ, Salihi A, Qadir FA, Muhammad HA, Shekha MS, Aziz FM, Amen KM, Najafi F, Yousefi-Manesh H, Falahati M. Combined chemo-magnetic field-photothermal breast cancer therapy based on porous magnetite nanospheres. Sci Rep. 2020;10:5925. doi: 10.1038/s41598-020-62429-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Deng S, Gigliobianco MR, Censi R, Di Martino P. Polymeric nanocapsules as nanotechnological alternative for drug delivery system: current status, challenges and opportunities. Nanomaterials (Basel) 2020;10:847. doi: 10.3390/nano10050847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Meiera W. Polymer nanocapsules. Chem Soc Rev. 2000 [Google Scholar]
- 130.Quintanar-Guerrero D, Allémann E, Doelker E, Fessi H. Preparation and characterization of nanocapsules from preformed polymers by a new process based on emulsification-diffusion technique. Pharm Res. 1998;15:1056–1062. doi: 10.1023/a:1011934328471. [DOI] [PubMed] [Google Scholar]
- 131.Verma G, Rajagopalan MD, Valluru R, Sridhar KA. Chapter 7-nanoparticles: a novel approach to target tumors. In: Grumezescu AM, editor. Nano- and microscale drug delivery systems. Elsevier; 2017. pp. 113–129. [Google Scholar]
- 132.Sánchez-Moreno P, Ortega-Vinuesa JL, Martín-Rodríguez A, Boulaiz H, Marchal-Corrales JA, Peula-García JM. Characterization of different functionalized lipidic nanocapsules as potential drug carriers. Int J Mol Sci. 2012;13:2405–2424. doi: 10.3390/ijms13022405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Zafar S, Akhter S, Ahmad I, Hafeez Z, Alam Rizvi MM, Jain GK, Ahmad FJ. Improved chemotherapeutic efficacy against resistant human breast cancer cells with co-delivery of docetaxel and thymoquinone by chitosan grafted lipid nanocapsules: formulation optimization, in vitro and in vivo studies. Colloids Surf B Biointerfaces. 2020;186:110603. doi: 10.1016/j.colsurfb.2019.110603. [DOI] [PubMed] [Google Scholar]
- 134.Caminade AM, Turrin CO, Laurent R, Ouali A, Delavaux-Nicot B, editors. Dendrimers: towards catalytic, material and biomedical uses. 2011. [Google Scholar]
- 135.Caminade AM, Turrin CO. Dendrimers for drug delivery. J Mater Chem B. 2014;2:4055–4066. doi: 10.1039/c4tb00171k. [DOI] [PubMed] [Google Scholar]
- 136.Nanjwade BK, Bechra HM, Derkar GK, Manvi FV, Nanjwade VK. Dendrimers: emerging polymers for drug-delivery systems. Eur J Pharm Sci. 2009;38:185–196. doi: 10.1016/j.ejps.2009.07.008. [DOI] [PubMed] [Google Scholar]
- 137.Abbasi E, Aval SF, Akbarzadeh A, Milani M, Nasrabadi HT, Joo SW, Hanifehpour Y, Nejati-Koshki K, Pashaei-Asl R. Dendrimers: synthesis, applications, and properties. Nanoscale Res Lett. 2014;9:247. doi: 10.1186/1556-276X-9-247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Uram Ł, Misiorek M, Pichla M, Filipowicz-Rachwał A, Markowicz J, Wołowiec S, Wałajtys-Rode E. The effect of biotinylated PAMAM G3 dendrimers conjugated with COX-2 inhibitor (celecoxib) and PPARγ agonist (Fmoc-L-leucine) on human normal fibroblasts, immortalized keratinocytes and glioma cells in vitro. Molecules (Basel, Switzerland) 2019;24:3801. doi: 10.3390/molecules24203801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Kulhari H, Pooja D, Shrivastava S, Kuncha M, Naidu VGM, Bansal V, Sistla R, Adams DJ. Trastuzumab-grafted PAMAM dendrimers for the selective delivery of anticancer drugs to HER2-positive breast cancer. Sci Rep. 2016;6:23179. doi: 10.1038/srep23179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Figueroa-Magalhães MC, Jelovac D, Connolly RM, Wolff AC. Treatment of HER2-positive breast cancer. Breast. 2014;23:128–136. doi: 10.1016/j.breast.2013.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Pinto AC, Ades F, de Azambuja E, Piccart-Gebhart M. Trastuzumab for patients with HER2 positive breast cancer: delivery, duration and combination therapies. Breast. 2013;22(Suppl 2):S152–S155. doi: 10.1016/j.breast.2013.07.029. [DOI] [PubMed] [Google Scholar]
- 142.Zhang Y, Huang Y, Li S. Polymeric micelles: nanocarriers for cancer-targeted drug delivery. AAPS PharmSciTech. 2014;15:862–871. doi: 10.1208/s12249-014-0113-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Croy SR, Kwon GS. Polymeric micelles for drug deliv ery. Curr Pharm Des. 2006;12:4669–4684. doi: 10.2174/138161206779026245. [DOI] [PubMed] [Google Scholar]
- 144.Subramani K, Ahmed W. Emerging nanotechnologies in dentistry. Elsevier; 2012. Nanoparticulate drug delivery systems for oral cancer treatment; pp. 333–345. [Google Scholar]
- 145.Torchilin VP. Cell penetrating peptide-modified pharmaceutical nanocarriers for intracellular drug and gene delivery. Biopolymers. 2008;90:604–610. doi: 10.1002/bip.20989. [DOI] [PubMed] [Google Scholar]
- 146.Kedar U, Phutane P, Shidhaye S, Kadam V. Advances in polymeric micelles for drug delivery and tumor targeting. Nanomedicine. 2010;6:714–729. doi: 10.1016/j.nano.2010.05.005. [DOI] [PubMed] [Google Scholar]
- 147.Gong J, Chen M, Zheng Y, Wang S, Wang Y. Polymeric micelles drug delivery system in oncology. J Control Release. 2012;159:312–323. doi: 10.1016/j.jconrel.2011.12.012. [DOI] [PubMed] [Google Scholar]
- 148.Pais-Silva C, de Melo-Diogo D, Correia IJ. IR780-loaded TPGS-TOS micelles for breast cancer photodynamic therapy. Eur J Pharm Biopharm. 2017;113:108–117. doi: 10.1016/j.ejpb.2017.01.002. [DOI] [PubMed] [Google Scholar]
- 149.Discher BM, Bermudez H, Hammer DA, Discher DE, Won YY, Bates FS. Cross-linked polymersome membranes: vesicles with broadly adjustable properties. J Phys Chem B. 2002;106:2848–2854. [Google Scholar]
- 150.Xu J, Zhao Q, Jin Y, Qiu L. High loading of hydrophilic/hydrophobic doxorubicin into polyphosphazene polymersome for breast cancer therapy. Nanomedicine. 2014;10:349–358. doi: 10.1016/j.nano.2013.08.004. [DOI] [PubMed] [Google Scholar]
- 151.Ezzati Nazhad Dolatabadi J, Valizadeh H, Hamishehkar H. Solid lipid nanoparticles as efficient drug and gene delivery systems: recent breakthroughs. Adv Pharm Bull. 2015;5:151–159. doi: 10.15171/apb.2015.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Lingayat VJ, Zarekar NS, Shendge RS. Solid lipid nanoparticles: a review. Nanoscience and Nanotechnology Research. 2017;4:67–72. [Google Scholar]
- 153.Kang KW, Chun MK, Kim O, Subedi RK, Ahn SG, Yoon JH, Choi HK. Doxorubicin-loaded solid lipid nanoparticles to overcome multidrug resistance in cancer therapy. Nanomedicine. 2010;6:210–213. doi: 10.1016/j.nano.2009.12.006. [DOI] [PubMed] [Google Scholar]
- 154.Chawla JS, Amiji MM. Biodegradable poly(epsilon-caprolactone) nanoparticles for tumor-targeted delivery of tamoxifen. Int J Pharm. 2002;249:127–138. doi: 10.1016/s0378-5173(02)00483-0. [DOI] [PubMed] [Google Scholar]
- 155.Lu B, Xiong SB, Yang H, Yin XD, Chao RB. Solid lipid nanoparticles of mitoxantrone for local injection against breast cancer and its lymph node metastases. Eur J Pharm Sci. 2006;28:86–95. doi: 10.1016/j.ejps.2006.01.001. [DOI] [PubMed] [Google Scholar]
- 156.da Rocha MCO, da Silva PB, Radicchi MA, Andrade BYG, de Oliveira JV, Venus T, Merker C, Estrela-Lopis I, Longo JPF, Báo SN. Docetaxel-loaded solid lipid nanoparticles prevent tumor growth and lung metastasis of 4T1 murine mammary carcinoma cells. J Nanobiotechnology. 2020;18:43. doi: 10.1186/s12951-020-00604-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Torchilin VP. Multifunctional nanocarriers. Adv Drug Deliv Rev. 2006;58:1532–1555. doi: 10.1016/j.addr.2006.09.009. [DOI] [PubMed] [Google Scholar]
- 158.Malam Y, Loizidou M, Seifalian AM. Liposomes and nanoparticles: nanosized vehicles for drug delivery in cancer. Trends Pharmacol Sci. 2009;30:592–599. doi: 10.1016/j.tips.2009.08.004. [DOI] [PubMed] [Google Scholar]
- 159.Naseri N, Valizadeh H, Zakeri-Milani P. Solid lipid nanoparticles and nanostructured lipid carriers: structure, preparation and application. Adv Pharm Bull. 2015;5:305–313. doi: 10.15171/apb.2015.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Mukherjee B, Mondal L, Chakraborty S, Paul P, Choudhury A, Bhattacharya S, Hossain CM. Size dependent variations of phospholipid based vesicular drug carriers in systemic drug activity. Curr Pharm Biotechnol. 2015;16:380–391. doi: 10.2174/1389201016666150206105149. [DOI] [PubMed] [Google Scholar]
- 161.Zununi Vahed S, Salehi R, Davaran S, Sharifi S. Liposome-based drug co-delivery systems in cancer cells. Mater Sci Eng C Mater Biol Appl. 2017;71:1327–1341. doi: 10.1016/j.msec.2016.11.073. [DOI] [PubMed] [Google Scholar]
- 162.Bozzuto G, Molinari A. Liposomes as nanomedical devices. Int J Nanomedicine. 2015;10:975–999. doi: 10.2147/IJN.S68861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Bangham AD. Liposomes: the Babraham connection. Chem Phys Lipids. 1993;64:275–285. doi: 10.1016/0009-3084(93)90071-a. [DOI] [PubMed] [Google Scholar]
- 164.Yingchoncharoen P, Kalinowski DS, Richardson DR. Lipid-based drug delivery systems in cancer therapy: what is available and what is yet to come. Pharmacol Rev. 2016;68:701–787. doi: 10.1124/pr.115.012070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Paliwal SR, Paliwal R, Agrawal GP, Vyas SP. Liposomal nanomedicine for breast cancer therapy. Nanomedicine. 2011;6:1085–1100. doi: 10.2217/nnm.11.72. [DOI] [PubMed] [Google Scholar]
- 166.Lee JY, Shin DH, Kim JS. Anticancer effect of metformin in herceptin-conjugated liposome for breast cancer. Pharmaceutics. 2020;12:11. doi: 10.3390/pharmaceutics12010011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Maeda H. Macromolecular therapeutics in cancer treatment: the EPR effect and beyond. J Control Release. 2012;164:138–144. doi: 10.1016/j.jconrel.2012.04.038. [DOI] [PubMed] [Google Scholar]
- 168.Gkionis L, Campbell RA, Aojula H, Harris LK, Tirella A. Manufacturing drug co-loaded liposomal formulations targeting breast cancer: influence of preparative method on liposomes characteristics and in vitro toxicity. Int J Pharm. 2020;590:119926. doi: 10.1016/j.ijpharm.2020.119926. [DOI] [PubMed] [Google Scholar]
- 169.Szekeres M, Tóth IY, Illés E, Hajdú A, Zupkó I, Farkas K, Oszlánczi G, Tiszlavicz L, Tombácz E. Chemical and colloidal stability of carboxylated core-shell magnetite nanoparticles designed for biomedical applications. Int J Mol Sci. 2013;14:14550–14574. doi: 10.3390/ijms140714550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Colombo M, Carregal-Romero S, Casula MF, Gutiérrez L, Morales MP, Böhm IB, Heverhagen JT, Prosperi D, Parak WJ. Biological applications of magnetic nanoparticles. Chem Soc Rev. 2012;41:4306–4334. doi: 10.1039/c2cs15337h. [DOI] [PubMed] [Google Scholar]
- 171.Soenen SJ, Rivera-Gil P, Montenegro JM, Parak WJ, De Smedt SC, Braeckmans K. Cellular toxicity of inorganic nanoparticles: common aspects and guidelines for improved nanotoxicity evaluation. Nano Today. 2011;6:446–465. [Google Scholar]
- 172.Wang AZ, Langer R, Farokhzad OC. Nanoparticle delivery of cancer drugs. Annu Rev Med. 2012;63:185–198. doi: 10.1146/annurev-med-040210-162544. [DOI] [PubMed] [Google Scholar]
- 173.Cheng SH, Lo LW. Inorganic nanoparticles for enhanced photodynamic cancer therapy. Curr Drug Discov Technol. 2011;8:250–268. doi: 10.2174/157016311796798982. [DOI] [PubMed] [Google Scholar]
- 174.Lucky SS, Soo KC, Zhang Y. Nanoparticles in photodynamic therapy. Chem Rev. 2015;115:1990–2042. doi: 10.1021/cr5004198. [DOI] [PubMed] [Google Scholar]
- 175.Montaseri H, Kruger CA, Abrahamse H. Inorganic nanoparticles applied for active targeted photodynamic therapy of breast cancer. Pharmaceutics. 2021;13:296. doi: 10.3390/pharmaceutics13030296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Elizabeth PS, Néstor MM, David QG. Nanobiomaterials in clinical dentistry. Elsevier; 2019. Nanoparticles as dental drug-delivery systems; pp. 567–593. [Google Scholar]
- 177.Yaqoob AA, Ahmad H, Parveen T, Ahmad A, Oves M, Ismail IMI, Qari HA, Umar K, Mohamad Ibrahim MN. Recent advances in metal decorated nanomaterials and their various biological applications: a review. Front Chem. 2020;8:341. doi: 10.3389/fchem.2020.00341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Patra JK, Das G, Fraceto LF, Campos EVR, del Pilar Rodriguez-Torres M, Acosta-Torres LS, Diaz-Torres LA, Grillo R, Swamy MK, Sharma S, Habtemariam S, Shin HS. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnology. 2018;16:71. doi: 10.1186/s12951-018-0392-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Bhattacharyya S, Kudgus RA, Bhattacharya R, Mukherjee P. Inorganic nanoparticles in cancer therapy. Pharm Res. 2011;28:237–259. doi: 10.1007/s11095-010-0318-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Sau TK, Rogach AL, Jäckel F, Klar TA, Feldmann J. Properties and applications of colloidal nonspherical noble metal nanoparticles. Adv Mater. 2010;22:1805–1825. doi: 10.1002/adma.200902557. [DOI] [PubMed] [Google Scholar]
- 181.Sperling RA, Gil PR, Zhang F, Zanella M, Parak WJ. Biological applications of gold nanoparticles. Chem Soc Rev. 2008;37:1896–1908. doi: 10.1039/b712170a. [DOI] [PubMed] [Google Scholar]
- 182.Conde J, Doria G, Baptista P. Noble metal nanoparticles applications in cancer. J Drug Deliv. 2012;2012:751075. doi: 10.1155/2012/751075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Chen X, Li QW, Wang XM. 8-Gold nanostructures for bioimaging, drug delivery and therapeutics. In: Baltzer N, Copponnex T, editors. Precious metals for biomedical applications. Woodhead Publishing; 2014. pp. 163–176. [Google Scholar]
- 184.Han G, Ghosh P, Rotello VM. Multi-functional gold nanoparticles for drug delivery. In: Chan WCW, editor. Bio-applications of nanoparticles. New York, NY: Springer New York; 2007. pp. 48–56. [DOI] [PubMed] [Google Scholar]
- 185.Han G, Ghosh P, Rotello VM. Functionalized gold nanoparticles for drug delivery. Nanomedicine. 2007;2:113–123. doi: 10.2217/17435889.2.1.113. [DOI] [PubMed] [Google Scholar]
- 186.Lee J, Chatterjee DK, Lee MH, Krishnan S. Gold nanoparticles in breast cancer treatment: promise and potential pitfalls. Cancer Lett. 2014;347:46–53. doi: 10.1016/j.canlet.2014.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Sani A, Cao C, Cui D. Toxicity of gold nanoparticles (AuNPs): a review. Biochem Biophys Rep. 2021;26:100991. doi: 10.1016/j.bbrep.2021.100991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Libutti SK, Paciotti GF, Byrnes AA, Alexander HR Jr, Gannon WE, Walker M, Seidel GD, Yuldasheva N, Tamarkin L. Phase I and pharmacokinetic studies of CYT-6091, a novel PEGylated colloidal gold-rhTNF nanomedicine. Clin Cancer Res. 2010;16:6139–6149. doi: 10.1158/1078-0432.CCR-10-0978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Nanospectra Biosciences Inc. Efficacy study of aurolase therapy in subjects with primary and/or metastatic lung tumors. Philadelphia, PA, USA: U.S. National Library of Medicine; 2016. [Google Scholar]
- 190.Nanospectra Biosciences Inc. MRI/US fusion imaging and biopsy in combination with nanoparticle directed focal therapy for ablation of prostate tissue. Baltimore, MD, USA: U.S. National Library of Medicine; 2016. [Google Scholar]
- 191.Bromma K, Chithrani DB. Advances in gold nanoparticle-based combined cancer therapy. Nanomaterials (Basel) 2020;10:1671. doi: 10.3390/nano10091671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Zhang C, Zhang F, Han M, Wang X, Du J, Zhang H, Li W. Co-delivery of 5-fluorodeoxyuridine and doxorubicin via gold nanoparticle equipped with affibody-DNA hybrid strands for targeted synergistic chemotherapy of HER2 overexpressing breast cancer. Sci Rep. 2020;10:22015. doi: 10.1038/s41598-020-79125-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Gnanasekar S, Balakrishnan D, Seetharaman P, Arivalagan P, Chandrasekaran R, Sivaperumal S. Chrysin-anchored silver and gold nanoparticle-reduced graphene oxide composites for breast cancer therapy. ACS Applied Nano Materials. 2020;3:4574–4585. [Google Scholar]
- 194.Graf C, Vossen DLJ, Imhof A, van Blaaderen A. A general method to coat colloidal particles with silica. Langmuir. 2003;19:6693–6700. doi: 10.1021/la100188w. [DOI] [PubMed] [Google Scholar]
- 195.Hepokur C, Kariper İA, Mısır S, Ay E, Tunoğlu S, Ersez MS, Zeybek Ü, Kuruca SE, Yaylım İ. Silver nanoparticle/capecitabine for breast cancer cell treatment. Toxicol In Vitro. 2019;61:104600. doi: 10.1016/j.tiv.2019.104600. [DOI] [PubMed] [Google Scholar]
- 196.Swanner J, Fahrenholtz CD, Tenvooren I, Bernish BW, Sears JJ, Hooker A, Furdui CM, Alli E, Li W, Donati GL, Cook KL, Vidi PA, Singh R. Silver nanoparticles selectively treat triple-negative breast cancer cells without affecting non-malignant breast epithelial cells in vitro and in vivo. FASEB Bioadv. 2019;1:639–660. doi: 10.1096/fba.2019-00021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Bigall NC, Härtling T, Klose M, Simon P, Eng LM, Eychmüller A. Monodisperse platinum nanospheres with adjustable diameters from 10 to 100 nm: synthesis and distinct optical properties. Nano Lett. 2008;8:4588–4592. doi: 10.1021/nl802901t. [DOI] [PubMed] [Google Scholar]
- 198.Manzoor S, Bashir DJ, Imtiyaz K, Rizvi MMA, Ahamad I, Fatma T, Agarwal NB, Arora I, Samim M. Biofabricated platinum nanoparticles: therapeutic evaluation as a potential nanodrug against breast cancer cells and drug-resistant bacteria. RSC Adv. 2021;11:24900–24916. doi: 10.1039/d1ra03133c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Srinivasan C. Carbon nanotubes in cancer therapy. Current Science-Bangalore. 2008;94:300. [Google Scholar]
- 200.Yang ST, Guo W, Lin Y, Deng XY, Wang HF, Sun HF, Liu YF, Wang X, Wang W, Chen M, Huang YP, Sun YP. Biodistribution of pristine single-walled carbon nanotubes in vivo. The Journal of Physical Chemistry C. 2007;111:17761–17764. [Google Scholar]
- 201.Deng X, Yang S, Nie H, Wang H, Liu Y. A generally adoptable radiotracing method for tracking carbon nanotubes in animals. Nanotechnology. 2008;19:075101. doi: 10.1088/0957-4484/19/7/075101. [DOI] [PubMed] [Google Scholar]
- 202.Cherukuri P, Gannon CJ, Leeuw TK, Schmidt HK, Smalley RE, Curley SA, Weisman RB. Mammalian pharmacokinetics of carbon nanotubes using intrinsic near-infrared fluorescence. Proc Natl Acad Sci U S A. 2006;103:18882–18886. doi: 10.1073/pnas.0609265103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Liu Z, Winters M, Holodniy M, Dai H. siRNA delivery into human T cells and primary cells with carbon-nanotube transporters. Angew Chem Int Ed Engl. 2007;46:2023–2027. doi: 10.1002/anie.200604295. [DOI] [PubMed] [Google Scholar]
- 204.Wu W, Wieckowski S, Pastorin G, Benincasa M, Klumpp C, Briand JP, Gennaro R, Prato M, Bianco A. Targeted delivery of amphotericin B to cells by using functionalized carbon nanotubes. Angew Chem Int Ed Engl. 2005;44:6358–6362. doi: 10.1002/anie.200501613. [DOI] [PubMed] [Google Scholar]
- 205.Liu Z, Tabakman S, Welsher K, Dai H. Carbon nanotubes in biology and medicine: in vitro and in vivo detection, imaging and drug delivery. Nano Res. 2009;2:85–120. doi: 10.1007/s12274-009-9009-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Shao N, Lu S, Wickstrom E, Panchapakesan B. Integrated molecular targeting of IGF1R and HER2 surface receptors and destruction of breast cancer cells using single wall carbon nanotubes. Nanotechnology. 2007;18:315101. [Google Scholar]
- 207.Casais-Molina ML, Cab C, Canto G, Medina J, Tapia A. Carbon nanomaterials for breast cancer treatment. Journal of Nanomaterials. 2018;2018:2058613. [Google Scholar]
- 208.McKernan P, Virani NA, Faria GNF, Karch CG, Prada Silvy R, Resasco DE, Thompson LF, Harrison RG. Targeted single-walled carbon nanotubes for photothermal therapy combined with immune checkpoint inhibition for the treatment of metastatic breast cancer. Nanoscale Res Lett. 2021;16:9. doi: 10.1186/s11671-020-03459-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Fellner C. Ipilimumab (yervoy) prolongs survival in advanced melanoma: serious side effects and a hefty price tag may limit its use. P T. 2012;37:503–530. [PMC free article] [PubMed] [Google Scholar]
- 210.Bobo D, Robinson KJ, Islam J, Thurecht KJ, Corrie SR. Nanoparticle-based medicines: a review of FDA-approved materials and clinical trials to date. Pharm Res. 2016;33:2373–2387. doi: 10.1007/s11095-016-1958-5. [DOI] [PubMed] [Google Scholar]
- 211.Chen Y, Chen H, Shi J. In vivo bio-safety evaluations and diagnostic/therapeutic applications of chemically designed mesoporous silica nanoparticles. Adv Mater. 2013;25:3144–3176. doi: 10.1002/adma.201205292. [DOI] [PubMed] [Google Scholar]
- 212.Swain S, Sahu PK, Beg S, Babu SM. Nanoparticles for cancer targeting: current and future directions. Curr Drug Deliv. 2016;13:1290–1302. doi: 10.2174/1567201813666160713121122. [DOI] [PubMed] [Google Scholar]
- 213.Mishra D, Hubenak JR, Mathur AB. Nanoparticle systems as tools to improve drug delivery and therapeutic efficacy. J Biomed Mater Res A. 2013;101:3646–3660. doi: 10.1002/jbm.a.34642. [DOI] [PubMed] [Google Scholar]
- 214.Carvalho A, Fernandes AR, Baptista PV. Applications of targeted nano drugs and delivery systems. Elsevier; 2019. Nanoparticles as delivery systems in cancer therapy: focus on gold nanoparticles and drugs; pp. 257–295. [Google Scholar]
- 215.Mahidhara G, Kanwar RK, Roy K, Kanwar JR. Oral administration of iron-saturated bovine lactoferrin-loaded ceramic nanocapsules for breast cancer therapy and influence on iron and calcium metabolism. Int J Nanomedicine. 2015;10:4081–4098. doi: 10.2147/IJN.S75877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Fu Q, Sun J, Zhang W, Sui X, Yan Z, He Z. Nanoparticle albumin-bound (NAB) technology is a promising method for anti-cancer drug delivery. Recent Pat Anticancer Drug Discov. 2009;4:262–272. doi: 10.2174/157489209789206869. [DOI] [PubMed] [Google Scholar]
- 217.Kratz F. A clinical update of using albumin as a drug vehicle-a commentary. J Control Release. 2014;190:331–336. doi: 10.1016/j.jconrel.2014.03.013. [DOI] [PubMed] [Google Scholar]
- 218.Elzoghby AO, Elgohary MM, Kamel NM. Implications of protein- and Peptide-based nanoparticles as potential vehicles for anticancer drugs. Adv Protein Chem Struct Biol. 2015;98:169–221. doi: 10.1016/bs.apcsb.2014.12.002. [DOI] [PubMed] [Google Scholar]
- 219.Langer K, Anhorn MG, Steinhauser I, Dreis S, Celebi D, Schrickel N, Faust S, Vogel V. Human serum albumin (HSA) nanoparticles: reproducibility of preparation process and kinetics of enzymatic degradation. Int J Pharm. 2008;347:109–117. doi: 10.1016/j.ijpharm.2007.06.028. [DOI] [PubMed] [Google Scholar]
- 220.Kouchakzadeh H, Shojaosadati SA, Shokri F. Efficient loading and entrapment of tamoxifen in human serum albumin based nanoparticulate delivery system by a modified desolvation technique. Chemical Engineering Research and Design. 2014;92:1681–1692. [Google Scholar]
- 221.Qu N, Sun Y, Li Y, Hao F, Qiu P, Teng L, Xie J, Gao Y. Docetaxel-loaded human serum albumin (HSA) nanoparticles: synthesis, characterization, and evaluation. Biomed Eng Online. 2019;18:11. doi: 10.1186/s12938-019-0624-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.KÖHler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256:495–497. doi: 10.1038/256495a0. [DOI] [PubMed] [Google Scholar]
- 223.Harbeck N, Beckmann MW, Rody A, Schneeweiss A, Müller V, Fehm T, Marschner N, Gluz O, Schrader I, Heinrich G, Untch M, Jackisch C. HER2 dimerization inhibitor pertuzumab-mode of action and clinical data in breast cancer. Breast Care. 2013;8:49–55. doi: 10.1159/000346837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Nami B, Maadi H, Wang Z. Mechanisms underlying the action and synergism of trastuzumab and pertuzumab in targeting her2-positive breast cancer. Cancers (Basel) 2018;10:342. doi: 10.3390/cancers10100342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Iqbal N, Iqbal N. Human epidermal growth factor receptor 2 (HER2) in cancers: overexpression and therapeutic implications. Mol Biol Int. 2014;2014:852748. doi: 10.1155/2014/852748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Mitri Z, Constantine T, O’Regan R. The HER2 receptor in breast cancer: pathophysiology, clinical use, and new advances in therapy. Chemother Res Pract. 2012;2012:743193. doi: 10.1155/2012/743193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Steinhauser I, Spänkuch B, Strebhardt K, Langer K. Trastuzumab-modified nanoparticles: optimisation of preparation and uptake in cancer cells. Biomaterials. 2006;27:4975–4983. doi: 10.1016/j.biomaterials.2006.05.016. [DOI] [PubMed] [Google Scholar]
- 228.White BE, White MK, Adhvaryu H, Makhoul I, Nima ZA, Biris AS, Ali N. Nanotechnology approaches to addressing HER2-positive breast cancer. Cancer Nanotechnology. 2020;11:12. [Google Scholar]
- 229.Mitchell MJ, King MR. Leukocytes as carriers for targeted cancer drug delivery. Expert Opin Drug Deliv. 2015;12:375–392. doi: 10.1517/17425247.2015.966684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Li Y, Tan Y, Ning Z, Sun S, Gao Y, Wang P. Design and fabrication of fluorescein-labeled starch-based nanospheres. Carbohydr Polym. 2011;86:291–295. [Google Scholar]
- 231.Mora-Huertas CE, Fessi H, Elaissari A. Polymer-based nanocapsules for drug delivery. Int J Pharm. 2010;385:113–142. doi: 10.1016/j.ijpharm.2009.10.018. [DOI] [PubMed] [Google Scholar]
- 232.Blanco-Padilla A, Soto KM, Hernández Iturriaga M, Mendoza S. Food antimicrobials nanocarriers. ScientificWorldJournal. 2014;2014:837215. doi: 10.1155/2014/837215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Barratt GM. Therapeutic applications of colloidal drug carriers. Pharm Sci Technol Today. 2000;3:163–171. doi: 10.1016/s1461-5347(00)00255-8. [DOI] [PubMed] [Google Scholar]
- 234.Huang Y, Liu M, Gao C, Yang J, Zhang X, Zhang X, Liu Z. Ultra-small and innocuous cationic starch nanospheres: Preparation, characterization and drug delivery study. Int J Biol Macromol. 2013;58:231–239. doi: 10.1016/j.ijbiomac.2013.04.006. [DOI] [PubMed] [Google Scholar]
- 235.Lamberti M, Zappavigna S, Sannolo N, Porto S, Caraglia M. Advantages and risks of nanotechnologies in cancer patients and occupationally exposed workers. Expert Opin Drug Deliv. 2014;11:1087–1101. doi: 10.1517/17425247.2014.913568. [DOI] [PubMed] [Google Scholar]
- 236.Bachar M, Mandelbaum A, Portnaya I, Perlstein H, Even-Chen S, Barenholz Y, Danino D. Development and characterization of a novel drug nanocarrier for oral delivery, based on self-assembled β-casein micelles. J Control Release. 2012;160:164–171. doi: 10.1016/j.jconrel.2012.01.004. [DOI] [PubMed] [Google Scholar]
- 237.Zhang XY, Zhang PY. Polymersomes in nanomedicine-a review. Curr Med Chem. 2017;13:124–129. [Google Scholar]
- 238.Vega-Vásquez P, Mosier NS, Irudayaraj J. Nanoscale drug delivery systems: from medicine to agriculture. Front Bioeng Biotechnol. 2020;8:79. doi: 10.3389/fbioe.2020.00079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Gaur PK, Mishra S, Purohit S. Solid lipid nanoparticles of guggul lipid as drug carrier for transdermal drug delivery. Biomed Res Int. 2013;2013:750690. doi: 10.1155/2013/750690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Felber AE, Dufresne MH, Leroux JC. pH-sensitive vesicles, polymeric micelles, and nanospheres prepared with polycarboxylates. Adv Drug Deliv Rev. 2012;64:979–992. doi: 10.1016/j.addr.2011.09.006. [DOI] [PubMed] [Google Scholar]
- 241.Isailović BD, Kostić IT, Zvonar A, Đorđević VB, Gašperlin M, Nedović VA, Bugarski BM. Resveratrol loaded liposomes produced by different techniques. Innovative Food Science & Emerging Technologies. 2013;19:181–189. [Google Scholar]
- 242.Ramachandran R, Paul W, Sharma CP. Synthesis and characterization of PEGylated calcium phosphate nanoparticles for oral insulin delivery. J Biomed Mater Res B Appl Biomater. 2009;88:41–48. doi: 10.1002/jbm.b.31241. [DOI] [PubMed] [Google Scholar]
- 243.Singh D, Dubey P, Pradhan M, Singh MR. Ceramic nanocarriers: versatile nanosystem for protein and peptide delivery. Expert Opin Drug Deliv. 2013;10:241–259. doi: 10.1517/17425247.2012.745848. [DOI] [PubMed] [Google Scholar]
- 244.[Internet]. 2022 [cited 28 January 2022] Available from: https://www.drugs.com/mtm/pertuzumab.html.
- 245.[Internet]. 2022 [cited 28 January 2022] Available from: https://www.drugs.com/monograph/trastuzumab.html.
