Home Recent developments and applications of smart nanoparticles in biomedicine
Article Open Access

Recent developments and applications of smart nanoparticles in biomedicine

  • Simona Campora and Giulio Ghersi EMAIL logo
Published/Copyright: July 11, 2022
Become an author with De Gruyter Brill

Abstract

Over the last decades, nanotechnology applied in medicine (nanomedicine) has sparked great interest from the scientific community, thanks to the possibility to engineer nanostructured materials, including nanoparticles (NPs), for a specific application. Their small size confers them unique properties because they are subject to physical laws in the middle between classical and quantum physics. This review is proposed to explain better how to design a specific NP and clarify the relationship between the type, size, and shape of NPs and the specific medical applications. NPs are classified into inorganic (metallic NPs, quantum dots, carbon-based nanostructures, mesoporous silica NPs) and organic (liposomes and micelles, dendrimers, and polymer NPs). Here, we report an accurate description of the potential of each NPs type focusing on their multiple areas of application, including theranostics drug delivery, imaging, tissue engineering, antimicrobial techniques, and nanovaccines. All these features make NPs a promise to revolutionize the new era of nanomedicine.

Graphical abstract

1 Introduction

Nanoscience represents one of the most exciting fields of modern science, with a highly interdisciplinary character; indeed, it develops by combining different doctrines like chemistry, biology, physics, and engineering, taking advantage of their principles and processes. It is based on understanding and knowledge of the matter properties on the nanometric scale (between 1 and 100 nm). The realization of materials, systems, and apparatuses on this size scale determines nanotechnology [1].

The term “nanotechnology” was first defined by Norio Taniguchi of Tokyo Science University in 1974 [2]. In the 1980s, the idea of nanotechnology as deterministic, rather than stochastic, handling of individual atoms and molecules was conceptually explored in depth by Dr. K. Eric Drexler, who called it molecular nanotechnology [3]. It is a continually evolving field that finds application in many productive sectors, including cosmetics, coating and paints, nano-hard disks, and memory chips. One of the significant applications concerns the biomedical environment (nanomedicine) focused on tissue engineering [4,5], drug delivery [6], nanovaccines [7], antimicrobial techniques [8], and bioimaging [9].

Nanoparticles (NPs) are dispersion solutions of atomic aggregates or solid particles with a size between 1 and 300 nm and specific properties, like the high surface-to-mass ratio. Furthermore, the small size permits them to circulate more freely in the human body. NPs present unique chemical, magnetic, mechanical, and biological properties that increase biocompatibility and cellular uptake. The possibility to engineer their surface permits multifunctional applications, especially in the clinical environment for diagnosis and therapies [10]. The classification and applications of NPs in biomedicine are illustrated in Figure 1. The combination of diagnosis and therapy (theranostic) applications has amply been employed for treatment of diseases. Depending on the nature and the functionalization (e.g., with fluorescence probes), NPs have been applied for different bioimaging studies, including magnetic resonance imaging (MRI) contrast agents, Positron emission tomography (PET), and optical, magnetic, and radioactive imaging (Figure 1). In this manner, NPs can be adopted for early disease detection, image-guided disease treatment, and the evaluation of a therapeutic effect.

Figure 1 
               Nanoparticles classification and applications in biomedicine. Inorganic NPs include metal and magnetic NPs (gold, silver, SPIONs), QDs, CBNs, and MSNs; organic NPs collect lipid-based NPs (liposomes and micelles), PNPs and dendrimers. Depending on the material, size, shape, and functionalization, NPs can be employed in nano-vaccines both for protein and RNA delivery; theranostics drug delivery that include treatment and diagnosis of many pathologies like cancer and NDs; antimicrobial (anti-viral, bacteria and biofilms) techniques; imaging for CT, MRI, SERS, and fluorescence analysis; tissue engineering in regenerative medicine for growth factors release or cell differentiation.
Figure 1

Nanoparticles classification and applications in biomedicine. Inorganic NPs include metal and magnetic NPs (gold, silver, SPIONs), QDs, CBNs, and MSNs; organic NPs collect lipid-based NPs (liposomes and micelles), PNPs and dendrimers. Depending on the material, size, shape, and functionalization, NPs can be employed in nano-vaccines both for protein and RNA delivery; theranostics drug delivery that include treatment and diagnosis of many pathologies like cancer and NDs; antimicrobial (anti-viral, bacteria and biofilms) techniques; imaging for CT, MRI, SERS, and fluorescence analysis; tissue engineering in regenerative medicine for growth factors release or cell differentiation.

Conversely, NPs can act like vectors able to carry biological molecules (e.g., drugs) to a specific tissue and release them with a controlled mechanism. These characteristics make them optimal candidates as drug delivery systems (DDS) in many pathologies, including neurodegenerative diseases (NDs) and tumours. Despite many progressions in the last decades, cancer remains one of the most devastating diseases globally, causing 10 million deaths in 2020 (data from OMS). The tumour heterogeneity limits the formulation of standard therapy. Therefore, conventional drug administration systems (CDASs; parenteral, oral, cutaneous, or topical) for diseases and, in particular, cancer chemotherapy can induce side effects because of their nonspecific action as they act both on healthy and malignant cells [11]. Based on the tumour cells’ high capability to divide out of control, CDASs are developed to destroy the rapidly dividing cells, unfortunately including the body’s other rapidly proliferating cells, such as cells in the hair follicles, myelopoietic bone marrow precursor, and intestinal epithelial cells [12]. Therefore, the knowledge of definite cancer physiology and structure can be the starting point for designing engineered NPs for a specific tumour targeting.

Furthermore, the drug dilution in the bodily fluids limits its absorption in the target tissue, so it is necessary to administer substantial doses to have a high local concentration. Conversely, the use of nanosystems as DDS permits a controlled release of the conjugated drug, depending on physiological conditions of the targeted site (site-specific targeting) and modulation during the release time (temporal modulation), related to the physical properties of the microenvironment [13].

Disease treatment is only one of the multiple applications in which NPs can be involved. Depending on their nature, NPs can present antibacterial or antiviral properties and also anti-angiogenetic and anti-neoplastic effects [14,15] (Figure 1). Furthermore, they are abundantly employed in tissue engineering to promote tissue differentiation, thanks to the possibility of local delivery of bioactive (growth factors, chemokines, inhibitors, cytokines, genes, etc.) and contrast agents in a controlled way [16].

Moreover, in the last decades, the use of different kinds of NPs as delivery systems in vaccines sparked great interest from the scientific community, thanks to their potential to improve vaccine efficacy and reduce the risk of attenuated vaccines. The encapsulation protects the antigens from early proteolytic degradation, permits a controlled antigen release, and helps antigen uptake and processing by antigen-presenting cells. In addition, the possibility of obtaining a specific target can improve vaccine formulation [17].

Therefore, NP systems, including the choice of the size, shape, composition (material), and surface properties, play a pivotal role in optimizing their use in biomedical applications.

2 Project of NPs

NP functionalization plays a crucial role in their activities in a wide range of delivery applications, including treatment of different diseases (tumours, neurodegenerative, and metabolic pathologies), bioimaging, tissue engineering, nanovaccines, and antimicrobial techniques (Figure 1). The engineered nanomaterials can be synthesized by two different approaches, top-down and bottom-up. Top-down is a physical approach that reduces macrostructures, named bulk materials, through incisions, grindings, and cuttings [18]. Conversely, bottom-up is a chemical approach that produces NPs from atoms or molecule aggregates [19]. It is the typical synthesis mechanism adopted in the biomedical field because it permits the formulation of nanostructures with the desired properties through a specific controlled process [20]. Starting from this, the size, shape, composition (material), and surface properties must be considered and analysed to increase the circulating half-life, biocompatibility, drug loading, corresponding site-specific release, and the specific site targeting [14].

2.1 NPs size

Morphological characteristics, like size and shape, play a pivotal role in NP-based drug delivery. Size change in the nanoscale can influence physical properties (like optical absorption or melting points that decrease in a size-dependent way), chemical reactions (like thermodynamic features), and magnetic properties, especially for metal NPs (MNPs) and electrical properties. The size of NPs can influence different aspects like body and tissue distribution, cellular response, and blood extravasation as shown in Figure 2.

Figure 2 
                  Effect of nanoparticles size on body and tissue distribution, cellular response, and blood extravasation. Depending on the size, NPs can localize differently in body compartments (pharmacokinetics) and can cleaned from different organs: kidney (<5 nm), liver (10–20 nm), spleen (20–200 nm), and lung (inhaled NPs >100 nm). Cellular response exploited cytotoxicity and cellular uptake mediated by micropinocytosis (big NPs), clathrin-mediated endocytosis (100 nm) and caveolae-mediated endocytosis (15–80 nm). NPs size influence also the EPR effect for passive tumour targeting and the cross thought BBB.
Figure 2

Effect of nanoparticles size on body and tissue distribution, cellular response, and blood extravasation. Depending on the size, NPs can localize differently in body compartments (pharmacokinetics) and can cleaned from different organs: kidney (<5 nm), liver (10–20 nm), spleen (20–200 nm), and lung (inhaled NPs >100 nm). Cellular response exploited cytotoxicity and cellular uptake mediated by micropinocytosis (big NPs), clathrin-mediated endocytosis (100 nm) and caveolae-mediated endocytosis (15–80 nm). NPs size influence also the EPR effect for passive tumour targeting and the cross thought BBB.

In fact, the size of NPs needs to be chosen with particular attention because nanosystems have to be small enough to escape the capture from the cells of the mononuclear phagocyte system (i.e., in the spleen and the liver) and big enough to avoid their rapid leakage into blood vessels following by the renal clearance [21]. Depending on the administration technique, cytotoxicity and adsorption of NPs across the epithelial barrier are related to their size (Figure 2). For example, inhalation enables penetration in the lung parenchyma, showing a different localization in the respiratory tract. Conversely, as reported by Braakhuis et al., the cytotoxic effect in rats of inhaled silver NPs is related to their dimension: NPs of 18 and 34 nm induced cell damage in a concentration-dependent way. Simultaneously, there was no dose-dependent toxicity of 60 and 160 nm NPs [22].

Many studies have evaluated the pharmacokinetics of NPs (in vivo distribution) and revealed a size-dependent different organ distribution as assessed by Ibrahim et al.: 5 nm gold nanoparticles (AuNPs) preferentially addressed to the liver, while bigger AuNPs of 20 and 50 nm localized on the spleen [23]. De Jong et al. also had analysed AuNPs size-dependent tissue distribution reporting an exclusive localization of 10 nm AuNPs in the testis, thymus, heart, and brain [24]. For instance, the size of NPs plays a pivotal role in passive tumour targeting by the enhanced permeability and retention effect due to the leaky vascularization of tumour tissues. It is unlikely that NPs bigger than 200 nm reach tumour mass due to spleen clearance, while NPs with a size smaller than 30 nm can travel back from the cancer mass to the blood vessels. Therefore, the optimal nanosystem size for passive tumour targeting is between 30 and 200 nm [25].

Smaller NPs are generally cleared by drainage organs like the kidney (<5 nm) or the liver (10–20 nm), while the bigger ones (20–200 nm) accumulate in the spleen, bone marrow, and other reticuloendothelial systems (RES) [26]. The clearance of NPs also depends on the inhalation process; for instance, pulmonary clearance by alveolar macrophages concerns big aerosol-based NPs (>100 nm). By comparing the biodistribution of 13 and 105 nm of inhaled AuNPs, Han and colleagues had noticed an initial rat lung deposit of both NPs, but significantly higher biodistribution from the lung to other organs of the smaller AuNPs. In addition, lung clearance was considerably higher for bigger NPs.

The optimal size of NPs must be ranged between 1 and 100 nm, especially to cross the blood–brain barrier (BBB), as suggested by neurodegenerative disease studies, including Alzheimer, Parkinson, or glioma. The biggest problem with treating cerebral pathologies is the impossibility or high limits of drugs to pass through the BBB. Their conjugation with NPs of different natures (i.e., polymeric, inorganic, or liposomes) permits them to cross the BBB by active (receptor-mediated or adsorption-mediated endocytosis or carrier-mediated transport) or passive (diffusion through endothelial cells) transport mechanisms [27]. NPs smaller than 10 nm cross BBB through a transcellular passage, while bigger NPs are involved in the transcytosis mechanism [28]. For example, one of the most used drugs for Alzheimer’s disease is the anti-amyloidogenic drug curcumin, which is unable to cross the BBB. Hence, Barbara et al. encapsulated it in polylactide-co-glycolic-acid (PLGA) NPs modified with a g7 ligand that permits the BBB crossing. An intensive decrease of Aβ aggregates in response to curcumin-loaded NPs was registered, suggesting a possible approach to treating Alzheimer’s disease [29].

A decrease in NP dimension corresponds to a higher surface area-to-volume ratio, suggesting that more conjugated drugs could be associated with or near the NP surface [30]. Furthermore, cellular uptake also depends on the size [31] (Figure 2). NPs are internalized faster and 15–250 times more than microparticles of 1–10 µm through many mechanisms: large NPs are generally involved in micropinocytosis; 100 nm NPs in clathrin-mediated endocytosis; and 15–80 nm NPs in caveolae-mediated endocytosis [32,33].

2.2 NP shape

The shape of NPs confers peculiar features that influence blood lifespan, macrophage uptake, and cell membrane interaction (Figure 3). Generally, NPs are injected into the blood vessels and are subjected to Brownian motion and convective forces, inducing rotation and rolling, especially for oblate-shaped spherical NPs [34]. In fact, blood circulation depends on nanosystem shape, as suggested by Geng and coworkers who showed that polymer filomicelles persisted in the circulation of rodents about ten times more than their spherical counterparts (more than 1 week against 2–3 days), probably due to the possibility to align to the blood fluid [35]. Zhao et al. also confirmed these data, which reported the more prolonged bloodstream circulation of the long rod mesoporous silica nanoparticles (NLR) compared with the short rod and spherical ones [36]. They also investigated the body biodistribution after rat oral administration; although the liver and kidney took up all the NPs, NLR had the longest residence time in the gastrointestinal.

Figure 3 
                  Factors affected by nanoparticles shape: blood circulation and lifetime; cellular internalization (depending on the NPs AR and the contact angle); and endocytosis pathways (clathrin-mediated endocytosis, caveolin-mediated endocytosis, and micropinocytosis).
Figure 3

Factors affected by nanoparticles shape: blood circulation and lifetime; cellular internalization (depending on the NPs AR and the contact angle); and endocytosis pathways (clathrin-mediated endocytosis, caveolin-mediated endocytosis, and micropinocytosis).

The major part of the NPs’ nonspecific clearance depends on the spleen and liver’s mononuclear phagocytic system (MPS). Their retention can induce an inflammation state [37]. Therefore, their blood lifetime is improved by evoking the macrophages’ phagocytosis in the reticuloendothelial system to reach the target tissue. Many strategies have been adopted to avoid the MPS, including the functionalization with polyethylene glycol (PEGylation), which permits the formation of a hydrating layer due to the association with water molecules [38]. This manner prevents the NP aggregation and interaction with blood components like opsonins, prolonging systemic circulation time [39]. PEGylated particles’ behaviour is related to PEG molecular weight and surface density, which influences its superficial properties [40]. Another approach consists of the NP’s “mimetic effect” by conjugating them with “self” molecules like CD47 peptides [41] or coating them with cell membranes extracted from autologous leukocytes [42] and red blood cells [43].

Cellular uptake and internalization depend on the NPs’ aspect ratio (AR) and the contact angle between NP and surface. Elongated rod nanosystems with very high AR attached better to the cell membrane than spheroidal or rod NPs with lower AR, but are phagocytosed less efficiently [44]. This is probably related to the alignment of the longer axis parallel to the cell membrane: in this case, its internalization is more difficult compared to sphere-shaped NPs. Furthermore, the geometry of the initial contact of the NP with the macrophage (tangent angles) determinates the cell response: the cell starts to remodel the actin cytoskeleton to cover and engulf the nanosystem only when the smaller axis of the oblate-shaped NP contacts the cell membrane. On the contrary, depending on the local particle shape, an incorrect interaction fails to correctly organize the actin, inducing a superficial spreading without any internalization [45,46]. Shape-dependent different macrophage uptake is also attributed to the different endocytosis pathways: spherical AuNPs are generally internalized by clathrin- and caveolin-mediated endocytosis. In contrast, the cylindrical ones are clathrin-mediated endocytosis. Moreover, the elongated shape induces a more efficient interleukin 6 inflammatory response than the shorter rod or spherical ones [47]. Shape-related differential uptake grade was also individuated in other cells, such as the tumour cells. For instance, breast cancer cells show a preferential uptake of rod NPs, followed by dishes and spheres [48].

Furthermore, the shape can also influence specific nanosystem features. Xu et al. reported a relation between morphology and reaction rate of silver NPs (AgNPs): the reaction rate of nanocubes was 14 times higher than that of triangular ones and four times more than the semi-spherical ones [49]. The morphology of AgNPs also influences their antibacterial effects: naocomplexes with a higher specific surface area resulted in more toxicity for bacteria than smaller ones due to the difference in the Ag ion release depending on the shape [50,51]. Conversely, the shape plays a pivotal role in mechanical properties and adhesion with hydrogel materials, as suggested by Arno et al. Analyzing the interaction between polymeric NPs and calcium-alginate hydrogels, they found an increase in the adhesion and the material’s mechanical strength concerning spherical or cylindrical counterparts [52].

2.3 NPs material

NPs can be classified into inorganic and organic depending on the material used. As shown in Figure 1, the first one includes MNPs, quantum dots (QDs), carbon-based nanostructures (CBNs), and mesoporous silica NPs (MSNs). At the same time, liposomes and micelles, dendrimers, and polymeric NPs represent the organic ones. A summary of the features of the major NP classes covered in this review is presented in Table 1.

Table 1

Advantages, disadvantages, and biomedical applications of each type of nanoparticle

NPs Advantages Disadvantages Functionalization Applications in nanomedicine Ref.
Metallic NPs AuNPs • Biocompatibility • Not biodegradable • Targeting molecules (antibodies; glycans, etc.) • Bio-imaging (CT/X-Ray) [61,64,66,67,68,72,73]
• Easy to synthesize and conjugate to biological molecules • Nanoparticles aggregation • PEG • Drug delivery
• High X-ray attenuation • Photothermal therapy
• SPR • Photodynamic therapy
• Tumour therapy
        • Nano-vaccines
AgNPs • Easy to synthesize • Toxic at higher concentrations • PEG • Drug delivery [76,79,80,81,82,84,85,90,91]
• Antibacterial and antiviral activity • Various ecological problems if released into the environment • Antiviral and antibacterial activity (inhibition of bacterial biofilm formation and EPS production)
• Anti-inflammatory and antitumor capacity • Antineoplastic effect
• Antiangiogenic effects
Magnetic NPs • Possibility to magnetize by the external field • Limitation to maintain efficacy in the target organ once the magnetic field is removed • Targeting molecules (folic acid, etc.) • Magnetic biosensing [98,99,101,104]
• Well-controlled activation/deactivation mechanism • Nanoparticles aggregation • Coated with silicon, dextran citrate or PEG • MRI
• Toxic effect • Magnetic separation
• Drug and gene delivery
• PTT
• PDT
        • RT
QDs • Imaging properties • Toxic effect of metal core • Biological molecules (peptides, proteins, nucleic acids, etc.) • Drug delivery [109,112,113,114,115,120,121,129]
• Capability to conjugate different biological molecules • Nanoparticles aggregation • Targeting molecules (antibodies, hyaluronic acid, folic acid, etc.) • Bio-imaging
• PEG • Cancer diagnosis and treatment
• Theranostic application
       
CBNs • Easy to synthesize and conjugate to biological molecules • Not biodegradable • Targeting molecules (antibodies, etc.) • Drug delivery [142,143,144,145,153,159,160]
• Large surface area • Potential material toxicity • Fluorescence probes • Bio-imaging
• Protect entrapped molecules • Poorly soluble in water • Biological molecules (proteins, nucleic acids, etc.) • Cancer diagnosis and treatment
• Drugs • Tissue engineering
• PEG • Photothermal therapy
  • Biosensors
MSNs • High surface to volume ratio to conjugate with biological molecules • Not biodegradable • Targeting molecules (antibodies, etc.) • Bio-imaging [185,186,187,189,192,193,196,197,198,200,202]
• Stability • Potential cell lysis caused by silanol groups interacting with membrane lipids • Fluorescence probes • Drug delivery
• Easy control of morphology, pore distribution, and size • Biological molecules (proteins, nucleic acids, etc.) • Cancer treatment
• Biocompatibility • Drugs • Tissue engineering
      • PEG • Nano-vaccines
Lipid-based NPs (LNPs) • Biocompatibility • Low solubility and stability • Targeting molecules (antibodies, etc.) • Drug delivery [213,215,218,219,221,224,226,228,230,231,232,264,268,273,274,277]
• Biodegradable • Tends to agglomerate • PEG or heparin or albumin or polysaccharides (chitosan) • Cancer treatment
• Amphiphilic • Some may be allergic • Hydrophilic and/or hydrophobic drugs • Neurodegenerative disease treatment
• Longer duration of circulation • May trigger an immune response • mRNA • Trojan Horse Liposome (THL) technology (e.g., to cross BBB)
  • Drugs • Nano-vaccines
Dendrimers • High drug loading ability • Immunoreaction • Targeting molecules (antibodies, RGD, etc.) • Drug delivery [293,301,304,306,308,309,311,349]
• Low polydispersity, • Haematological toxicity • Biological molecules (peptides, siRNA, small DNA, etc.) • Cancer treatment
• Reproducible pharmacodynamics and pharmacokinetic behaviour • Toxicity for prokaryotic and eukaryotic cells • Drugs • Antiviral and antibacterial activity
• High cellular uptake • PEG • Tissue engineering
  • Capability to cross BBB   • Anionic groups (acid or sulfonate residues)  
Polymer NPs • Biocompatibility • Inflammatory response • Targeting molecules (antibodies, folic acid, etc.) • Drug delivery [32,337,339,340,341,343,344,345,346,347]
• Biodegradable • Nanoparticles aggregation depending on the polymer used • PEG • Cancer treatment
• Variety for chemical composition • Biological molecules (GF, peptides, etc.) • Tissue engineering
• Stability • Nano-vaccines

2.3.1 Inorganic NPs

Inorganic NPs play a fundamental role in modern materials science due to their unique physical characteristics like size-dependent optical, magnetic, electronic, and catalytic properties. Moreover, they can be quickly and cheaply synthesized and mass-produced, and hence, they can also be more readily used for many applications. These inorganic NPs include metallic ones like gold and silver, QDs, CBNs, and MSNs [53].

2.3.2 Metallic nanoparticles and magnetic nanoparticles

MNPs are amply employed in biomedical applications such as targeted drug delivery, antimicrobial activity, bio-imaging, and diagnosis and disease therapy [54] (Figure 4; Table 1). In addition, they can be used as imaging probes in many techniques like ultrasound (US), X-ray, computed tomography (CT), PET, MRI, optical imaging, and surface-enhanced Raman imaging (SERS) [55].

Figure 4 
                     Metal and magnetic nanoparticles (MNPs and MgNPs) including AuNPs, AgNPs, and SPIONs. They present a natural antimicrobial activity (antibacterial and antiviral); can act as delivery carriers both in drug delivery and nanovaccines; can be employed in diagnostics (CT/X-ray and fluorescence analysis) and in disease therapy (e.g., tumour therapy, photothermal, and photodynamic therapy).
Figure 4

Metal and magnetic nanoparticles (MNPs and MgNPs) including AuNPs, AgNPs, and SPIONs. They present a natural antimicrobial activity (antibacterial and antiviral); can act as delivery carriers both in drug delivery and nanovaccines; can be employed in diagnostics (CT/X-ray and fluorescence analysis) and in disease therapy (e.g., tumour therapy, photothermal, and photodynamic therapy).

In this context, AuNPs have recently attracted interest for their use as CT imaging contrast agents, thanks to their high X-ray attenuation, simple synthesis, surface properties, and biocompatibility [56]. Furthermore, they present peculiar absorption and scattering properties like surface plasmon resonance (SPR) that can be tuned by controlling the specific size and shape (sphere, rod, and clusters), depending on the synthesis method [57]. AuNPs can be produced through physical (like the microwave and ultraviolet irradiation or laser ablation), chemical (like the Turkevich method that consists of the reduction of gold chloride with sodium citrate), and biological (plants and microorganisms mediated) ways [58]. The last one is relatively new and attracted great attention because of its eco-friendly character because microorganisms can adsorb gold atoms and collect AuNPs by secreting enzymes involved in the enzymatic reduction of gold ions [59].

AuNPs are widely used in academic research for the tumour treatment [60]. Some studies revealed their potential in limiting angiogenesis and tumour progression; Li et al. suggested and demonstrated the AuNPs inhibition effects on epithelial–mesenchymal transition and tumour vasculature normalization [61,62].

Furthermore, their functionalization with targeting molecules permits their specific cancer mass penetration and the release of the associated anticancer drug [63]. Pedrosa et al. had designed multifunctional AuNPs carrying a novel chemotherapeutic candidate (ZnD) and monoclonal antibody cetuximab to recognize the epidermal growth factor receptor (EGFR) overexpressed in cancer cells. Data suggested a specific tumour targeting in a colorectal DOX-resistant model, leading to a reduction in the tumour growth without systemic toxicity [64].

Furthermore, AuNPs show an optical scattering that can be exploited for the nanophotolysis technique using a short-pulse laser. Laser photothermal therapy (PTT) is based on the capability of cancer-targeted AuNPs to absorb light and convert it into heat, which leads to thermal explosion when the threshold laser flow is 25–40 mJ/cm2 [65]. Many studies suggest this approach for cancer therapy because it is an effective way to selectively kill tumour cells without affecting healthy ones. In this context, Zheng et al. have synthesized a hybrid nanoplatform made of photothermal gold nanostars and a glycopolymer containing both galactose groups (to target the tumour asialoglycoprotein receptor) and glucose groups (to recognize concanavalin A [ConA], which is cytotoxic to hepatoma cells). A decrease in the tumour size was observed under infrared laser irradiation due to the synergic effect of photothermal and drug therapy [66]. Another form of light therapy adopted against cancer cells and pathogenic bacteria is photodynamic therapy (PDT) based on AuNPs as a photosensitizer (PS). When excited by near-infrared (NIR) light, AuNPs transfer energy to the surrounding O2 to generate reactive oxygen species (ROS), inducing cell death [67]. PTT and PDT can be combined with radiation therapy (RT) to produce a multifunctional nanotheranostic gold nanocage, as reported by Xy and colleagues. They had synthesized hyaluronic acid-modified Au nanocages (AuNCs-HA) acting as a contrast agent for enhanced photoacoustic (PA) imaging to provide contour, size, and location information of the tumour. By combining radiotherapy and phototherapy, AuNCs-HA could inhibit the tumour growth compared to each therapy alone [68].

All the AuNP features make them an optimal candidate for vaccine formulations acting both as delivery systems and adjuvants [69]. Peptide–AuNP conjugates can be internalized by macrophages, resulting in their activation, and AuNPs of 8–17 nm size bring a strong antibody response with low cytotoxicity [70,71]. The NPs’ efficiency as adjuvants could depend on their shape as observed by Tazaki et al.: only gold nanorods, but not spherical ones, were able to enhance the intranasal inactivated influenza vaccine adjuvanticity [72]. Conversely, Gulla et al. have proposed AuNPs as vectors for in vivo mannose receptor-mediated targeting of DNA vaccines to mouse dendritic cells (DCs). The formulated nanovaccines delivered the melanoma antigen (pCMV-MART1) encoded DNA to DCs, inducing a long-lasting immune response against murine melanoma and significant inhibition of the melanoma growth [73].

AuNPs can be recognized from the plasma proteins (opsonization) and processed by the RES in the bloodstream. To prevent this process, AuNPs can be functionalized by adding PEG (PEGylation), which prolongs their blood circulation [74].

PEGylation is also adopted for AgNPs because it increases human cell biocompatibility and inhibits platelet aggregation under flow conditions [75]. Furthermore, due to their low-cost production and low toxicity and immunological response [76], AgNPs are amply used in biomedical applications for drug delivery, bioimaging, and molecular diagnostics, and some of them are FDA (Food and Drug Administration) approved and available in the market (Table 2) [77].

Table 2

Approved marketed nanoparticles and their application in biomedicine

Name NPs type/drug Application Approval date Ref.
LNPs
Diprivan Liposome/propofol Anaesthesia 1989 [203,352]
Doxil Liposome/doxorubicin Kaposi sarcoma, ovarian cancer 1995 [203,352,353,354]
DaunoXome Liposome/daunorubicin Kaposi sarcoma 1996 [203,352,353,354]
AmBisome Liposome/amphotericin B Fungal infection 1997 [352,353,354]
DepoCyt Liposome/cytarabine Lymphoma 1999 [352,353]
Visudyne Liposome/verteporfin Myopia, ocular histoplasmosis 2000 [203,352,353,354]
Mepact Liposome/mifamurtide Myosarcoma 2009 [203,352]
Marquibo Liposome/vincristine Acute lymphoblastic, leukaemia 2012 [203,352,353,354]
Onivyde Liposome/irinotecan Pancreatic cancer 2015 [203,352,353,354]
Vyxeos Liposome/cytarabine/daunorubicin Acute myeloid leukaemia 2017 [203,352,353,354]
Onpattro Liposome/patisiran sodium Transthyretin-mediated amyloidosis 2018 [203,352,354]
Comirnaty [BNT162b2] [Pfizer-BioNTech] Lipid-based NPs/mRNA COVID-19 2020 [352,354]
Spikevax [mRNA-1273] [Moderna] Lipid- based NPs/mRNA COVID-19 2020 [352,354]
Polymer-based nanoparticles
Oncaspar Polymer-protein/pegaspargase Acute lymphoblastic leukaemia 1994 [352,353,354]
Copaxone Polymer-protein/glatiramer acetate Multiple sclerosis 1996 [352,353,354]
Verelan PLGA NPs/verapamil HCl Hypertension, angina, rhythm disorders 1998 [352]
Renagel polyallylamine hydrochloride/epichlorohydrin Renal disease, hyperphosphatemia 2000 [353]
PegIntron PEGylated IFN alpha 2B Hepatitis C 2001 [352,354]
Neulasta PEGylated GCSFprotein Neutropenia, chemotherapy induced 2002 [352,353,354]
Pegasys PEGylated IFN alpha 2A Hepatitis Band C 2002 [352,353]
Eligard PLGH/leuprolide acetate Prostate cancer 2002 [352,353,354]
Somavert PEGylated/visomant Acromegaly 2003 [352,353]
Estrasorb Micellar/estradiol Vasomotor symptoms of menopause 2003 [353]
Cimzia PEGylated/certolizumab Crohn’s disease, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis 2008 [352,353,354]
Krystexxa Polymer-protein/pegloticase Chronic gout 2010 [352,353]
Plegridy Polymer-protein/peginterferon beta-1A Multiple sclerosis 2014 [352,353,354]
Adynovate Polymer-protein/pegylated factor VIII Haemophilia 2015 [352,353,354]
Zilretta PLGA hydrogel/trimconolone acetonide Osteoarthritis 2017 [352,353]
Inorganic nanoparticles
INFeD Iron dextran colloid Iron-deficient anaemia 1992 [203,354]
DexFerrum Iron dextran colloid Iron-deficient anaemia 1996 [203,354]
Ferrlecit Iron gluconate colloid Iron deficiency in chronic kidney disease 1999 [203,354]
Venofer Iron sucrose colloid Iron deficiency in chronic kidney disease 2000 [203,354]
Feraheme Iron polyglucose sorbitol carboxymethylether colloid Iron deficiency in chronic kidney disease 2009 [203,353,354]
Injectafer Iron carboxymaltose colloid Iron-deficient anaemia 2013 [203,354]
Ferumoxytol Magnetic nanopartilces MRI 2009 [103]
Nanoparticles for imaging applications
Optison Human serum albumin-stabilized perflutren microspheres Ultrasound contrast agent 1997 [203]
Definity Perflutren lipid microspheres Ultrasound contrast agent 2001 [203]
SonoVue Phospholipid stabilized microbubble Ultrasound contrast agent 2001 [203]

Moreover, AgNPs present excellent antimicrobial and antiviral properties. AgNPs interact with microorganisms and release Ag+ ions that can bind the negatively charged cell walls (due to carboxyl and phosphate groups) and alter the cell permeability [78]. Consequently, ion uptake inhibits the mitochondria respiratory enzymes leading to ROS production, resulting in oxidative stress, ATP production inhibition, and DNA replication [79]. Gram-negative bacteria are more susceptible to AgNPs than Gram-positive ones: in fact, the thick peptidoglycan layer of Gram-positive microorganisms acts as a protective barrier, limiting AgNP internalization [80].

The antibacterial effect of AgNPs is correlated with their size and shape. Hong et al. reported a higher antibacterial effect to E. coli of silver nanocubes to spheres and wires [81]. At the same time, their antimicrobial activity decreases with the increasing particle size, as reported by Raza et al. [50,82]. Therefore smaller spherical AgNPs are more inclined to release silver ions due to their large surface area, bringing a high antimicrobial effect [83]. Many studies suggest the role of AgNPs in inhibiting bacterial biofilm formation and extracellular polymeric substance (EPS) production, mainly when associated with the plant-derived drug-like quercetin [84,85].

The antiviral activity is due to the interference of AgNPs with (1) viral proteins involved in cellular interaction and consequently in cell internalization inhibition or (2) viral DNA/RNA by blocking virus replication and propagation [86]. For instance, AgNPs (especially in NPs size ranging from 1 to 10 nm) are involved in HIV-1 virus infection by interacting with glycoprotein (e.g., gp 120) and preventing CD4-dependent virion binding [87,88,89]. On the other hand, Morris et al. demonstrated the effect of AgNPs on reducing respiratory syncytial virus (RDV) replication and pro-inflammatory cytokine production both in vitro and in vivo experiments [90]. Recently, their potential inhibition effect on SARS-CoV-2 (severe acute respiratory syndrome coronavirus-2) was also observed. By comparing AgNPs of different sizes, Jeremiah et al. had recently demonstrated that 10 nm AgNPs could disrupt viral integrity in vitro [91]. Meanwhile, Balagna et al. applied the antiviral effect of silver to confer an antimicrobial/virucidal effect on individual protection equipment (e.g., facial masks) by a silver nanocluster/silica composite sputtered coating, directly applied on the FFP3 (filtering facepiece 3) masks [92].

AgNPs also act on eukaryotic cells as antineoplastic drugs by inducing apoptosis. They increase ROS levels by reducing mitochondrial membrane potential, releasing cytochrome C into the cytosol, activating JNK, and translocating Bax to mitochondria [93]. This occurs, for example, in Dalton’s lymphoma ascites cell lines both in vitro and in vivo as reported by Sriram et al.: AgNP treatment reduces the volume of the ascitic fluid in tumour-bearing mice by 65%, increasing their survival time by about 50% in comparison with tumour controls [94]. The cytotoxic effect of NPs was also evaluated on healthy human tissues. Many studies have suggested a correlation between toxicology and NP size and morphology: smaller particles can affect cytoplasm and cellular organelles more than bigger ones [95].

In recent decades, great interest was also given to MgNPs, thanks to their unique properties. MgNPs are typically produced from pure metals (such as Fe, Co, Ni) or alloys (such as FeCo, FePt, PtCo; FePd), iron carbides (such as Fe5C2, Fe3C, Fe2C), or ferrites (such as CoFe2O4) [96]. There are different MgNP synthesis methods including chemical and physical (like the ball milling method, coprecipitation, thermal decomposition, hydrothermal, microemulsion, sol-gel method) or biological processes starting from bacteria, plants, fungi, or algae [97].

The possibility to magnetize MgNPs by an external field opens a wide opportunity to be used in a huge range of biomedical applications including magnetic biosensing (diagnostics), magnetic particle resonance (MRI), magnetic separation, drug and gene delivery, and PTT, PDT, RT, etc. In addition, once the external magnetic fields are removed, the magnetization of MgNPs is extinguished, permitting a well-controlled activation/deactivation mechanism.

Magnetic hyperthermia is based on the possibility to generate heat when an external alternating magnetic field is applied to MgNPs. This technique is amply adopted for tumour therapy because the lower pH of the tumour microenvironment makes cancer cells less thermotolerant and, therefore, more susceptible to hyperthermia [98]. The specific cancer effect can be increased by conjugating specific cancer-targeting molecules like folic acid (FA) to obtain a synergistic anti-tumour effect of chemotherapy and heat treatment as suggested by Wang et al. [99]. Superparamagnetic iron oxide nanoparticles (SPIONs) are the most commonly used MgNPs for tumour therapy because of their high biocompatibility, low toxicity for healthy cells, and the capability to induce ROS-mediated cancer cell death by Fenton reaction: Fe2+ ions released by SPIONs can destabilize the hydrogen peroxide (H2O2) generating ˙OH radical groups [100]. Fenton metals, like iron, are amply employed in MRI as contrast agents able to deeply penetrate the tumour mass and provide multimodal imaging modalities [101]: the acid tumour microenvironment induces NP clustering limiting their escape into the bloodstream and increasing the imaging resolution [102]. Therefore, some SPIONs like Ferumoxytol and Endorem were approved by FDA as contrast agents for MRI [103] (Table 2).

ROS generation is involved also in the antibacterial mechanism adopted by MgNPs: in fact, ROS can interfere with the major of bacteria components. This permits the use of magnetic NPs against multi-drug-resistant bacteria and biofilm [104]. In the same manner, NIR-triggered PTT can destroy bacterial cell membranes and cause protein denaturation, selectively killing microorganisms [105]. Under physiological conditions, iron oxide NPs present low solubility and can aggregate in the bloodstream. To reduce these limitations, magnetic NPs can be coated with silicon, dextran citrate, or PEG especially when they are used as contrast agents in target organs [106].

2.3.3 QDs

QDs are very small (2–10 nm) NPs or nanocrystals with an inorganic core of semi-conductor of group II/IV (e.g., cadmium/selenium, cadmium/technetium) and an aqueous organic coated shell (e.g., zinc sulphide, cadmium sulphide). Typically, their semiconducting nature confers unique optical and electronic properties. Depending on the core structure and composition, QDs can emit different colours over a wide spectral range if excited by the same light source (Figure 5). Therefore, they are amply employed as fluorescent probes in cellular and in vivo molecular imaging [107]. In this context, Zhou et al. have developed the faster method point of care testing quantum dot fluorescence immunoassay to detect the high-sensitivity cardiac troponin (a significant biomarker of myocardial injury and necrosis) in whole blood samples [108].

Figure 5 
                     QDs properties and biomedical applications. (a) If excited by a light source, QDs can emit different colours over a wide spectral range. This feature makes them optimal candidate for cellular (on both eukaryotic and prokaryotic cells) and in vivo bioimaging and diagnosis (if conjugated to targeting molecules). (b) QDs present also a cytotoxic effect that can be exploited as antimicrobial, antibiofilm, and anticancer activity. When are photoexcited, QDs generate ROS, bringing cell death. The same effect can be obtained by conjugating killing drugs that can be released in a specific tissue.
Figure 5

QDs properties and biomedical applications. (a) If excited by a light source, QDs can emit different colours over a wide spectral range. This feature makes them optimal candidate for cellular (on both eukaryotic and prokaryotic cells) and in vivo bioimaging and diagnosis (if conjugated to targeting molecules). (b) QDs present also a cytotoxic effect that can be exploited as antimicrobial, antibiofilm, and anticancer activity. When are photoexcited, QDs generate ROS, bringing cell death. The same effect can be obtained by conjugating killing drugs that can be released in a specific tissue.

On the other hand, the outer shell can be functionalized by conjugating different molecules like peptides, proteins, or DNA acting as diagnostic and therapeutic agents for cancer diagnosis, PDT cell labelling, and biosensors [109,110] (Table 1). For instance, conjugation with specific antibodies permits a specific targeting of the tumour so that Ab-modified QDs can be used for the detection of primary tumours (such as ovarian, breast, prostate, and pancreatic cancer), as well as local lymph nodes and detached metastases [111,112] (Figure 5). Karakoçak et al. formulated hyaluronan-conjugated carbon QDs for bioimaging of tumour cells. In vivo studies demonstrated the ability of conjugated hyaluronic acid to recognize CD44 receptors overexpressed by breast cancer tissue and, therefore, selectively detect tumour mass compared to the surrounding tissue [113].

Many studies reported the use of QDs for cellular and targeted drug delivery in cancer treatment as proposed by Ruzycka-Ayoush et al. who synthesized FA-conjugated Ag–In–Zn–S QDs modified with 11-mercaptoundecanoic acid (MUA), l-cysteine, and lipoic acid encapsulated with the anticancer drug doxorubicin. Their studies had demonstrated specific targeting, controlled drug delivery, and imaging on adenocarcinoma human alveolar basal epithelial cells overexpressing folate receptors [114]. Conversely, Qin et al. demonstrated the double effect of carbon quantum dots (CQDs) for imaging and photocatalytic inactivation of cancer cells. After cell internalization, CQDs emitted a yellow fluorescence signal when excited under blue light and simultaneously started to produce ROS, bringing a 40% decrease in relative cell viability [115].

Despite their extraordinary potential as fluorescence probes, QDs present some biomedical application limitations because of their high toxicity for eukaryotic cells: cadmium could cause interferences in DNA repair or stimulate free radical synthesis [116,117]. In this context, recently, cadmium-free QDs (Cd-free QDs) made of indium/palladium were amply used because of their higher biocompatibility [118,119]. For example, fluorescent Ag–In–S/ZnS quantum dots (AIS/ZnS QDs) are Cd-free QDs amply adopted in bioimaging, including tumour draining lymph node imaging [120] and visualization of transplanted adipose tissue-derived stem cells [121].

Overall, the QD application had given many results in vitro systems, especially cellular pathways’ understanding. Anyway, even if QDs were removed from heavy metals, the translation to in vivo systems still presents some limitations due to the RES system action and side effects. QDs’ potential toxicity can be limited by surface modification, like adding PEG [122] or carbohydrates to QDs [123,124]. PEGylation can reduce the liver and spleen uptake decreasing their clearance [125,126]. Therefore, the length and the molecular weight of the PEG and the degree of substitution can modulate the circulation half-life. For example, mPEG-5000-coated QDs circulate longer in mice than mPEG-750-coated QDs that were completely cleared from the bloodstream after 1 h of injection [127].

PEGylation (just as, for instance, the adding of polyethyleneimine and poly-l-lysine) can be also employed to enhance the intrinsic QDs antimicrobial activity. Many studies suggested them as an efficient alternative to traditional antibiotic drugs because they can (1) destroy cell walls/cell membranes, (2) act as oxidizing agents producing ROS, and (3) inhibit cell proliferation by binding with nucleic material (DNA/RNA) [128]. These phenomena are more evident in positively charged carbon quantum dots (PC-CQDs) than in negatively charged CDs, thanks to the more robust interaction with the bacterial cells. The PC-CQDs showed a stronger antibacterial effect on Gram-positive bacteria than on Gram-negative ones, as suggested by Hao et al. who tested its efficiency on different microorganisms such as Staphylococcus aureus, methicillin-resistant S. aureus, Listeria monocytogenes, Enterococcus faecalis, Escherichia coli, Serratia marcescens, Pseudomonas aeruginosa, drug-resistant E. coli, and drug-resistant P. aeruginosa isolated from the clinic [129]. In vivo experiments demonstrated the PC-CQDs ability to support the antibacterial treatment of mixed S. aureus and E. coli infected wounds in rats with low in vivo toxicity. Conversely, Ristic et al. synthesized graphene quantum dots (GQD) NPs that present higher biocompatibility for eukaryotic cells and antibacterial activity in infectious diseases. If photoexcited, GQDs generate ROS due to increased propidium iodide cellular uptake, resulting in the killing of the pathogenic bacteria strains such as methicillin-resistant S. aureus and E. coli as illustrated in Figure 5 [130]. Moreover, QDs optical properties also permit their use for sensing microorganisms, including their detection, Gram type identification, biofilm imaging, and microbial viability assessment [131,132,133,134].

2.3.4 CBN

CBNs are amply employed in many biological applications like bioimaging, drug delivery, tissue engineering, diagnosis, and cancer therapy due to their unique features, including thermal, mechanical, electrical, optical, and structural properties [135,136,137] (Table 1). CBNs include graphene oxide (GO), carbon nanotubes (CNTs), fullerenes, carbon nanohorns, carbon nanodots (CDs), and nanodiamonds (Figure 6). Graphene is a two-dimensional sheet of hexagonally arranged carbon atoms isolated from its three-dimensional parent material, graphite [138]. The oxide form consists of single-atom-thick carbon sheets with carboxylate groups on the periphery, providing pH-dependent negative surface charge and colloidal stability [139]. The basal surfaces contain functional groups of hydroxyl (–OH) that permit the conjugation with many molecules such as antibodies, fluorescence probes, proteins, drugs, or nucleic acids permitting different biomedical applications including tumour therapy, tissue engineering, PTT, and bioimaging (Figure 6) [140]. The basal planes also include unmodified graphene domains that are hydrophobic and capable of steaking (π–π) interactions employed on biological molecules’ adsorption like nucleic acid, including small interfering RNA (siRNA) for drug delivery applications [141]. siRNA gene therapy can also be combined with PTT, as proposed by Yin et al. for pancreatic cancer. They developed PEGylated GO nanosheets conjugated with the tumour targeting molecule FA to co-deliver two siRNA causing apoptosis, proliferation, inhibition, and cell cycle arrest. The synergistic combination of gene silencing and NIR light thermotherapy in vivo mouse model showed tumour volume growth inhibition by >80% [142].

Figure 6 
                     CBNs including graphene NPs and its wrapping structure carbon nanotubes, carbon nanodots, fullerenes, nanodiamonds, and carbon nanohorns. Thanks to the possibility to conjugate fluorescence molecules, antibodies, proteins, drugs, or nucleic acids, CBNs can be employed in tumour therapy, tissue engineering, photothermal therapy, and bioimaging.
Figure 6

CBNs including graphene NPs and its wrapping structure carbon nanotubes, carbon nanodots, fullerenes, nanodiamonds, and carbon nanohorns. Thanks to the possibility to conjugate fluorescence molecules, antibodies, proteins, drugs, or nucleic acids, CBNs can be employed in tumour therapy, tissue engineering, photothermal therapy, and bioimaging.

Another significant feature of graphene NPs is the capability to promote the growth, proliferation, and differentiation of mesenchymal stem cells (MSCs) [143], neural stem cells [144], and induced pluripotent stem cells [145] into tissues of various lineages [146,147,148]. Therefore, its possible employment in tissue engineering and regenerative medicine has generated significant interests, thanks to the further possibility of combining it with other materials like poly-l-lactide (PLLA) [149]. For example, 3 wt% of graphene added to PLLA scaffolds facilitates the differentiation of bone marrow-derived MSCs/stromal cells (BMSCs) and increases the calcium deposition and formation of collagen type I [150].

Furthermore, graphene-specific features, including the electrical conductivity, high electron transfer rate, and capability to immobilize different molecules, make them optimal candidates as biosensors for the electrochemical detection of nucleic acids (through dsDNA), ions and molecules (through enzymes), or cells and microorganisms (through antibodies) [151]. In fact, graphene-based materials are amply used in diagnostics, thanks to their ability to recognize specific molecular biomarkers (i.e., circulating tumour cells, exosomes, circulating nucleic acid, etc.) in liquid biopsies [152]. Recently, the use of graphene for SARS-CoV-2 detection has sparked great interest in the scientific community. For instance, Seo et al. developed a graphene field-effect transistor (FET)-based biosensing device conjugated to anti-SARS-CoV-2 spike protein antibody to detect the virus in human nasopharyngeal swab specimens [153].

CNTs are also amply used in biosensing applications, thanks to their unique features like high AR, stability and thermal and electrical conductivity, strong mechanical strengths, and fast electron transfer rate [154,155]. They originate by wrapping graphene into a cylinder structure forming a tubular structure of 1–2 nm in diameter: the rolled sheets can be single (single-walled CNTs- SWCNTs), double (double-walled CNTs DWNTs), or more than two (multi-walled CNTs – MWCNTs) [136,156]. Their limited solubility on all solvents generates toxicity problems that can be solved by chemical modification with peptides, proteins, nucleic acid, and therapeutic molecules that can increase the cellular uptake and drug release when used as DDS [136,157,158]. Su et al. conjugated iRGD-polyethyleneimine (PEI) and candesartan (CD) to develop MWCNTs targeting the tumour endothelium and lung cancer cells (by recognition of αvβ3-integrin and AT1R). In addition, plasmid AT2 (pAT2) was assembled to form iRGD-PEI-MWNT-SS-CD/pAT2 complexes. Co-delivery of CD and pAT2 synergistically inhibited angiogenesis by downregulating vascular endothelial growth factor and inducing tumour growth suppression in A549 xenograft nude mice [159]. Moreover, the nature of CNTs makes them ideal elements for tissue engineering; for instance, Vaithilingam et al. introduced multi-walled CNTs to 3D scaffolds to stimulate human pluripotent stem cells to differentiate into cardiomyocytes and modulate their behaviour [160].

Similar to SWCNTs, carbon nanohorns (CNHs) present a conical hollow configuration constituted by sp2-hybridized carbon atoms highly resistant to oxidation. Although easily synthesised and functionalised, CNHs present some limitations during biomedical applications because of their aggregation into spherical clusters [161]. Some strategies were developed to increase CHHs polydispersity such as the addition of potassium naphthalenide [162,163]. On the other hand, CDs present water-dispersible properties in addiction to fluorescent ones. In fact, they are fluorescent carbon nanostructures with a size less than 10 nm amply employed in biological imaging. The modification with nitrogen, sulphur, and phosphorus can improve the bioluminescence features, while the conjugation with drugs or targeting molecules increases their interest in biomedical applications, especially in cancer theranostics [164]. For instance, when CDs are irradiated by NIR light, ROS that selectively kills tumour cells is generated (NIR light-irradiated PDT) [165,166]. Notwithstanding the interesting properties, in vivo CD applications present some limits due to the concentration-related toxicity for the blood compound: a concentration higher than 0.1 mg/mL induces red blood cells lysis, complement activation, and platelet-mediated coagulation stimulation [167].

Fullerens consist of hexagonal and pentagonal rings than confer the typical curvature to generate hollow spheres (named buckyballs), ellipsoids, or tubes. Buckminsterfullerene (C 60) is the most common fulleren amply adopted in biological applications, thanks to its electrochemical, physical, and photographic features [168]. Although fullerens are soluble only in organic solvents, their structure permits the addition of hydroxyl groups (fullerenols) that increase their solubility in water and also the conjugation with a huge range of biological molecules for biomedical applicants in tissue engineering and photothermal and cancer therapy and also antibacterial and antiviral activities [169,170]. Therefore, the first water-soluble fullerene form was developed in 1993 by Wudl and coworkers who studied its action as an inhibitor of the HIV-1 protease (HIV-1 PR) [171,172].

Conversely, the capabilities to bind a high amount of drugs and to easily enter into the cells make nanodiamonds (NDs optimal candidates for drug delivery in biological systems. They present a truncated octahedral structure, faceted or with a rounded shape, often modified by additional functional groups (like hydroxyl, carbonyl, carboxyl, anhydrides, and lactones) that limit aggregation, improving biocompatibility [173]. Their good stability, biocompatibility, and good optical properties permit them to be used as biological markers for bioimaging investigation, as DDS for cancer or metabolic diseases and as fillers of biocomposite scaffolds for tissue engineering. NDs present a low osteogenic differentiation capability that can be increased by conjugating chemical compounds like icariin, as proposed by Choi et al. [174,175].

2.3.5 MSNs

MSNs are porous spherical particles of 50–300 nm constituted by a polymeric structure of siloxane (–Si–O–Si–O–) rich in silanol (Si–OH) groups on their surface that can be modified by conjugation with biological molecules to obtain multifunctional nanoconjugates [176]. The porous structure permits conjugating molecules (e.g., drugs, fluorescence probes, targeting molecules, nucleic acid) in the inner part and surface (Figure 7). The controlled chemical synthesis permits to regulate their morphology, pore distribution, size, and biodegradability [177]. Parameters like pH, surfactant, silica precursor, and temperature can modulate NP size and shape that play a crucial role in cellular uptake, immune escape, and elimination rate [178]. For example, long-rod MSNs are captured by the spleen with a low elimination rate, while the short-rod ones are localized in the liver [179].

Figure 7 
                     Mesoporous silica nanoparticles (MSNs) functionalization and biomedical applications. Biological molecules (e.g., fluorescent probes, drugs, DNA, and RNA) can be encapsulated in the pores or conjugated to the surface of MSNs just as targeting molecules and PEG. Fictionalized MSNs are employed not only in tissue engineering, bioimaging, tumour therapy, and vaccines but also in antibacterial and antibiofilm treatment.
Figure 7

Mesoporous silica nanoparticles (MSNs) functionalization and biomedical applications. Biological molecules (e.g., fluorescent probes, drugs, DNA, and RNA) can be encapsulated in the pores or conjugated to the surface of MSNs just as targeting molecules and PEG. Fictionalized MSNs are employed not only in tissue engineering, bioimaging, tumour therapy, and vaccines but also in antibacterial and antibiofilm treatment.

Generally, MSNs are highly biocompatible because they degrade to silicic acid naturally present in body fluids and connective tissue, such as hair, nails, bone, skin, and tendons, and are rapidly eliminated through urine [180,181,182] (Table 1). In this context, MSN circulating time can be highly regulated: surface modification like the PEGylation can prolong their permanence in the bloodstream. At the same time, the increase of their pore size or the addition of metal ions can accelerate their elimination from the body [183,184]. By analysing the MSNs biodistribution and clearance kinetics in healthy rats, Dogra et al. showed that big NPs (142 nm) presented lower accumulation in the liver and spleen compared to the smaller ones (32 nm) [185].

Furthermore, the significant amount of pores and channels that confers a high surface-to-volume ratio permits to accommodate a large number of biological molecules, including therapeutic agents or drugs that make them optimal candidates as drug delivery carriers for tumour therapy as suggested by Duo et al. [186,187] (Table 1, Figure 7). They synthesized doxorubicin-loaded MSNs coated with polydopamine (PDA) to obtain a pH-sensitive drug release. Furthermore, NPs were functionalized with PEG to increase the stability and biocompatibility of nanosystems. In vitro and in vivo analysess in the breast cancer model had suggested a higher cellular uptake and a controlled drug release with an improved anticancer activity than a free drug [187].

Silica can also reduce the toxicity of other NPs containing, for example, heavy metals like gadolinium (Gd), which is typically used as a contrast agent in MRI [188]. In the same manner, their functionalization can occur for other bio-imaging applications such as optical imaging, PET, and CT [189,190,191]. Recently, MSNs are also employed in PA imaging, thanks to the possibility to encapsulate the PA contrast agent ICG that produces heat under NIR irradiation [192]. Due to the heat-induced thermoelastic change, the tissue generates US waves detected by US transducers [193].

Zhang et al., to limit the aggregation and increase the drug loading to AuNPs, synthesized a mesoporous silica shell-coated AuNPs conjugated with doxorubicin as an anticancer agent [194,195]. At the same time, Ramasamy et al. developed AuNPs coated with silica to deliver the antibiofilm agent cinnamaldehyde to eradicate bacteria [196]. The MSN structure is ideal for loading, protecting, and transporting antimicrobial molecules to the target bacteria and/or biofilm and releasing them in a stimuli-responsive way, as reported by Yang et al. [197,198]. They described the in vitro and in vivo antibacterial efficiency of MSNs carrying gentamicin and bacteria-targeting peptide ubiquicidin coated by bacterial toxin responsive lipid bilayer surface shell. Their data demonstrated the S. aureus growth inhibition and the downregulation of inflammation-related gene expression in infected preosteoblast or macrophage [198].

Moreover, MSNs could be incorporated on scaffolds for tissue engineering, especially bone tissue engineering: MSNs loaded with bioactive factors can be combined with scaffolds to improve repair efficacy [199] (Figure 7). MSNs can release Si ions that can influence stem cell behaviour, especially in gene expression in differentiation and osteogenesis [200].

Moreover, mesoporous silica NPs present an interesting potential as a vaccine adjuvant, as suggested by Oliveira et al. who had investigated the MSNs vector efficiency against the parasite Schistosoma mansoni. In particular, they had developed MSNs associated with SWAP (soluble worm antigenic preparation) to test their higher immunization activity compared to a conventional immunization system (SWAP-associated aluminium salt) [201]. In this scenario, MSNs pore sizes play a key role in the presentation of peptide-major histocompatibility complex (MHC I) complexes to CD8+ T cells, as suggested by Hong et al. They had shown that the association of ovalbumin (OVA) tumour antigen with MSNs enhanced both antibody and T cell responses and, in particular, the large-pore MSNs had established the strongest antitumor effects and immune response. Nanosystems, indeed, facilitated OVA escape from lysosomal degradation for MHC I restricted [202].

2.3.6 Organic NPs

In the last years, many researchers have focused their studies on the possible use of organic (ONPs) in different sectors, especially biomedical ones. Indeed, the organic nature of these systems highly reduces their toxicity and, therefore, side effects. There are different types of ONPs depending on their composition and structure like liposomes and micelles, dendrimers, polymeric NPs, and nanogels.

2.3.7 Lipid-based nanoparticles

Lipid-based nanoparticles (LNPs) are the most common FDA-approved NP type due to their features like the easy synthesis process, biocompatibility, and capability to deliver molecules of different nature (Table 2, Figure 8) [203]. It is a vast family of different types of nanosystems, including liposomes, micelles, exosomes, niosomes, solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), lipid hybrid nanoparticles (LPHNPs), and lipid calcium phosphate nanoparticles (LCPNPs) that differ for the lipid composition and are employed in many applications like the “Trojan horse liposome” (THL) technology, tumour therapy, tissue engineering, and vaccines (Figure 8).

Figure 8 
                     LNPs including liposomes, micelles, exosomes, niosomes, solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), lipid hybrid nanoparticles (LPHNPs), and LCPNPs and their application in biomedical research. Liposomes can encapsulate hydrophilic molecules in the inner core, while the hydrophobic ones are associated with the phospholipid membranes; micelles can link lipophilic molecules in the hydrophobic core; LNPs can encapsulate nucleic acid like RNA. SLNs are composed by solid lipids, while NLCs by both solid and liquids. Thanks to their characteristics, LNPs are amply studied in Trojan Horse Liposome (THL) technology for NDs, cancer therapy, tissue engineering, and in vaccine formulations.
Figure 8

LNPs including liposomes, micelles, exosomes, niosomes, solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), lipid hybrid nanoparticles (LPHNPs), and LCPNPs and their application in biomedical research. Liposomes can encapsulate hydrophilic molecules in the inner core, while the hydrophobic ones are associated with the phospholipid membranes; micelles can link lipophilic molecules in the hydrophobic core; LNPs can encapsulate nucleic acid like RNA. SLNs are composed by solid lipids, while NLCs by both solid and liquids. Thanks to their characteristics, LNPs are amply studied in Trojan Horse Liposome (THL) technology for NDs, cancer therapy, tissue engineering, and in vaccine formulations.

Liposomes, an early version of LNPs, are vesicles constituted by a self-assembled phospholipid bilayer that assumes a spherical shape delimiting an aqueous core of 50–1,000 nm diameter [204]. Depending on the bilayer’s number, it is possibly classified into small unilamellar vesicles (SUVs), large unilamellar vesicles (LUVs), giant unilamellar vesicles (GUVs), multivesicular vesicles (MVs), and multilamellar vesicles (MLVs), in which the layers are separated by aqueous spaces [205]. This unique structure permits to load of both hydrophilic and hydrophobic molecules: the hydrophilic ones are localized in the inner core or between the bilayers; in contrast, the hydrophobic molecules are associated with the phospholipid membranes [206]. This leads to multidrug loading and, consequently, a sequential drug release from the two different compartments. They are primarily employed in the DDS because they can fuse with the plasmatic membrane and release the drug inside the cell [207]. Furthermore, PS inclusion in the liposomes permits a light-induced cargo release (light-induced liposome technology) [208]. Instead, micelles are characterized by a single lipid layer that defines a spherical structure with a hydrophobic core, fundamental for transporting lipophilic molecules like many antitumor drugs [209].

Thanks to their nature, liposomes and micelles are non-toxic, biocompatible, biodegradable, and non-immunogenic (Table 1). Moreover, their chemical–physical properties can be accurately modified by mixing different lipids molecules and changing the superficial charge, size, and functionalization [210].

Even if they have a good distribution in the organism, they present some disadvantages like low solubility and half-life, the possibility to leak the loaded drugs, especially when they occasionally change lipid components with high- or low-density lipoprotein (HDL and LDL respectively) and thus modify their size and composition, and also their accumulation into the tissues [211]. In addition, surface modification like PEGylation can elongate the circulating time in the bloodstream, while the addition of targeting molecules can improve targeted delivery [212]. In this context, PEGylated liposomes are amply studied in the THL technology for transvascular non-viral gene therapy of the brain. Cationic liposomes of THL carrying non-viral gene expression plasmids are functionalized with specific antibodies able to recognize antigens on the BBB and, in this way, permit its crossing [213,214]. Cationic liposomes’ encapsulation of genetic material limits their degradation from ubiquitous nucleases. The exogenous gene is expressed within 1 day of a single intravenous administration, as Jiang et al. had demonstrated; indeed, THL treatment reduced tissue inclusion bodies in the brain and peripheral organs [215,216]. This technique is also applied in NDs like Parkinson’s disease [217,218] and tumour therapy, as described by Zhang et al. They reported that monoclonal antibody-targeted THLs carrying a siRNA knocking down the EGFR were capable of increasing the survival time of mice with intracranial brain cancer [219,220]. Liposomes and micelles are the most primarily studied vector for drug targeting macrophages in treating diseases like salmonellosis, leishmaniosis, tuberculosis, rheumatoid arthritis, and cancer [221,222,223,224,225,226]. Furthermore, liposomes can mimic pathogens’ features, inducing humoral and cellular immune responses, thanks to their capability to present antigens to antigen-presenting cells. Therefore, they can be optimal candidates as vaccines. Depending on the saturation grade of the lipids, they could induce a Th2 (if they are unsaturated) or Th1 (if they are saturated) response [227]. For example, Huang et al. reported the improved efficiency of the Pfs230 malaria transmission-blocking antigen candidate when it was loaded on liposomes containing cobalt–porphyrin–phospholipid and the synthetic monophosphoryl lipid A (PHAD). They had shown an increase in anti-Pfs230C1 IgG response in mice after 250 days, the inhibition of parasite transmission, and the immunization of rabbits [228].

Cationic lipid NPs are amply employed as nucleic acid-based vaccines (mRNA-carrying LPNs). They differ from liposomes because, inside the core of the particles, they present micellar structures in which nucleic acid, including mRNA, can be protected from enzymatic degradation. Generally, they consist of cationic or ionisable lipids that interact with negatively charged nucleic acids and helper lipids (like phospholipids, cholesterol, and/or PEGylated lipids) that increase their stability and promote cellular uptake and consequently the nucleic acid delivery [229]. Espeseth et al. demonstrated a higher cellular immune response of mRNA/lipid NPs than to the protein-based vaccine. They tested lipid NP-encapsulated mRNA vaccine encoding respiratory syncytial virus F protein on rodent animals, highlighting the total absence of vaccine-enhanced respiratory disease that generally occurs after protein immunization [230]. Furthermore, two of the most used COVID-19 vaccines (mRNA 1273 and BNT162b2) were developed using LPNs as a vector to deliver mRNA encoding for a full-length, perfusion stabilized spike (S) protein of SARS-CoV-2 [231,232]. After intramuscular injection, LNPs are internalized by host cells in which the mRNA is release, translated into the S protein that is exposed in the cell membrane to immune system cells [233].

Exosomes are small extracellular vesicles constituted by a phospholipid-bilayer membrane that enclose various molecules depending on the type of mammalian cell that secreted them [234]. Based on this, they can be used as potential biomarkers for early detection and monitoring of the progression of cancer or other pathologies like neurodegenerative ones [235,236]. In the same manner, exosomes can deliver drugs or other biomolecules acting as DDS [237]. Generally, they are isolated by ultracentrifugation and filtration, even if these techniques limit the purity and can partially destroy the NPs. Other methodologies were optimized like size-exclusion chromatography (SEC) or polymer precipitation, but the isolation protocol could be optimized to obtain more pure exosomes [238]. Because of their nature, exosomes are highly biocompatible, present innate stability and low immunogenicity, and can easily enter cells and tissues. Conversely, they present some disadvantages like the limited drug loading, the rapid clearance by the reticuloendothelial system, and the difficulty in specific targeting that depends on the homing sources. For example, tumour cell-derived exosomes can target metastatic and cancer cells, while macrophage-derived exosomes can cross the BBB [239]. Despite their great potential, further improvements need to be carried out for their use in clinical.

Niosomes present also a bilayer structure that, in this case, is formed by self-association of non-ionic surfactants and lipids such as cholesterol in an aqueous phase. Depending on the type of components, the size, the surface charge, and the number of lamellae, niosomes can be classified into three groups: SUV (10–100 nm), LUV (100–3,000 nm), and MLV, where more than one bilayer is present [240]. As liposomes, niosomes are able to load both hydrophobic and hydrophilic drugs. However, they present some advantages compared to liposomes [241]. The uncharged single-chain surfactants confer higher stability in contrast with neutral or changed double-chain phospholipids of the liposomes. Furthermore, the lower cholesterol concentration in niosomes bring to higher drug loading and longer self-life [242,243]. Thanks to their nature, different administration methods can be adopted, like parental and oral. It has been amply reported that niosomes can enhance the oral bioavailability of different drugs, including paclitaxel, griseofulvin, carvedilol, and nateglinide [244,245]. Conversely, the nasal administration permits brain uptake through the direct nose-to-brain pathway for the neurodegenerative or glioma therapies [246], while the possibility to use with the stratum corneum permitting drug penetration in dermal tissue (dermal administration) [247]. Finally, niosomes could prolong residence time in the cornea and could be incorporated in foaming gels, creams, and ointments for ocular administration [248].

In 1991, SLNs were developed by combining features of the classical NPs (metal or polymeric NPs at that time) and the lipid-based liposomes [249,250]. SLNs are generally spherical with a diameter between 50 and 1,000 nm and are constituted by lipids (including fatty acids, triglycerides, steroids, and waxes) and surfactants as stabilizing agents [251]. Surfactants, like lecithin phospholipids, Tween, Span, sodium cholate, polysorbates, poloxamers, and bile acids, increase the SLN stability by limiting the interfacial tension and the aggregation of the NPs, especially if used as a mixture [252]. SLNs present not only good biocompatibility and biodegradability as well as other lipid NPs but also easy sterilization and increased stability due to the rigid core lipid matrix, permitting the encapsulation of both lipophilic and hydrophilic drugs [253,254]. Differently from liposomes, they are synthesized by organic solvent-free methods (through high-pressure homogenization technique or micro-emulsification processes), allowing large-scale manufacture at a low cost. Another significant advantage of SLNs is the wide range of administration routes, including oral [255], intravenous [256], pulmonary [257], ocular [258], and transdermal [259].

In contrast to other NPs, the lipid nature of SLNs makes them an optimal candidate for oral administration that is feasible as aqueous dispersions or in the dosage forms of capsules, tablets, and pellets. Indeed, the solid lipid matrix of SLNs can enhance their bioavailability by protecting encapsulated drugs from chemical and enzymatic activity in the gastrointestinal tract (GIT) [260]. SLNs exploit the oral drug delivery via intestinal lymphatic transport, reaching the lymphatic system through microfold cells (M cells) [261,262]. Moreover, SLNs can be surface modified by adding heparin, albumin, PEG, and polysaccharides like chitosan to prevent the high drug release due to the low pH of the stomach [263]. The bioavailability could also be increased by conjugating P-gp inhibitors as proposed by Shah et al. They synthetized SLNs as carriers for the anti-diabetic drug linagliptin (LGP) and functionalized them with Tween 80 as a P-gp inhibitor. Data reported that this formulation increased the oral bioavailability from 29.5% (LGP alone) to 300%, carrying to a reduction in glucose levels compared to the drug alone [264]. Oral administration of NPs can also be adopted for the treatment of other pathologies including, cardiovascular diseases [265], cancer [266], and neuropathologies, thanks to their ability to cross BBB [267]. For example, Shen et al. developed doxorubicin and SPION-loaded SLNs for a chemo/magnetothermal colon tumour therapy. The active targeting was obtained by adding folate (FA), and the entire complex was covered and protected by a dextran shell. Once arrived in the small intestine, the shell is degraded by dextranase, bringing the FA exposure and the NPs colon tumour cellular uptake. In this manner, the drug stability was increased not only by the SLNs themselves but also by the possibility to functionalize them (e.g., dexatran). Compared with the drug alone, the anticancer efficiency of the nanocomplex was significantly higher, bringing to the effective inhibition of primary colon tumour and metastasis [268].

Despite several advantages, SLNs have drawbacks including poor drug-loading capacity and drug expulsion during long-term storage because of lipid crystallization [252]. Therefore, NLCs were developed to overcome these limits. NLCs consist of both solid and liquid lipids that reduce the crystallinity of the lipid core, increasing the drug loading and long-term stability. Based on this, NLCs can be classified into imperfect, amorphous, and multiple types [269]. Imperfect type is constituted by different lipids, like glycerides carrying fatty acids of different lengths and saturation, engendering imperfections in the crystal order. The amount of loaded drug depends on the imperfections grade (a higher imperfection grade corresponds to a higher drug loading). In the amorphous NLCs, the solid and liquid lipids are mixed to form an amorphous unstructured non-crystalline matrix. Multiple NLSs are oil-in-fat-water-carriers (O/F/W) in which oil nano-compartments, hosting drugs, are surrounded by solid lipids acting as a barrier to protect drugs and control their release [270].

As reported earlier for SLPs, NLCs can also be administered in different ways like oral, ocular, transdermal, intranasal, intravenous, and pulmonary routes [271]. The intranasal (IN) route is a non-invasive approach amply adopted for nose-to-brain delivery using NLCs as vectors to increase the drug bioavailability. Three pathways can be involved in this process: the systemic ones in which, after reaching the systemic circulation, drugs can cross the BBB; the olfactory pathway, where drugs go into the brain tissue via the olfactory bulb; and the trigeminal way where the drugs arrive in the nervous system through trigeminal nerve [272]. Since the drug absorption can be limited by the mucociliary clearance in the nasal cavity, NLCs can be superficially modified with cationic polysaccharides, such as chitosan, which were able to interact with the negative charge of the mucosa. For instance, Singh et al. developed chitosan-coated NLC loaded with the antipsychotic drug asenapine for intranasal delivery [273]. Their data showed that NLCs as carriers increased the systemic and brain bioavailability by 2.3- and 4-folds compared to drugs alone. Pai and Vavia also adopted the chitosan coating to make mucoadhesive on the surface of NLCs [274]. They used NLCs as a vector for the antineoplastic eye drug etoposide for ocular administration, showing NP localization around the ocular surface confirmed by the high drug concentration. Furthermore, many assays have demonstrated non-toxicity and the absence of ocular irritancy.

On the other hand, all the mentioned SLNs and NLCs properties make them optimal also for cosmetic and pharmaceutics applications [249]. In particular, many studies reported the NLCs’ ability to improve wound healing by enhancing drug penetration and maintaining skin hydration and moisture [275,276]. Starting from this, Chato-Astrain et al. combined the NLCs properties to bioartificial human dermis substitute for tissue engineering application [277]. In particular, they functionalized fibrin-agarose biomaterials with antimicrobial-loaded NLCs to treat infected wounds, typical of severe burns. Their data showed that the antibacterial effect was directly proportional to the NLC amount and that these NPs were able to improve some important biomechanical properties of the artificial construct.

The special properties of LNPs can be combined with those of polymeric nanoparticles (PNPs) to develop the LPHNPs. Lipid component increases the biocompatibility, stability, and permeation of drug across the cell membrane, while the nature of PNPs permits to regulate the drug loading and release [278]. Taken together, these characteristics make LPHNPs an optimal candidate for DDS with excellent drug release kinetics, high encapsulation efficiency, and low toxicity. LPHNPs are constituted by (1) a hydrophobic polymeric core, in which the therapeutic drugs are encapsulated, surrounded by (2) a phospholipid monolayer that makes more biocompatible the polymeric core, and (3) an outer lipid-PEG layer that confers stability increasing the in vivo blood circulation time of the LPHNPs. Based on their structure, LPHNPs can be classified into [279,280]:

  1. Monolithic hybrid NPs (random dispersion of lipids in a polymeric matrix);

  2. Polymer-core lipid-shell NPs (polymeric core surrounding by lipid shell);

  3. Hollow core lipid–polymer–lipid NPs (hollow aqueous core surrounding by cationic lipids followed by polymer layer and a neutral lipids layer);

  4. Erythrocyte membrane-coated LPHNPs (polymeric core surrounding by a cell-derived membrane);

  5. Polymer-caged liposomes (liposomes coated by an external polymer layer).

By integrating two different kinds of NPs, LPHNPs can be employed in a huge range of biomedical applications including cancer therapy because the polymeric part permits the conjugation with hydrophobic chemotherapeutic drugs and the lipid part protects them from early degradation and permit the association with targeting molecules, like FA [281]. For instance, Khan et al. developed FA-conjugated chitosan LPNPs and showed an increase in their breast cancer cellular uptake and efficiency compared to non-targeted NPs [282]. Many studies also reported the use of LPHNPs for gene therapy, thanks to their large-DNA incorporation ability [283,284]. Cationic LPHNPs are employed also for siRNA delivery as proposed by D’Angelo et al. who investigated the use of PLGA and dipalmitoylphosphatidylcholine-based LPHNPs for siRNA pulmonary delivery for cystic fibrosis (CF) therapy. Their data highlighted the efficiency of these nanosystems to be internalized inside the airway epithelial barrier and are able to transport siRNA and release it reducing the relative protein expression [285]. Despite good in vitro results amply reported in the literature, few data of in vivo data can be found, suggesting that more investigation into the use of LPHNPs for gene therapy needs to be carried out.

Conversely, it is amply reported in the literature on the use of LCPNPs for gene delivery. Calcium ions are able to complex with nucleic acids protecting them from the serum nucleases, and calcium phosphate (CaP) is dissolvable at a pH of 4–5 [286]. This pH is typical of the lysosomes in which CaPs can go after cellular internalization, permitting the drug release. The coating of CaP with cationic lipid compound form LCPNPs, and this increases their stability and the siRNA delivery, as suggested by Tang et al. [287]. The NP’s features, including size and drug loading, depend on the Ca2+/ PO 4 3 ratio. Due to the pivotal role of the calcium and phosphate ions in bone resorption and deposition regulation (healthy bone cells communicate through Ca2+ and PO 4 3 channels), LCPNPs are amply involved in bone tissue regeneration [288]. In fact, many studies demonstrated the use of this type of NPs as a delivery system of factors for bone application, including antibiotics, anti-inflammatory agents, and growth factors (GFs) [289]. Among these, bone morphogenetic factors (BMPs) are the most used GFs. Some studies reported also the delivery of small plasmid DNA carrying genes like BMP, microRNA, or siRNA [290].

2.3.8 Dendrimers

Dendrimers are nanovectors with a spherical shape constituted by polymeric macromolecules capable of self-assembling. They present three different parts: a central hydrophobic core available for the encapsulation of drug molecules; ramification repeated units named “dendrons” that determinate the generation of the dendrimer and its globular structure; hydrophilic functional groups at the outer side that can be conjugated with specific molecules for the complex formation or other functionalizations, as shown in Figure 9 [291]. Their synthesis is based on the polymerization process of the ramification units from the surface to the core (convergent synthesis) or vice versa (divergent synthesis), and it can be patterned to control the drug release [292]. Thanks to this globular shape, they present a high drug loading ability through both covalent and noncovalent bonds, low polydispersity, reproducible pharmacodynamics, and pharmacokinetics (Table 1). Moreover, the positive charge on their surface due to amino groups’ presence permits the interaction with nucleic acids, like siRNA or small DNA and the association with cell membranes with cellular uptake [293,294] (Figure 9).

Figure 9 
                     Dendrimers features and biomedical applications. Thanks to their ability as gene and drug delivery systems, dendrimers are amply employed in cancer treatment, tissue engineering, and antimicrobial (bacterial and viral) activity.
Figure 9

Dendrimers features and biomedical applications. Thanks to their ability as gene and drug delivery systems, dendrimers are amply employed in cancer treatment, tissue engineering, and antimicrobial (bacterial and viral) activity.

On the contrary, the cationic charge makes them toxic for both prokaryotic and eukaryotic cells so that they are rapidly eliminated from the bloodstream by the mononuclear phagocyte system [295,296]. Therefore, generally, they are modified with molecules like PEG that are able to shield the positive charge, improving circulation time and making them more biocompatible even if it depends on PEG molecular weight, degree of PEGylation, and tested cell lines [297]. The antibacterial activity is related to the ratio of surface cationic charge to hydrophobicity. It is probably mediated by disrupting the bacterial outer and inner membranes due to positive charges of terminal amino groups, as Kannan et al. have suggested [298]. Furthermore, Valikala et al. demonstrated this effect against both Gram-positive and Gram-negative bacteria even after PEGylation [299]. Conversely, their functionalization with anionic groups, such as acid or sulfonate residues, permits limiting the eukaryotic cell toxicity and determinates artificial mimics of the anionic cell surfaces to exploit an antiviral function. Based on the virus–cell interaction depending on the binding to the cell membrane’s sulphated residues, dendrimers can compete with cells for binding of virus and, therefore, stop the infection [300,301].

There are over 100 families of dendrimers depending on their functionalization moieties and on the initiator cores (carbon, nitrogen, and phosphorus). They are classified as polyamidoamine (PAMAM), polypropylenimine (PPI), carbosilane (CBS), poly-l-lysine (PLL), and phosphorus dendrimers [302]. PAMAM and PPI are amply employed in pharmaceutical sciences and biomedical engineering, thanks to the possibility to work as a delivery system and overcome drug resistance. In this context, they are amply used to treat infectious diseases like malaria, leishmania, schistosomiasis, toxoplasmosis, HIV, meningitis, hepatitis, and herpes and especially in tumour therapy [303,304,305,306,307,308] (Table 1 and Figure 9). Easy surface and core modifications with DNA, siRNA, plasmids, peptides, antibodies, or drugs make them optimal candidates for drug delivery, especially for brain tumours like glioma, because they can cross the BBB and deliver biological molecules in a controlled way like that proposed by Lu et al. They had formulated PAMAM dendrimers conjugated with the PEG, the Arg-Gly-Asp (RGD) tripeptide for tumour targeting, and the anticancer drug arsenic trioxide (ATO): the use of the dendrimer vector permits the drug crossing of the BBB, enhancing its antitumor effect [309,310]. The capability to incorporate many biological molecules and the ramified structure are optimal tissue engineering applications [311]. In this context, dendrimers can act as a polymerizing agent in hydrogel scaffolds and simultaneously release growth factors in a controlled manner. In particular, they are amply used in bone tissue engineering, as reported by Oliveira et al. who showed an increase in the ectopic early osteogenic differentiation of rat bone marrow stromal cells in osteoblasts in hydroxyapatite (HA) and SPCL (starch–polycaprolactone) scaffolds in the presence of Dex-loaded CMCht/PAMAM dendrimer NPs (dexamethasone-loaded carboxymethylchitosan/poly(amidoamine) dendrimer) [16]. Furthermore, the presence of PMAM dendrimers in hydrogel scaffolds can be related to its specific mechanical properties, as reported by Pistone et al. They developed a chitosan–dendrimer–HA hydrogel doped with an anti-inflammatory drug (ketoprofen). They identified a correlation between dendrimer molecular weight and chitosan–HA matrix’s rheological properties, investigating its drug release kinetic [312].

2.3.9 Polymer nanoparticles

PNPs present a size ranging from 1 to 1,000 nm and are classified into nanocapsules and nanospheres [313]. The first one is constituted by an oily core in which the drug or the biological molecule is retained, surrounded by a polymeric shell that controls the drug release mechanism. Conversely, nanospheres present a polymeric matrix able to encapsulate the drug: in this way, the drug is carried both in the inner and outer parts. Based on the polymer source of origin, it is possible to individuate natural and synthetic polymers [314]. Natural polymers include sodium alginate [315], albumin [316], chitosan [317], polypeptides [318], cellulose [319], inulin [320], and gelatin [321]. Conversely, some examples of the synthetic ones are PLGAs [322,323], polyglycolides [324], poly(malic acid) [325], poly(methyl methacrylate) [326], polyacrylamide [327], poly(N-vinyl pyrrolidone) (PVP) [328], polyorthoesters [329], poly(methacrylic acid)[330], and poly-l-lactide [331,332] that can be modified with particles like HA for bone tissue engineering applications [333]. Polymers can also cross-link to form nanogels that are able to encapsulate theranostic cargos and conjugate targeting molecules to obtain a specific drug release profile [334].

Generally, PNPs can be formulated by direct monomeric polymerization or dispersion of pre-existing polymers. The polymerization process can be obtained from monomers by different preparation techniques like emulsion, miniemulsion or microemulsion, interfacial, controlled/living radical (C/LRP). At the same time, polymer dispersion can be developed by solvent evaporation, nanoprecipitation, salting out, dialysis, and supercritical fluid technology [335]. Conversely, amphiphilic copolymers with distinct hydrophobic and hydrophilic segments can self-assemble to form micelles in an aqueous solution, wherein water-insoluble elements form the core hydrophilic components form the corona [336]. These features permit a huge biomedical application range, so that many PNPs are currently FDA approved and used as therapeutic agents (Table 2).

Polymers present many functional groups permitting the conjugation with biological molecules involved in specific targeting or a controlled drug release mechanism as shown in Figure 10. Adamo et al., for example, designed PVP nanogels bringing both FA for a specific tumour targeting and the pro-apoptotic Bcl-2 siRNA through a redox-sensitive linker. Their data suggested a selective death induction only on cancer cells [337,338]. Their biodegradability and biocompatibility make them optimum candidates for the treatment of cancer and neurodegenerative disorders and cardiovascular diseases[339] (Table 1, Figure 10). By investigating the use of NPs for Alzheimer’s disease (AD), Carradori et al. had demonstrated the therapeutic efficacy of PNPs conjugated with the antibody against Aβ1–42 peptide to reduce soluble forms of Aβ and rescue memory in AD mice [340]. Conversely, Tan et al. had developed PNPs to encapsulate apomorphine (AMP) drug commonly used in Parkinson’s disease. In this manner, AMP was able to cross the BBB and was protected by oxidation preventing toxic form formation [341]. Moreover, many studies suggest their application for ischemic protection since passive targeting may be doable because the blood–brain barrier’s permeability increases upon ischemia [342]. Zamanlu et al., for example, had formulated PEGylated PLGA NPs conjugated with the tissue plasminogen activator for the treatment of ischemic stroke: circulating time and thrombolytic activity were increased by association with the nanocomplex [343]. PLGA and the other PNPs can be functionalized to obtain a sustained, spatial, and temporally controlled delivery of growth factors (GFs) involved in cell growth and differentiation. GFs can be loaded, together with cells, into solid scaffolds or hydrogels to elaborate 3D structures for tissue engineering, as proposed by Sokolova et al. [344]. They encapsulated DNA-loaded calcium phosphate NPs into PLGA/hydroxylapatite (nHAP) composite scaffolds for bone tissue engineering. HeLa cells’ test suggested a higher cell attachment, higher cell viability, and good gene transfection activity. In this context, Park et al. developed a co-delivery system of a gene and a protein into cells [345]. They synthesized Runt-related transcription factor 2 (RUNX2) protein-loaded PLGA NPs conjugated with bone morphogenetic protein 2 (BMP-2) plasmid DNA to enhance osteogenesis. In vitro analysis of human mesenchymal stem cells (hMSCs) treated with these NPs showed increased differentiation grade. Furthermore, in vivo experiments demonstrated NPs’ ability to enhance the osteogenesis of the hMSCs.

Figure 10 
                     PNPs and their application in biomedical research. PNPs can be conjugated with targeting molecules (specific targeting); with biological molecules including drugs and RNA (drug and gene delivery) and fluorescence probes (bioimaging). Furthermore, PNPs are amply used also in tissue engineering and vaccines.
Figure 10

PNPs and their application in biomedical research. PNPs can be conjugated with targeting molecules (specific targeting); with biological molecules including drugs and RNA (drug and gene delivery) and fluorescence probes (bioimaging). Furthermore, PNPs are amply used also in tissue engineering and vaccines.

Furthermore, many studies have been dedicated to PNPs application as a controlled-release vaccine delivery system (Table 1 and Figure 10). Some parameters like surface charge and antigen loading can influence the immune responses, as suggested by Gu et al. who had tested PLGA NPs positively or negatively charged conjugated to ovalbumin (OVA) by adsorption or encapsulation. They reported that the negative charge facilitated the cytoplasmic antigen delivery by inducing the activation of DCs in lymph nodes 5 days after the primary vaccination. Conversely, when the antigen was encapsulated, more potent and long-term antigen-specific antibody responses were registered, compared to those of antigen-adsorbed NPs [346]. Moreover, NP immunogenicity can be improved by adding adjuvants like chitosan or glycol chitosan. In vivo studies suggested that glycol chitosan induces significantly higher systemic and mucosal immune responses than only chitosan or NPs alone [347].

3 Conclusions

With a particular focus on nanomedicine, nanotechnology provides an entirely new concept and new approaches in vast fields of modern science and medicine. The small size of NPs confers them unique properties because they are subject to physical laws in the middle between classical and quantum physics. In this context, the NPs’ project plays a key role because the material, size, shape, and functionalization need to be chosen and optimized to reach the desiderated aim, as suggested by this review. The NP size-dependent cytotoxic effect was amply analysed as well as the influence of size on cellular uptake and cytotoxicity. On the other hand, the shape is deeply related to body distribution, blood lifespan, macrophage uptake, and membrane internalization.

Moreover, the synthesis processes and the material adopted can modulate other features like biocompatibility, aggregation, and stability. Indeed, each type of NPs presents specific advantages and disadvantages (Table 1), which confer unique properties for specific biomedical applications. Furthermore, the possibility of conjugating them with a large number of different molecules permits obtaining a controlled release mechanism (i.e., mediated by pH, temperature, redox state) and specific targeting, as adopted in cancer treatment to overcome chemotherapy limitations [348]. In this manner, NPs can be employed as DDS not only to act on malignant cells selectively but also for diseases diagnosis, thanks to the capability to detect, for example, primary tumours, lymph nodes, and metastasis or to act as contrast agents in medical imaging techniques. In addition, nanosystems are amply employed in tissue engineering and regenerative medicine to promote tissue differentiation, thanks to the possibility of local delivery of bioactive molecules (i.e., growth factors). Finally, it is recently an object of interest by the scientific community to recruit nanotechnology in vaccine delivery (Table 1), as adopted for BNT162b2 mRNA Covid-19 vaccine, consisting of lipid NPs to deliver mRNA vaccine [231].

Taken together, these features make NPs the starting point for the future of nanomedicine, having a considerable impact on human health. The potential application sectors in which they can be involved are more than those reported in this review. For example, these molecules could permit personalized DDS by improving the patient’s life quality or could be involved as nano-robots to make repairs at the cellular level. Undoubtedly, intelligent multifunctional nanosystems will be the most promising candidates as vectors of biological molecules for a vast range of applications in nanomedicine. Furthermore, different features of each kind of NPs can be integrated to generate inorganic/organic hybrid NPs like lipid-polymer hybrids NPs or mesoporous silica shell-coated AuNPs, cell membrane-coated NPs, or dendrimer-conjugated MNPs [349,350,351]. Therefore, the use of nanotechnology in biomedical application continues to be an evolving field.


tel: +39-09123897409

  1. Funding information: The publication was funded by “Europe Union – FESR o FSE, PON Ricerca e Innovazione 2014-2020 – DM 1062/2021.”

  2. Author contributions: S.C. and G.G. outlined the initial structure of the manuscript. S.C. wrote the manuscript and ideated the figures. All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  3. Conflict of interest: The authors state no conflict of interest.

References

[1] Shahbazi MA, Faghfouri L, Ferreira MPA, Figueiredo P, Maleki H, Sefat F, et al. The versatile biomedical applications of bismuth-based nanoparticles and composites: Therapeutic, diagnostic, biosensing, and regenerative properties. Chem Soc Rev. 2020;49:1253–321.10.1039/C9CS00283ASearch in Google Scholar PubMed

[2] Taniguchi N. On the basic concept of ‘nano-technology. Proceedings of the International Conference on Production Engineering, Tokyo; 1974.Search in Google Scholar

[3] Drexler KE Engines of creation: The coming era of nanotechnology chapter 1: engines of construction. http://www.foresight.org/EOC/EOC_Chapter_1.html. (Accessed 8 February 2021).Search in Google Scholar

[4] Scaffaro R, Re GL, Rigogliuso S, Ghersi G. 3D polylactide-based scaffolds for studying human hepatocarcinoma processes in vitro. Sci Technol Adv Mater. 2012 Aug;13:045003. 10.1088/1468-6996/13/4/045003.Search in Google Scholar PubMed PubMed Central

[5] Pavia FC, La Carrubba V, Ghersi G, Brucato V. Poly-left-lactic acid tubular scaffolds via diffusion induced phase separation: Control of morphology. Polym Eng Sci. 2013;53:431–42.10.1002/pen.23273Search in Google Scholar

[6] Begines B, Ortiz T, Pérez-Aranda M, Martínez G, Merinero M, Argüelles-Arias F, et al. Polymeric nanoparticles for drug delivery: Recent developments and future prospects. Nanomaterials. 2020;10:1–41.10.3390/nano10071403Search in Google Scholar PubMed PubMed Central

[7] Pippa N, Gazouli M, Pispas S. Recent advances and future perspectives in polymer-based nanovaccines. Vaccines. 2021 May 26;9:558. 10.3390/VACCINES9060558.Search in Google Scholar PubMed PubMed Central

[8] Fatima F, Siddiqui S, Khan WA. Nanoparticles as novel emerging therapeutic antibacterial agents in the antibiotics resistant era. Biol Trace Elem Res. 2021;199:2552–64.10.1007/s12011-020-02394-3Search in Google Scholar PubMed

[9] Yuan D, Ellis CM, Davis JJ. Mesoporous silica nanoparticles in bioimaging. Mater (Basel, Switz). 1 September 2020;13(17):3795. 10.3390/MA13173795.Search in Google Scholar PubMed PubMed Central

[10] Lombardo D, Kiselev MA, Caccamo MT. Smart nanoparticles for drug delivery application: development of versatile nanocarrier platforms in biotechnology and nanomedicine. J Nanomater. 2019;2019:1–26. 10.1155/2019/3702518 Search in Google Scholar

[11] Schirrmacher V. From chemotherapy to biological therapy: A review of novel concepts to reduce the side effects of systemic cancer treatment (Review). Int J Oncol. 2019;54:407–19.10.3892/ijo.2018.4661Search in Google Scholar

[12] Ke X, Shen L. Molecular targeted therapy of cancer: The progress and future prospect. Front Lab Med. 2017;1:69–75.10.1016/j.flm.2017.06.001Search in Google Scholar

[13] Jain KK. An overview of drug delivery systems. Methods in molecular biology. Humana New York, NY: Humana Press Inc.; p. 1–54. 10.1007/978-1-4939-9798-5.Search in Google Scholar

[14] Sun T, Zhang YS, Pang B, Hyun DC, Yang M, Xia Y. Engineered nanoparticles for drug delivery in cancer therapy. Angew Chem – Int Ed. 2014;53:12320–64.10.1201/9780429027819-2Search in Google Scholar

[15] Yin IX, Zhang J, Zhao IS, Mei ML, Li Q, Chu CH. The antibacterial mechanism of silver nanoparticles and its application in dentistry. Int J Nanomed. 2020;15:2555–62.10.2147/IJN.S246764Search in Google Scholar PubMed PubMed Central

[16] Oliveira JM, Sousa RA, Kotobuki N, Tadokoro M, Hirose M, Mano JF, et al. The osteogenic differentiation of rat bone marrow stromal cells cultured with dexamethasone-loaded carboxymethylchitosan/poly(amidoamine) dendrimer nanoparticles. Biomaterials. 2009;30:804–13.10.1016/j.biomaterials.2008.10.024Search in Google Scholar PubMed

[17] Lung P, Yang J, Li Q. Nanoparticle formulated vaccines: Opportunities and challenges. Nanoscale. 2020;12:5746–63.10.1039/C9NR08958FSearch in Google Scholar

[18] Fu X, Cai J, Zhang X, Li WD, Ge H, Hu Y. Top-down fabrication of shape-controlled, monodisperse nanoparticles for biomedical applications. Adv Drug Delivery Rev. 2018;132:169–87.10.1016/j.addr.2018.07.006Search in Google Scholar PubMed

[19] Belusso LCS, Lenz GF, Fiorini EE, Pereira AJ, Sequinel R, Bini RA, et al. Synthesis of silver nanoparticles from bottom up approach on borophosphate glass and their applications as SERS, antibacterial and glass-based catalyst. Appl Surf Sci. 2019;473:303–12.10.1016/j.apsusc.2018.12.155Search in Google Scholar

[20] Pareek V, Bhargava A, Gupta R, Jain N, Panwar J. Synthesis and applications of noble metal nanoparticles: a review. Adv Sci Eng Med. 2017;9:527–44.10.1166/asem.2017.2027Search in Google Scholar

[21] Avila-Olias M, Pegoraro C, Battaglia G, Canton I. Inspired by nature: Fundamentals in nanotechnology design to overcome biological barriers. Ther Deliv. 2013;4:27–43.10.4155/tde.12.126Search in Google Scholar PubMed

[22] Braakhuis HM, Cassee FR, Fokkens PH, de la Fonteyne LJ, Oomen AG, Krystek P, et al. Identification of the appropriate dose metric for pulmonary inflammation of silver nanoparticles in an inhalation toxicity study. Nanotoxicology. 2016;10:63–73.10.1016/j.toxlet.2016.06.1953Search in Google Scholar

[23] Ibrahim K, Al-Mutary M, Bakhiet A, Khan, Khan HH. Histopathology of the liver, kidney, and spleen of mice exposed to gold nanoparticles. Molecules. 2018;23:1848.10.3390/molecules23081848Search in Google Scholar PubMed PubMed Central

[24] De Jong WH, Hagens WI, Krystek P, Burger MC, Sips AJ, Geertsma RE. Particle size-dependent organ distribution of gold nanoparticles after intravenous administration. Biomaterials. 2008;29:1912–9.10.1016/j.biomaterials.2007.12.037Search in Google Scholar PubMed

[25] Cabral H, Matsumoto Y, Mizuno K, Chen Q, Murakami M, Kimura M, et al. Accumulation of sub-100 nm polymeric micelles in poorly permeable tumours depends on size. Nat Nanotechnol. 2011;6:815–23.10.1038/nnano.2011.166Search in Google Scholar PubMed

[26] Liu GW, Pippin JW, Eng DG, Lv S, Shankland SJ, Pun SH, et al. Nanoparticles exhibit greater accumulation in kidney glomeruli during experimental glomerular kidney disease. Physiol Rep. 2020;8:e14545.10.14814/phy2.14545Search in Google Scholar PubMed PubMed Central

[27] Zhou Y, Peng Z, Seven ES, Leblanc RM. Crossing the blood-brain barrier with nanoparticles. J Controlled Rel. 2018;270:290–303.10.1016/j.jconrel.2017.12.015Search in Google Scholar PubMed

[28] Bose T, Latawiec D, Mondal PP, Mandal S. Overview of nano-drugs characteristics for clinical application: The journey from the entry to the exit point. J Nanopart Res. 2014;16:1–25.10.1007/s11051-014-2527-7Search in Google Scholar

[29] Barbara R, Belletti D, Pederzoli F, Masoni M, Keller J, Ballestrazzi A, et al. Novel Curcumin loaded nanoparticles engineered for Blood-Brain Barrier crossing and able to disrupt Abeta aggregates. Int J Pharm. 2017;526:413–24.10.1016/j.ijpharm.2017.05.015Search in Google Scholar PubMed

[30] Hua X, Tan S, Bandara HM, Fu Y, Liu S, Smyth HD, et al. Externally controlled triggered-release of drug from plga micro and nanoparticles. PLoS One. 2014;9:e114271.10.1371/journal.pone.0114271Search in Google Scholar PubMed PubMed Central

[31] Cartaxo AL, Costa-Pinto AR, Martins A, Faria S, Gonçalves V, Tiritan ME, et al. Influence of PDLA nanoparticles size on drug release and interaction with cells. J Biomed Mater Res Part A. 2019;107:482–93.10.1002/jbm.a.36563Search in Google Scholar PubMed

[32] Mauro N, Campora S, Ada Mo G, Scialabba C, Ghersi G, Giammona, et al. Polyaminoacid-doxorubicin prodrug micelles as highly selective therapeutics for targeted cancer therapy. RSC Adv. 2016;6:77256–66. 10.1039/C6RA14935A.Search in Google Scholar

[33] Kou L, Sun J, Zhai Y, He Z. The endocytosis and intracellular fate of nanomedicines: Implication for rational design. Asian J Pharm Sci. 2013;8:1–10.10.1016/j.ajps.2013.07.001Search in Google Scholar

[34] Shaw S, Ganguly S, Sibanda P, Chakraborty S. Dispersion characteristics of blood during nanoparticle assisted drug delivery process through a permeable microvessel. Microvasc Res. 2014;92:25–33.10.1016/j.mvr.2013.12.007Search in Google Scholar PubMed

[35] Geng Y, Dalhaimer P, Cai S, Tsai R, Tewari M, Minko T, et al. Shape effects of filaments versus spherical particles in flow and drug delivery. Nat Nanotechnol. 2007;2:249–55.10.1038/nnano.2007.70Search in Google Scholar PubMed PubMed Central

[36] Zhao Y, Wang Y, Ran F, Cui Y, Liu C, Zhao Q, et al. A comparison between sphere and rod nanoparticles regarding their in vivo biological behavior and pharmacokinetics. Sci Rep. 2017;7:1–11.10.1038/s41598-017-03834-2Search in Google Scholar PubMed PubMed Central

[37] MacParland SA, Tsoi KM, Ouyang B, Ma XZ, Manuel J, Fawaz A, et al. Phenotype determines nanoparticle uptake by human macrophages from liver and blood. ACS Nano. 2017;11:2428–43.10.1021/acsnano.6b06245Search in Google Scholar PubMed

[38] Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Delivery Rev. 2016;99:28–51.10.1016/j.addr.2015.09.012Search in Google Scholar PubMed PubMed Central

[39] Patsula V, Horák D, Kučka J, Macková H, Lobaz V, Francová P, et al. Synthesis and modification of uniform PEG-neridronate-modified magnetic nanoparticles determines prolonged blood circulation and biodistribution in a mouse preclinical model. Sci Rep. 2019;9:1–12.10.1038/s41598-019-47262-wSearch in Google Scholar PubMed PubMed Central

[40] Xue W, Liu Y, Zhang N, Yao Y, Ma P, Wen H, et al. Effects of core size and PEG coating layer of iron oxide nanoparticles on the distribution and metabolism in mice. Int J Nanomed. 2018;13:5719–31.10.2147/IJN.S165451Search in Google Scholar PubMed PubMed Central

[41] Rodriguez PL, Harada T, Christian DA, Pantano DA, Tsai RK, Discher DE. Minimal ‘self’ peptides that inhibit phagocytic clearance and enhance delivery of nanoparticles. Sci (80-). 2013;339:971–5.10.1126/science.1229568Search in Google Scholar PubMed PubMed Central

[42] Parodi A, Quattrocchi N, Van De Ven AL, Chiappini C, Evangelopoulos M, Martinez JO, et al. Synthetic nanoparticles functionalized with biomimetic leukocyte membranes possess cell-like functions. Nat Nanotechnol. 2013;8:61–8.10.1038/nnano.2012.212Search in Google Scholar PubMed PubMed Central

[43] Hu CM, Zhang L, Aryal S, Cheung C, Fang RH, Zhang L. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc Natl Acad Sci U S A. 2011;108:10980–5.10.1073/pnas.1106634108Search in Google Scholar PubMed PubMed Central

[44] Sharma G, Valenta DT, Altman Y, Harvey S, Xie H, Mitragotri S, et al. Polymer particle shape independently influences binding and internalization by macrophages. J Control Rel. 2010;147:408–12.10.1016/j.jconrel.2010.07.116Search in Google Scholar PubMed PubMed Central

[45] Champion JA, Mitragotri S. Role of target geometry in phagocytosis. Proc Natl Acad Sci U S A. 2006;103:4930–4.10.1073/pnas.0600997103Search in Google Scholar PubMed PubMed Central

[46] Tan J, Shah S, Thomas A, Ou-Yang HD, Liu Y. The influence of size, shape and vessel geometry on nanoparticle distribution. Microfluid Nanofluidics. 2013;14:77–87.10.1007/s10404-012-1024-5Search in Google Scholar PubMed PubMed Central

[47] Li Z, Sun L, Zhang Y, Dove AP, O'Reilly RK, Chen G. Shape effect of glyco-nanoparticles on macrophage cellular uptake and immune response. ACS Macro Lett. 2016;5:1059–64.10.1021/acsmacrolett.6b00419Search in Google Scholar PubMed PubMed Central

[48] Barua S, Yoo JW, Kolhar P, Wakankar A, Gokarn YR, Mitragotri S. Particle shape enhances specificity of antibody-displaying nanoparticles. Proc Natl Acad Sci U S A. 2013;110:3270–5.10.1073/pnas.1216893110Search in Google Scholar PubMed PubMed Central

[49] Xu R, Wang D, Zhang J, Li Y. Shape-dependent catalytic activity of silver nanoparticles for the oxidation of styrene. Chem – An Asian J. 2006;1:888–93.10.1002/asia.200600260Search in Google Scholar PubMed

[50] Helmlinger J, Sengstock C, Groß-Heitfeld C, Mayer C, Schildhauer TA, Köller M, et al. Silver nanoparticles with different size and shape: Equal cytotoxicity, but different antibacterial effects. RSC Adv. 2016;6:18490–501.10.1039/C5RA27836HSearch in Google Scholar

[51] Cheon JY, Kim SJ, Rhee YH, Kwon OH, Park WH. Shape-dependent antimicrobial activities of silver nanoparticles. Int J Nanomed. 2019;14:2773–80.10.2147/IJN.S196472Search in Google Scholar PubMed PubMed Central

[52] Arno MC, Inam M, Weems AC, Li Z, Binch A, Platt CI, et al. Exploiting the role of nanoparticle shape in enhancing hydrogel adhesive and mechanical properties. Nat Commun. 2020;11:1–9.10.1038/s41467-020-15206-ySearch in Google Scholar PubMed PubMed Central

[53] Ladj R, Bitar A, Eissa M, Mugnier Y, Le Dantec R, Fessi H, et al. Individual inorganic nanoparticles: Preparation, functionalization and in vitro biomedical diagnostic applications. J Mater Chem B. 2013;1:1381–96.10.1039/c2tb00301eSearch in Google Scholar PubMed

[54] McNamara K, Tofail SAM. Nanosystems: The use of nanoalloys, metallic, bimetallic, and magnetic nanoparticles in biomedical applications. Phys Chem Chem Phys. 2015;17:27981–95.10.1039/C5CP00831JSearch in Google Scholar

[55] Chakravarty R, Goel S, Dash A, Cai W. Radiolabeled inorganic nanoparticles for positron emission tomography imaging of cancer: An overview. Q J Nucl Med Mol Imaging. 2017;61:181–204.10.23736/S1824-4785.17.02969-7Search in Google Scholar PubMed PubMed Central

[56] Patra JK, Das G, Fraceto LF, Campos E, Rodriguez-Torres M, Acosta-Torres LS, et al. Nano based drug delivery systems: Recent developments and future prospects 10 Technology 1007 Nanotechnology 03 Chemical Sciences 0306 Physical Chemistry (incl. Structural) 03 Chemical Sciences 0303 Macromolecular and Materials Chemistry 11 Medical and Health Sciences 1115 Pharmacology and Pharmaceutical Sciences 09 Engineering 0903 Biomedical Engineering Prof Ueli Aebi, Prof Peter Gehr. J Nanobiotechnol. 2018;16:71.10.1186/s12951-018-0392-8Search in Google Scholar PubMed PubMed Central

[57] Hong X, Hall EAH. Contribution of gold nanoparticles to the signal amplification in surface plasmon resonance. Analyst. 2012;137:4712–9.10.1039/c2an35742aSearch in Google Scholar PubMed

[58] Dong J, Carpinone PL, Pyrgiotakis G, Demokritou P, Moudgil BM. Synthesis of precision gold nanoparticles using Turkevich method. KONA Powder Part J. 2020;37:224–32.10.14356/kona.2020011Search in Google Scholar PubMed PubMed Central

[59] Li J, Li Q, Ma X, Tian B, Li T, Yu J, et al. Biosynthesis of gold nanoparticles by the extreme bacterium – Deinococcus radiodurans – and an evaluation of their antibacterial properties. Int J Nanomed. 2016;11:5931–44.10.2147/IJN.S119618Search in Google Scholar PubMed PubMed Central

[60] Kim CK, Ghosh P, Rotello VM. Multimodal drug delivery using gold nanoparticles. Nanoscale. 2009;1:61–7.10.1039/b9nr00112cSearch in Google Scholar PubMed

[61] Li W, Li X, Liu S, Yang W, Pan F, Yang XY, et al. Gold nanoparticles attenuate metastasis by tumor vasculature normalization and epithelial-mesenchymal transition inhibition. Int J Nanomed. 2017;12:12–3509.10.2147/IJN.S128802Search in Google Scholar PubMed PubMed Central

[62] Ghersi G. Roles of molecules involved in epithelial/mesenchymal transition during angiogenesis. Front Biosci. 2008;13:2335–55.10.2741/2848Search in Google Scholar PubMed

[63] Sulaiman GM, Waheeb HM, Jabir MS, Khazaal SH, Dewir YH, Naidoo Y. Hesperidin loaded on gold nanoparticles as a drug delivery system for a successful biocompatible, anti-cancer, anti-inflammatory and phagocytosis inducer model. Sci Rep. 2020;10:1–16.10.1038/s41598-020-66419-6Search in Google Scholar PubMed PubMed Central

[64] Pedrosa P, Corvo ML, Ferreira-Silva M, Martins P, Carvalheiro MC, Costa PM, et al. Targeting cancer resistance via multifunctional gold nanoparticles. Int J Mol Sci. 2019;20:5510.10.3390/ijms20215510Search in Google Scholar PubMed PubMed Central

[65] Letfullin RR, Joenathan C, George TF, Zharov VP. Laser-induced explosion of gold nanoparticles: Potential role for nanophotothermolysis of cancer. Nanomedicine. 2006;1:473–80.10.2217/17435889.1.4.473Search in Google Scholar PubMed

[66] Zheng Y, Zhang Y, Zhang T, Cai H, Xie X, Yang Y, et al. AuNSs@Glycopolymer-ConA hybrid nanoplatform for photothermal therapy of hepatoma cells. Chem Eng J. 2020;389:124460.10.1016/j.cej.2020.124460Search in Google Scholar

[67] Yang Y, Hu Y, Du H, Ren L, Wang H. Colloidal plasmonic gold nanoparticles and gold nanorings: shape-dependent generation of singlet oxygen and their performance in enhanced photodynamic cancer therapy. Int J Nanomed. 2018;13:2065–78.10.2147/IJN.S156347Search in Google Scholar PubMed PubMed Central

[68] Xu X, Chong Y, Liu X, Fu H, Yu C, Huang J, et al. Multifunctional nanotheranostic gold nanocages for photoacoustic imaging guided radio/photodynamic/photothermal synergistic therapy. Acta Biomater. 2019;84:328–38.10.1016/j.actbio.2018.11.043Search in Google Scholar PubMed

[69] Tao W, Ziemer KS, Gill HS. Gold nanoparticle-M2e conjugate coformulated with CpG induces protective immunity against influenza A virus. Nanomedicine. 2014;9:237–51.10.2217/nnm.13.58Search in Google Scholar PubMed PubMed Central

[70] Bastús NG, Sánchez-Tilló E, Pujals S, Farrera C, Kogan MJ, Giralt E, et al. Peptides conjugated to gold nanoparticles induce macrophage activation. Mol Immunol. 2009;46:743–8.10.1016/j.molimm.2008.08.277Search in Google Scholar PubMed

[71] Chen YS, Hung YC, Lin WH, Huang GS, Huang GS. Assessment of gold nanoparticles as a size-dependent vaccine carrier for enhancing the antibody response against synthetic foot-and-mouth disease virus peptide. Nanotechnology. 2010;21:195101. 10.1088/0957-4484/21/19/195101 Search in Google Scholar PubMed

[72] Tazaki T, Tabata K, Ainai A, Ohara Y, Kobayashi S, Ninomiya T, et al. Shape-dependent adjuvanticity of nanoparticle-conjugated RNA adjuvants for intranasal inactivated influenza vaccines. RSC Adv. 2018;8:16527–36.10.1039/C8RA01690ASearch in Google Scholar

[73] Gulla SK, Rao BR, Moku G, Jinka S, Nimmu NV, Khalid S, et al. In vivo targeting of DNA vaccines to dendritic cells using functionalized gold nanoparticles. Biomater Sci. 2019;7:773–88.10.1039/C8BM01272ESearch in Google Scholar PubMed

[74] Chen TY, Chen MR, Liu SW, Lin JY, Yang YT, Huang HY, et al. Assessment of polyethylene glycol-coated gold nanoparticle toxicity and inflammation in vivo using nf-κb reporter mice. Int J Mol Sci. 2020;21:1–16.10.3390/ijms21218158Search in Google Scholar PubMed PubMed Central

[75] Xu M, Liu J, Xu X, et al. Synthesis and comparative biological properties of Ag-PEG nanoparticles with tunable morphologies from Janus to multi-core shell structure. Mater (Basel). 20 September 2018;11. 10.3390/ma11101787.Search in Google Scholar PubMed PubMed Central

[76] Samuel MS, Jose S, Selvarajan E, Mathimani T, Pugazhendhi A. Biosynthesized silver nanoparticles using Bacillus amyloliquefaciens; Application for cytotoxicity effect on A549 cell line and photocatalytic degradation of p-nitrophenol. J Photochem Photobiol B Biol. 2020 Jan;202:111642. 10.1016/j.jphotobiol.2019.111642.Search in Google Scholar PubMed

[77] Pala R, Zeng Y, Pattnaik S, Busi S, Alomari N, Nauli SM, et al. Functionalized silver nanoparticles for sensing, molecular imaging and therapeutic applications. Curr Nanomed. 2019;8:234–50.10.2174/2468187308666180508144919Search in Google Scholar

[78] Abbaszadegan A, Ghahramani Y, Gholami A, Hemmateenejad B, Dorostkar S, Nabavizadeh M, et al. The effect of charge at the surface of silver nanoparticles on antimicrobial activity against gram-positive and gram-negative bacteria: A preliminary study. J Nanomater. 2015;2015:1–8. 10.1155/2015/720654.Search in Google Scholar

[79] Liao C, Li Y, Tjong SC. Bactericidal and cytotoxic properties of silver nanoparticles. Int J Mol Sci. 2 January 2019;20. 10.3390/ijms20020449.Search in Google Scholar PubMed PubMed Central

[80] Meikle TG, Dyett BP, Strachan JB, White J, Drummond CJ, Conn CE. Preparation, characterization, and antimicrobial activity of cubosome encapsulated metal nanocrystals. ACS Appl Mater Interfaces. 2020;12:6944–54.10.1021/acsami.9b21783Search in Google Scholar PubMed

[81] Hong X, Wen J, Xiong X, Hu Y. Shape effect on the antibacterial activity of silver nanoparticles synthesized via a microwave-assisted method. Env Sci Pollut Res. 2016;23:4489–97.10.1007/s11356-015-5668-zSearch in Google Scholar PubMed

[82] Raza MA, Kanwal Z, Rauf A, Sabri AN, Riaz S, Naseem S. Size- and shape-dependent antibacterial studies of silver nanoparticles synthesized by wet chemical routes. Nanomaterials. 2016;6:74.10.3390/nano6040074Search in Google Scholar PubMed PubMed Central

[83] Shanmuganathan R, MubarakAli D, Prabakar D, Muthukumar H, Thajuddin N, Kumar SS, et al. An enhancement of antimicrobial efficacy of biogenic and ceftriaxone-conjugated silver nanoparticles: green approach. Env Sci Pollut Res. 2018;25:10362–70.10.1007/s11356-017-9367-9Search in Google Scholar PubMed

[84] Yu L, Shang F, Chen X, Ni J, Yu L, Zhang M, et al. The anti-biofilm effect of silvernanoparticle-decorated quercetin nanoparticles on a multi-drug resistant Escherichia coli strain isolated from a dairy cow with mastitis. PeerJ. 2018;2018:e5711.10.7717/peerj.5711Search in Google Scholar PubMed PubMed Central

[85] Siddique MH, Aslam B, Imran M, Ashraf A, Nadeem H, Hayat S, et al. Effect of silver nanoparticles on biofilm formation and eps production of multidrug-resistant Klebsiella pneumoniae. Biomed Res Int. 2020;2020:6398165. 10.1155/2020/6398165.Search in Google Scholar PubMed PubMed Central

[86] Salleh A, Naomi R, Utami ND, Mohammad AW, Mahmoudi E, Mustafa N, et al. The potential of silver nanoparticles for antiviral and antibacterial applications: a mechanism of action. Nanomaterials. 2020;10:1566.10.3390/nano10081566Search in Google Scholar PubMed PubMed Central

[87] Elechiguerra JL, Burt JL, Morones JR, Camacho-Bragado A, Gao X, Lara HH, et al. Interaction of silver nanoparticles with HIV-1. J Nanobiotechnol. 2005;3:6.10.1186/1477-3155-3-6Search in Google Scholar PubMed PubMed Central

[88] Lara HH, Ayala-Nuñez NV, Ixtepan-Turrent L, Rodriguez-Padilla C. Mode of antiviral action of silver nanoparticles against HIV-1. J Nanobiotechnol. 2010;8:1.10.1186/1477-3155-8-1Search in Google Scholar PubMed PubMed Central

[89] Tsai CH, Whiteley CG, Lee DJ. Interactions between HIV-1 protease, silver nanoparticles, and specific peptides. J Taiwan Inst Chem Eng. 2019;103:20–32.10.1016/j.jtice.2019.07.019Search in Google Scholar

[90] Morris D, Ansar M, Speshock J, Ivanciuc T, Qu Y, Casola A, et al. Antiviral and immunomodulatory activity of silver nanoparticles in experimental RSV infection. Viruses. 2019;11:732.10.3390/v11080732Search in Google Scholar PubMed PubMed Central

[91] Jeremiah SS, Miyakawa K, Morita T, Yamaoka Y, Ryo A. Potent antiviral effect of silver nanoparticles on SARS-CoV-2. Biochem Biophys Res Commun. 2020;533:195–200.10.1016/j.bbrc.2020.09.018Search in Google Scholar PubMed PubMed Central

[92] Balagna C, Perero S, Percivalle E, Nepita EV, Ferraris M. Virucidal effect against coronavirus SARS-CoV-2 of a silver nanocluster/silica composite sputtered coating. Open Ceram. 2020;1:100006.10.1016/j.oceram.2020.100006Search in Google Scholar

[93] Hsin Y-H, Chen C-F, Huang S, Shih TS, Lai PS, Chueh PJ. The apoptotic effect of nanosilver is mediated by a ROS- and JNK-dependent mechanism involving the mitochondrial pathway in NIH3T3 cells. Toxicol Lett. 2008;179:130–9.10.1016/j.toxlet.2008.04.015Search in Google Scholar PubMed

[94] Sriram MI, Kanth SB, Kalishwaralal K, Gurunathan S. Antitumor activity of silver nanoparticles in Dalton’s lymphoma ascites tumor model. Int J Nanomed. 2010;5:753–62.10.2147/IJN.S11727Search in Google Scholar

[95] Perde-Schrepler M, Florea A, Brie I, Virag P, Fischer-Fodor E, Vâlcan A, et al. Size-dependent cytotoxicity and genotoxicity of silver nanoparticles in cochlear cells in vitro. J Nanomater. 2019;2019:1–12. 10.1155/2019/6090259.Search in Google Scholar

[96] Farzin A, Etesami SA, Quint J, Memic A, Tamayol A. Magnetic nanoparticles in cancer therapy and diagnosis. Adv Healthc Mater. 2020;9:e1901058.10.1002/adhm.201901058Search in Google Scholar PubMed PubMed Central

[97] Ali A, Shah T, Ullah R, Zhou P, Guo M, Ovais M, et al. Review on recent progress in magnetic nanoparticles: synthesis, characterization, and diverse applications. Front Chem. 2021;9:548.10.3389/fchem.2021.629054Search in Google Scholar PubMed PubMed Central

[98] Li X, Lovell JF, Yoon J, Chen X. Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat Rev Clin Oncol. 2020;2020(17):657–74.10.1038/s41571-020-0410-2Search in Google Scholar PubMed

[99] Wang X, Yang R, Yuan C, An Y, Tang Q, Chen D. Preparation of folic acid-targeted temperature-sensitive magnetoliposomes and their antitumor effects in vitro and in vivo. Target Oncol. 2018;13:481–94.10.1007/s11523-018-0577-ySearch in Google Scholar PubMed

[100] Huang G, Chen H, Dong Y, Luo X, Yu H, Moore Z, et al. Superparamagnetic iron oxide nanoparticles: amplifying ROS stress to improve anticancer drug efficacy. Theranostics. 2013;3:116–26.10.7150/thno.5411Search in Google Scholar PubMed PubMed Central

[101] Barrow M, Taylor A, Fuentes-Caparrós AM, Sharkey J, Daniels LM, Mandal P, et al. SPIONs for cell labelling and tracking using MRI: magnetite or maghemite? Biomater Sci. 2017;6:101–6.10.1039/C7BM00515FSearch in Google Scholar

[102] Nelson NR, Port JD, Pandey MK. Use of superparamagnetic iron oxide nanoparticles (spions) via multiple imaging modalities and modifications to reduce cytotoxicity: an educational review. J Nanotheranostics. 2020;1:105–35.10.3390/jnt1010008Search in Google Scholar

[103] Zhang W, Cao S, Liang S, Tan CH, Luo B, Xu X, et al. Differently charged super-paramagnetic iron oxide nanoparticles preferentially induced M1-like phenotype of macrophages. Front Bioeng Biotechnol. 2020;8:537.10.3389/fbioe.2020.00537Search in Google Scholar PubMed PubMed Central

[104] Abdal Dayem A, Hossain MK, Lee SB, Kim K, Saha SK, Yang GM, et al. The role of reactive oxygen species (ROS) in the biological activities of metallic nanoparticles. Int J Mol Sci. 10 January 2017;18. 10.3390/IJMS18010120.Search in Google Scholar

[105] Mao C, Xiang Y, Liu X, Zheng Y, Yeung K, Cui Z, et al. Local photothermal/photodynamic synergistic therapy by disrupting bacterial membrane to accelerate reactive oxygen species permeation and protein leakage. ACS Appl Mater Interfaces. 2019;11:17902–14.10.1021/acsami.9b05787Search in Google Scholar PubMed

[106] Singh N, Jenkins GJ, Asadi R, Doak SH. Potential toxicity of superparamagnetic iron oxide nanoparticles (SPION). Nano Rev. 2010;1:5358.10.3402/nano.v1i0.5358Search in Google Scholar PubMed PubMed Central

[107] Tandale P, Choudhary N, Singh J, Sharma A, Shukla A, Sriram P, et al. Fluorescent quantum dots: An insight on synthesis and potential biological application as drug carrier in cancer. Biochem Biophys Rep. 2021;26:100962.10.1016/j.bbrep.2021.100962Search in Google Scholar PubMed PubMed Central

[108] Zhou P, Liu H, Gong L, Tang B, Shi Y, Yang C, et al. A faster detection method for high-sensitivity cardiac troponin – POCT quantum dot fluorescence immunoassay. J Thorac Dis. 2019;11:1506–13.10.21037/jtd.2019.03.25Search in Google Scholar PubMed PubMed Central

[109] Zhao MX, Zeng EZ. Application of functional quantum dot nanoparticles as fluorescence probes in cell labeling and tumor diagnostic imaging. Nanoscale Res Lett. 2015;10:1–9.10.1186/s11671-015-0873-8Search in Google Scholar PubMed PubMed Central

[110] Nicosia A, Cavallaro G, Costa S, Utzeri MA, Cuttitta A, Giammona G, et al. Carbon nanodots for on demand chemophotothermal therapy combination to elicit necroptosis: overcoming apoptosis resistance in breast cancer cell lines. Cancers (Basel). 2020;12:3114.10.3390/cancers12113114Search in Google Scholar PubMed PubMed Central

[111] Radenkovic D, Kobayashi H, Remsey-Semmelweis E, Seifalian AM. Quantum dot nanoparticle for optimization of breast cancer diagnostics and therapy in a clinical setting. Nanomed Nanotechnol Biol Med. 2016;12:1581–92.10.1016/j.nano.2016.02.014Search in Google Scholar PubMed

[112] Nifontova G, Ramos-Gomes F, Baryshnikova M, Alves F, Nabiev I, Sukhanova A. Cancer cell targeting with functionalized quantum dot-encoded polyelectrolyte microcapsules. Front Chem. 2019;7:34.10.3389/fchem.2019.00034Search in Google Scholar PubMed PubMed Central

[113] Karakoçak BB, Laradji A, Primeau T, Berezin MY, Li S, Ravi N. Hyaluronan-conjugated carbon quantum dots for bioimaging use. ACS Appl Mater Interfaces. 2021;13:277–86.10.1021/acsami.0c20088Search in Google Scholar PubMed PubMed Central

[114] Ruzycka-Ayoush M, Kowalik P, Kowalczyk A, Bujak P, Nowicka AM, Wojewodzka M, et al. Quantum dots as targeted doxorubicin drug delivery nanosystems. Cancer Nanotechnol. 2021;12:8.10.1186/s12645-021-00077-9Search in Google Scholar

[115] Qin X, Liu J, Zhang Q, Chen W, Zhong X, He J. Synthesis of yellow-fluorescent carbon nano-dots by microplasma for imaging and photocatalytic inactivation of cancer cells. Nanoscale Res Lett. 2021;16:14.10.1186/s11671-021-03478-2Search in Google Scholar PubMed PubMed Central

[116] Lovrić J, Bazzi HS, Cuie Y, Fortin GR, Winnik FM, Maysinger D. Differences in subcellular distribution and toxicity of green and red emitting CdTe quantum dots. J Mol Med. 2005;83:377–85.10.1007/s00109-004-0629-xSearch in Google Scholar PubMed

[117] Oh E, Liu R, Nel A, Gemill KB, Bilal M, Cohen Y, et al. Meta-analysis of cellular toxicity for cadmium-containing quantum dots. Nat Nanotechnol. 2016;11:479–86.10.1038/nnano.2015.338Search in Google Scholar PubMed

[118] Xu G, Zeng S, Zhang B, Swihart MT, Yong KT, Prasad PN. New generation cadmium-free quantum dots for biophotonics and nanomedicine. Chem Rev. 2016;116:12234–327.10.1021/acs.chemrev.6b00290Search in Google Scholar PubMed

[119] Supreet SG. Recent advances on cadmium free quantum dots-liquid crystal nanocomposites. Appl Mater Today. 2020;21:100840.10.1016/j.apmt.2020.100840Search in Google Scholar

[120] Sun X, Shi M, Zhang C, Yuan J, Yin M, Du S, et al. Fluorescent Ag-In-S/ZnS quantum dots for tumor drainage lymph node imaging in vivo. ACS Appl Nano Mater. 2021;2021:1029–37.10.1021/acsanm.0c02542Search in Google Scholar

[121] Ogihara Y, Yukawa H, Kameyama T, Nishi H, Onoshima D, Ishikawa T, et al. Labeling and in vivo visualization of transplanted adipose tissue-derived stem cells with safe cadmium-free aqueous ZnS coating of ZnS-AgInS 2 nanoparticles. Sci Rep. 2017;7:1–12.10.1038/srep40047Search in Google Scholar PubMed PubMed Central

[122] Ali M, Zayed D, Ramadan W, Kamel OA, Shehab M, Ebrahim S. Synthesis, characterization and cytotoxicity of polyethylene glycol-encapsulated CdTe quantum dots. Int Nano Lett. 2019;9:61–71.10.1007/s40089-018-0262-2Search in Google Scholar

[123] Ashree J, Wang Q, Chao Y. Glyco-functionalised quantum dots and their progress in cancer diagnosis and treatment. Front Chem Sci Eng. 2020;14:365–77.10.1007/s11705-019-1863-7Search in Google Scholar

[124] Yang Y, Cui J, Zheng M, Hu C, Tan S, Xiao Y, et al. One-step synthesis of amino-functionalized fluorescent carbon nanoparticles by hydrothermal carbonization of chitosan. Chem Commun. 2012;48:380–2.10.1039/C1CC15678KSearch in Google Scholar PubMed

[125] Schipper ML, Iyer G, Koh AL, Cheng Z, Ebenstein Y, Aharoni A, et al. Particle size, surface coating, and PEGylation influence the biodistribution of quantum dots in living mice. Small. 2009;5:126–34.10.1002/smll.200800003Search in Google Scholar PubMed PubMed Central

[126] Hong E, Liu L, Li C, Shan D, Cao H, Wang B. Study on cytotoxicity of polyethylene glycol and albumin bovine serum molecule-modified quantum dots prepared by hydrothermal method. J Mater Res. 2020;35:1135–42.10.1557/jmr.2020.78Search in Google Scholar

[127] Ballou B, Lagerholm BC, Ernst LA, Bruchez MP, Waggoner AS. Noninvasive imaging of quantum dots in mice. Bioconjug Chem. 2004;15:79–86.10.1021/bc034153ySearch in Google Scholar PubMed

[128] Rajendiran K, Zhao Z, Pei DS, Fu A. Antimicrobial activity and mechanism of functionalized quantum dots. Polymers. 1 October 2019;11. 10.3390/polym11101670 Search in Google Scholar PubMed PubMed Central

[129] Hao X, Huang L, Zhao C, Chen S, Lin W, Lin Y, et al. Antibacterial activity of positively charged carbon quantum dots without detectable resistance for wound healing with mixed bacteria infection. Mater Sci Eng C. 2021;123:111971.10.1016/j.msec.2021.111971Search in Google Scholar PubMed

[130] Ristic BZ, Milenkovic MM, Dakic IR, Todorovic-Markovic BM, Milosavljevic MS, Budimir MD, et al. Photodynamic antibacterial effect of graphene quantum dots. Biomaterials. 2014;35:4428–35.10.1016/j.biomaterials.2014.02.014Search in Google Scholar PubMed

[131] Lin F, Bao Y-W, Wu F-G. Carbon dots for sensing and killing. Microorg C. 2019;5:33.10.3390/c5020033Search in Google Scholar

[132] Yang J, Gao G, Zhang X, Ma Y-H, Chen X, Wu F-G. One-step synthesized carbon dots with bacterial contact-enhanced fluorescence emission property: Fast Gram-type identification and selective Gram-positive bacterial inactivation. Carbon N Y. 2019;146:827–39.10.1016/j.carbon.2019.02.040Search in Google Scholar

[133] Yang J, Zhang X, Ma YH, Gao G, Chen X, Jia HR, et al. Carbon dot-based platform for simultaneous bacterial distinguishment and antibacterial applications. ACS Appl Mater Interfaces. 2016;8:32170–81.10.1021/acsami.6b10398Search in Google Scholar PubMed

[134] Huang Z, Zhou T, Yuan Y, Natalie Kłodzińska S, Zheng T, Sternberg C, et al. Synthesis of carbon quantum dot-poly lactic-co-glycolic acid hybrid nanoparticles for chemo-photothermal therapy against bacterial biofilms. J Colloid Interface Sci. 2020;577:66–74.10.1016/j.jcis.2020.05.067Search in Google Scholar PubMed

[135] Wang H, Chen Q, Zhou SH. Carbon-based hybrid nanogels: a synergistic nanoplatform for combined biosensing, bioimaging, and responsive drug delivery. Chem Soc Rev. 2018;47:4198–232. 10.1039/C7CS00399D.Search in Google Scholar PubMed

[136] Maiti D, Tong X, Mou X, Yang K. Carbon-based nanomaterials for biomedical applications: a recent study. Front Pharmacol. 2019;9:1401.10.3389/fphar.2018.01401Search in Google Scholar PubMed PubMed Central

[137] Liu F, Mou X, Song J, Li Q, Liu J. Novel carbon-based magnetic luminescent nanocomposites for multimodal imaging. Front Chem. 2020;8:611.10.3389/fchem.2020.00611Search in Google Scholar PubMed PubMed Central

[138] Priyadarsini S, Mohanty S, Mukherjee S, Basu S, Mishra M. Graphene and graphene oxide as nanomaterials for medicine and biology application. J Nanostructure Chem. 2018;8:123–37.10.1007/s40097-018-0265-6Search in Google Scholar

[139] Jagiełło J, Chlanda A, Baran M, Gwiazda M, Lipińska L. Synthesis and characterization of graphene oxide and reduced graphene oxide composites with inorganic nanoparticles for biomedical applications. Nanomaterials. 2020;10:1846.10.3390/nano10091846Search in Google Scholar PubMed PubMed Central

[140] Smith AT, LaChance AM, Zeng S, Liu B, Sun L. Synthesis, properties, and applications of graphene oxide/reduced graphene oxide and their nanocomposites. Nano Mater Sci. 2019;1:31–47.10.1016/j.nanoms.2019.02.004Search in Google Scholar

[141] Campora S, Mauro N, Griffiths P, Giammona G, Ghersi G. Graphene nanosystems as supports in siRNA delivery. Chem Eng Trans. 2018;64:415–20. 10.3303/CET1864070.Search in Google Scholar

[142] Yin F, Hu K, Chen Y, Yu M, Wang D, Wang Q, et al. SiRNA delivery with PEGylated graphene oxide nan osheets for combined photothermal and genetherapy for pancreatic cancer. Theranostics. 2017;7:1133–48.10.7150/thno.17841Search in Google Scholar PubMed PubMed Central

[143] Newby SD, Masi T, Griffin CD, King WJ, Chipman A, Stephenson S, et al. Functionalized graphene nanoparticles induce human mesenchymal stem cells to express distinct extracellular matrix proteins mediating osteogenesis. Int J Nanomed. 2020;15:2501–13.10.2147/IJN.S245801Search in Google Scholar PubMed PubMed Central

[144] Qi Z, Guo W, Zheng S, Fu C, Ma Y, Pan S, et al. Enhancement of neural stem cell survival, proliferation and differentiation by IGF-1 delivery in graphene oxide-incorporated PLGA electrospun nanofibrous mats. RSC Adv. 2019;9:8315–25.10.1039/C8RA10103ESearch in Google Scholar PubMed PubMed Central

[145] Saburi E, Islami M, Hosseinzadeh S, Moghadam AS, Mansour RN, Azadian E, et al. In vitro osteogenic differentiation potential of the human induced pluripotent stem cells augments when grown on Graphene oxide-modified nanofibers. Gene. 2019;696:72–9.10.1016/j.gene.2019.02.028Search in Google Scholar PubMed

[146] Chen GY, Pang DW, Hwang SM, Tuan HY, Hu YC. A graphene-based platform for induced pluripotent stem cells culture and differentiation. Biomaterials. 2012;33:418–27.10.1016/j.biomaterials.2011.09.071Search in Google Scholar PubMed

[147] Lee WC, Lim CH, Shi H, Tang LA, Wang Y, Lim CT, et al. Origin of enhanced stem cell growth and differentiation on graphene and graphene oxide. ACS Nano. 2011;5:7334–41.10.1021/nn202190cSearch in Google Scholar PubMed

[148] Menaa F, Abdelghani A, Menaa B. Graphene nanomaterials as biocompatible and conductive scaffolds for stem cells: Impact for tissue engineering and regenerative medicine. J Tissue Eng Regenerative Med. 2015;9:1321–38.10.1002/term.1910Search in Google Scholar PubMed

[149] Dubey N, Bentini R, Islam I, Cao T, Castro Neto AH, Rosa V. Graphene: A Versatile Carbon-Based Material for Bone Tissue Engineering. Stem Cell Int. 2015;2015:804213. 10.1155/2015/804213.Search in Google Scholar PubMed PubMed Central

[150] Duan S, Yang X, Mei F, Tang Y, Li X, Shi Y, et al. Enhanced osteogenic differentiation of mesenchymal stem cells on poly(l-lactide) nanofibrous scaffolds containing carbon nanomaterials. J Biomed Mater Res Part A. 2015;103:1424–35.10.1002/jbm.a.35283Search in Google Scholar PubMed

[151] Peña-Bahamonde J, Nguyen HN, Fanourakis SK, Rodrigues DF. Recent advances in graphene-based biosensor technology with applications in life sciences. J Nanobiotechnol. 2018;16:75.10.1186/s12951-018-0400-zSearch in Google Scholar PubMed PubMed Central

[152] Cordaro A, Neri G, Sciortino MT, Scala A, Piperno A. Graphene-based strategies in liquid biopsy and in viral diseases diagnosis. Nanomaterials. 2020;10:1014.10.3390/nano10061014Search in Google Scholar PubMed PubMed Central

[153] Seo G, Lee G, Kim MJ, Baek SH, Choi M, Ku KB, et al. Rapid detection of COVID-19 causative virus (SARS-cov-2) in human nasopharyngeal swab specimens using field-effect transistor-based biosensor. ACS Nano. 2020;14:5135–42.10.1021/acsnano.0c02823Search in Google Scholar PubMed PubMed Central

[154] Norizan MN, Moklis MH, Ngah Demon SZ, Halim NA, Samsuri A, Mohamad IS, et al. Carbon nanotubes: Functionalisation and their application in chemical sensors. RSC Adv. 2020;10:43704–32.10.1039/D0RA09438BSearch in Google Scholar PubMed PubMed Central

[155] Rathinavel S, Priyadharshini K, Panda D. A review on carbon nanotube: An overview of synthesis, properties, functionalization, characterization, and the application. Mater Sci Eng B: Solid-State Mater Adv Technol. 2021;268:115095.10.1016/j.mseb.2021.115095Search in Google Scholar

[156] Negri V, Pacheco-Torres J, Calle D, López-Larrubia P. Carbon nanotubes in biomedicine. Top Curr Chem. 1 February 2020;378(1):15. 10.1007/s41061-019-0278-8.Search in Google Scholar PubMed

[157] Xu H, Liu M, Lan M, Yuan H, Yu W, Tian J, et al. Mussel-inspired PEGylated carbon nanotubes: Biocompatibility evaluation and drug delivery applications. Toxicol Res (Camb). 2016;5:1371–9.10.1039/C6TX00094KSearch in Google Scholar PubMed PubMed Central

[158] Singh S, Mehra NK, Jain NK. Development and characterization of the paclitaxel loaded riboflavin and thiamine conjugated carbon nanotubes for cancer treatment. Pharm Res. 2016;33:1769–81.10.1007/s11095-016-1916-2Search in Google Scholar PubMed

[159] Su Y, Hu Y, Wang Y, Xu X, Yuan Y, Li Y, et al. A precision-guided MWNT mediated reawakening the sunk synergy in RAS for anti-angiogenesis lung cancer therapy. Biomaterials. 2017;139:75–90.10.1016/j.biomaterials.2017.05.046Search in Google Scholar PubMed

[160] Vaithilingam J, Sanjuan-Alberte P, Campora S, et al. Multifunctional bioinstructive 3D architectures to modulate cellular behavior. Adv Funct Mater. 2019;29(38):1902016. 10.1002/adfm.201902016.Search in Google Scholar

[161] Villarreal CC, Pham T, Ramnani P, Mulchandani A. Carbon allotropes as sensors for environmental monitoring. Curr Opin Electrochem. 2017;3:106–13.10.1016/j.coelec.2017.07.004Search in Google Scholar

[162] Voiry D, Pagona G, Del Canto E, Ortolani L, Morandi V, Noé L, et al. Reductive dismantling and functionalization of carbon nanohorns. Chem Commun. 2015;51:5017–9.10.1039/C4CC10389KSearch in Google Scholar PubMed

[163] Karousis N, Suarez-Martinez I, Ewels CP, Tagmatarchis N. Structure, properties, functionalization, and applications of carbon nanohorns. Chem Rev. 2016;116:4850–83.10.1021/acs.chemrev.5b00611Search in Google Scholar PubMed

[164] Gomez IJ, Arnaiz B, Cacioppo M, Arcudi F, Prato M. Nitrogen-doped carbon nanodots for bioimaging and delivery of paclitaxel. J Mater Chem B. 2018;6:5540–8.10.1039/C8TB01796DSearch in Google Scholar PubMed

[165] Wang J, Xu M, Wang D, Li Z, Primo FL, Tedesco AC, et al. Copper-doped carbon dots for optical bioimaging and photodynamic therapy. Inorg Chem. 2019;58:13394–402.10.1021/acs.inorgchem.9b02283Search in Google Scholar PubMed

[166] Zhou Z, Song J, Nie L, Chen X. Reactive oxygen species generating systems meeting challenges of photodynamic cancer therapy. Chem Soc Rev. 2016;45:6597–626.10.1039/C6CS00271DSearch in Google Scholar PubMed PubMed Central

[167] Li S, Guo Z, Zhang Y, Xue W, Liu Z. Blood compatibility evaluations of fluorescent carbon dots. ACS Appl Mater Interfaces. 2015;7:19153–62.10.1021/acsami.5b04866Search in Google Scholar PubMed

[168] Gaur M, Misra C, Yadav AB, Swaroop S, Maolmhuaidh FÓ, Bechelany M, et al. Biomedical applications of carbon nanomaterials: fullerenes, quantum dots, nanotubes, nanofibers, and graphene. Mater (Basel). 1 October 2021;14(20):5978. 10.3390/MA14205978.Search in Google Scholar

[169] Prylutskyy YI, Petrenko VI, Ivankov OI, Kyzyma OA, Bulavin LA, Litsis OO, et al. On the origin of C 60 fullerene solubility in aqueous solution. Langmuir. 2014;30:3967–70. 10.1021/la404976k.Search in Google Scholar PubMed

[170] Moussa F. [60]Fullerene and derivatives for biomedical applications. Nanobiomater Nanostruct Mater Biomed Appl. 2018;113–36.10.1016/B978-0-08-100716-7.00005-2Search in Google Scholar

[171] Schinazi RF, Sijbesma R, Srdanov G, Hill CL, Wudl F. Synthesis and virucidal activity of a water-soluble, configurationally stable, derivatized C60 fullerene. Antimicrob Agents Chemother. 1993;37:1707–10.10.1128/AAC.37.8.1707Search in Google Scholar PubMed PubMed Central

[172] Martinez ZS, Castro E, Seong CS, Cerón MR, Echegoyen L, Llano M. Fullerene derivatives strongly inhibit hiv-1 replication by affecting virus maturation without impairing protease activity. Antimicrob Agents Chemother. 2016;60:5731–41.10.1128/AAC.00341-16Search in Google Scholar PubMed PubMed Central

[173] Uthappa UT, Arvind OR, Sriram G, Losic D, Ho-Young-Jung, Kigga M, et al. Nanodiamonds and their surface modification strategies for drug delivery applications. J Drug Deliv Sci Technol. 2020;60:101993.10.1016/j.jddst.2020.101993Search in Google Scholar

[174] Choi S, Noh SH, Lim CO, Kim HJ, Jo HS, Min JS, et al. Icariin-functionalized nanodiamonds to enhance osteogenic capacity in vitro. Nanomater (Basel, Switz). 2020;10:1–14.10.3390/nano10102071Search in Google Scholar PubMed PubMed Central

[175] Zhang Q, Mochalin VN, Neitzel I, Knoke IY, Han J, Klug CA, et al. Fluorescent PLLA-nanodiamond composites for bone tissue engineering. Biomaterials. 2011;32:87–94.10.1016/j.biomaterials.2010.08.090Search in Google Scholar PubMed

[176] Chen F, Hableel G, Zhao ER, Jokerst JV. Multifunctional nanomedicine with silica: Role of silica in nanoparticles for theranostic, imaging, and drug monitoring. J Colloid Interface Sci. 2018;521:261–79.10.1016/j.jcis.2018.02.053Search in Google Scholar PubMed PubMed Central

[177] Li Z, Mu Y, Peng C, Lavin MF, Shao H, Du Z. Understanding the mechanisms of silica nanoparticles for nanomedicine. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 1 January 2021;13(1):e1658. 10.1002/wnan.1658.Search in Google Scholar

[178] Ghaferi M, Koohi Moftakhari Esfahani M, Raza A, Al Harthi S, Ebrahimi Shahmabadi H, Alavi SE. Mesoporous silica nanoparticles: synthesis methods and their therapeutic use-recent advances. J Drug Target. 2021;29:131–54.10.1080/1061186X.2020.1812614Search in Google Scholar

[179] Jafari S, Derakhshankhah H, Alaei L, Fattahi A, Varnamkhasti BS, Saboury AA. Mesoporous silica nanoparticles for therapeutic/diagnostic applications. Biomed Pharmacotherapy. 2019;109:1100–11.10.1016/j.biopha.2018.10.167Search in Google Scholar

[180] Lugowski SJ, Smith DC, Bonek H, Lugowski J, Peters W, Semple J. Analysis of silicon in human tissues with special reference to silicone breast implants. J Trace Elem Med Biol. 2000;14:31–42.10.1016/S0946-672X(00)80021-8Search in Google Scholar

[181] Park JH, Gu L, Von Maltzahn G, Ruoslahti E, Bhatia SN, Sailor MJ. Biodegradable luminescent porous silicon nanoparticles for in vivo applications. Nat Mater. 2009;8:331–6.10.1038/nmat2398Search in Google Scholar PubMed PubMed Central

[182] Bhavsar D, Patel V, Sawant K. Systemic investigation of in vitro and in vivo safety, toxicity and degradation of mesoporous silica nanoparticles synthesized using commercial sodium silicate. Microporous Mesoporous Mater. 2019;284:343–52.10.1016/j.micromeso.2019.04.050Search in Google Scholar

[183] Kempen PJ, Greasley S, Parker KA, Campbell JL, Chang HY, Jones JR, et al. Theranostic mesoporous silica nanoparticles biodegrade after pro-survival drug delivery and ultrasound/magnetic resonance imaging of stem cells. Theranostics. 2015;5:631–42.10.7150/thno.11389Search in Google Scholar PubMed PubMed Central

[184] Kuang G, Zhang Q, He S, Liu Y. Curcumin-loaded PEGylated mesoporous silica nanoparticles for effective photodynamic therapy. RSC Adv. 2020;10:24624–30.10.1039/D0RA04778CSearch in Google Scholar PubMed PubMed Central

[185] Dogra P, Adolphi NL, Wang Z, Lin YS, Butler KS, Durfee PN, et al. Establishing the effects of mesoporous silica nanoparticle properties on in vivo disposition using imaging-based pharmacokinetics. Nat Commun. 2018;9:1–14.10.1038/s41467-018-06730-zSearch in Google Scholar PubMed PubMed Central

[186] Zhuang J, Chen S, Hu Y, Yang F, Huo Q, Xie N, et al. Tumour-targeted and redox-responsive mesoporous silica nanoparticles for controlled release of doxorubicin and an siRNA against metastatic breast cancer. Int J Nanomed. 2022;2021(16):1961–76.10.2147/IJN.S375352Search in Google Scholar PubMed PubMed Central

[187] Duo Y, Li Y, Chen C, Liu B, Wang X, Zeng X, et al. DOX-loaded pH-sensitive mesoporous silica nanoparticles coated with PDA and PEG induce pro-death autophagy in breast cancer. RSC Adv. 2017;7:39641–50.10.1039/C7RA05135BSearch in Google Scholar

[188] Carniato F, Tei L, Botta M. Gd-based mesoporous silica nanoparticles as MRI probes. Eur J Inorg Chem. 2018;2018:4936–54.10.1002/ejic.201801039Search in Google Scholar

[189] Jeong HJ, Yoo RJ, Kim JK, Kim MH, Park SH, Kim H, et al. Macrophage cell tracking PET imaging using mesoporous silica nanoparticles via in vivo bioorthogonal F-18 labeling. Biomaterials. 2019;199:32–9.10.1016/j.biomaterials.2019.01.043Search in Google Scholar PubMed

[190] Hayashi K, Wataru S, Yogo T. Iodinated silica/porphyrin hybrid nanoparticles for X-ray computed tomography/fluorescence dual-modal imaging of tumors. J Asian Ceram Soc. 2014;2:429–34.10.1016/j.jascer.2014.09.003Search in Google Scholar

[191] Ciccione J, Jia T, Coll JL, Parra K, Amblard M, Jebors S, et al. Unambiguous and controlled one-pot synthesis of multifunctional silica nanoparticles. Chem Mater. 2016;28:885–9.10.1021/acs.chemmater.5b04398Search in Google Scholar

[192] Ferrauto G, Carniato F, Di Gregorio E, Botta M, Tei L. Photoacoustic ratiometric assessment of mitoxantrone release from theranostic ICG-conjugated mesoporous silica nanoparticles. Nanoscale. 2019;11:18031–6.10.1039/C9NR06524ESearch in Google Scholar

[193] Lv R, Wang D, Xiao L, Chen G, Xia J, Prasad PN. Stable ICG-loaded upconversion nanoparticles: Silica core/shell theranostic nanoplatform for dual-modal upconversion and photoacoustic imaging together with photothermal therapy. Sci Rep. 2017;7:1–11.10.1038/s41598-017-16016-xSearch in Google Scholar PubMed PubMed Central

[194] Zhang Z, Wang L, Wang J, Jiang X, Li X, Hu Z, et al. Mesoporous silica-coated gold nanorods as a light-mediated multifunctional theranostic platform for cancer treatment. Adv Mater. 2012;24:1418–23.10.1002/adma.201104714Search in Google Scholar PubMed

[195] Ramasamy T, Ruttala HB, Sundaramoorthy P, Poudel BK, Youn YS, Ku SK, et al. Multimodal selenium nanoshell-capped Au@mSiO2 nanoplatform for NIR-responsive chemo-photothermal therapy against metastatic breast cancer. NPG Asia Mater. 2018;10:197–216.10.1038/s41427-018-0034-5Search in Google Scholar

[196] Ramasamy M, Lee J-H, Lee J. Development of gold nanoparticles coated with silica containing the antibiofilm drug cinnamaldehyde and their effects on pathogenic bacteria. Int J Nanomed. 2017;12:2813–28.10.2147/IJN.S132784Search in Google Scholar PubMed PubMed Central

[197] Chen X, Liu Y, Lin A, Huang N, Long L, Gang Y, et al. Folic acid-modified mesoporous silica nanoparticles with pH-responsiveness loaded with Amp for an enhanced effect against anti-drug-resistant bacteria by overcoming efflux pump systems. Biomater Sci. 2018;6:1923–35.10.1039/C8BM00262BSearch in Google Scholar PubMed

[198] Yang S, Han X, Yang Y, Qiao H, Yu Z, Liu Y, et al. Bacteria-targeting nanoparticles with microenvironment-responsive antibiotic release to eliminate intracellular staphylococcus aureus and associated infection. ACS Appl Mater Interfaces. 2018;10:14299–311.10.1021/acsami.7b15678Search in Google Scholar PubMed

[199] Gan Q, Zhu J, Yuan Y, Liu H, Qian J, Li Y, et al. A dual-delivery system of pH-responsive chitosan-functionalized mesoporous silica nanoparticles bearing BMP-2 and dexamethasone for enhanced bone regeneration. J Mater Chem B. 2015;3:2056–66.10.1039/C4TB01897DSearch in Google Scholar

[200] Shi M, Xia L, Chen Z, Lv F, Zhu H, Wei F, et al. Europium-doped mesoporous silica nanosphere as an immune-modulating osteogenesis/angiogenesis agent. Biomaterials. 2017;144:176–87.10.1016/j.biomaterials.2017.08.027Search in Google Scholar PubMed

[201] Oliveira DCP, de Barros ALB, Belardi RM, de Goes AM, de Oliveira Souza BK, Soares DCF. Mesoporous silica nanoparticles as a potential vaccine adjuvant against Schistosoma mansoni. J Drug Deliv Sci Technol. 2016;35:234–40.10.1016/j.jddst.2016.07.002Search in Google Scholar

[202] Hong X, Zhong X, Du G, Hou Y, Zhang Y, Zhang Z, et al. The pore size of mesoporous silica nanoparticles regulates their antigen delivery efficiency. Sci Adv. 1 June 2020;6(25):eaaz4462. 10.1126/sciadv.aaz4462.Search in Google Scholar PubMed PubMed Central

[203] Anselmo AC, Mitragotri S. Nanoparticles in the clinic: An update. Bioeng Transl Med. September 2019;4:10143. 10.1002/btm2.10143.Search in Google Scholar PubMed PubMed Central

[204] Panahi Y, Farshbaf M, Mohammadhosseini M, Mirahadi M, Khalilov R, Saghfi S, et al. Recent advances on liposomal nanoparticles: synthesis, characterization and biomedical applications. Artif Cells Nanomed Biotechnol. 2017;45:788–99.10.1080/21691401.2017.1282496Search in Google Scholar PubMed

[205] Fan Y, Marioli M, Zhang K. Analytical characterization of liposomes and other lipid nanoparticles for drug delivery. J Pharm Biomed Anal. 2021;192:113642.10.1016/j.jpba.2020.113642Search in Google Scholar PubMed

[206] Antimisiaris SG, Marazioti A, Kannavou M, et al. Overcoming barriers by local drug delivery with liposomes. Adv Drug Deliv Rev. 2 February 2021;174:53–86. 10.1016/j.addr.2021.01.019.Search in Google Scholar PubMed

[207] Daraee H, Etemadi A, Kouhi M, Alimirzalu S, Akbarzadeh A. Application of liposomes in medicine and drug delivery. Artif Cells, Nanomed Biotechnol. 2016;44:381–91.10.3109/21691401.2014.953633Search in Google Scholar PubMed

[208] Miranda D, Lovell JF. Mechanisms of light‐induced liposome permeabilization. Bioeng Transl Med. 2016;1:267–76.10.1002/btm2.10032Search in Google Scholar PubMed PubMed Central

[209] Jhaveri AM, Torchilin VP. Multifunctional polymeric micelles for delivery of drugs and siRNA. Front Pharmacology. 2014 5 APR;5:77.10.3389/fphar.2014.00077Search in Google Scholar PubMed PubMed Central

[210] Riaz MK, Riaz MA, Zhang X, Lin C, Wong KH, Chen X, et al. Surface functionalization and targeting strategies of liposomes in solid tumor therapy: A review. Int J Mol Sci. 9 January 2018;19. 10.3390/ijms19010195 Search in Google Scholar PubMed PubMed Central

[211] Bozzuto G, Molinari A. Liposomes as nanomedical devices. Int J Nanomed. 2015;10:975–99.10.2147/IJN.S68861Search in Google Scholar PubMed PubMed Central

[212] Nunes SS, Fernandes RS, Cavalcante CH, da Costa César I, Leite EA, Lopes S, et al. Influence of PEG coating on the biodistribution and tumor accumulation of pH-sensitive liposomes. Drug Deliv Transl Res. 2019;9:123–30.10.1007/s13346-018-0583-8Search in Google Scholar PubMed PubMed Central

[213] Xue X, Huang Y, Bo R, Jia B, Wu H, Yuan Y, et al. Trojan Horse nanotheranostics with dual transformability and multifunctionality for highly effective cancer treatment. Nat Commun. 2018;9:1–15.10.1038/s41467-018-06093-5Search in Google Scholar PubMed PubMed Central

[214] Lee H, Jiang D, Pardridge WM. Lyoprotectant optimization for the freeze-drying of receptor-targeted trojan horse liposomes for plasmid DNA delivery. Mol Pharm. 2020;17:2165–74.10.1021/acs.molpharmaceut.0c00310Search in Google Scholar PubMed

[215] Jiang D, Lee H, Pardridge WM. Plasmid DNA gene therapy of the Niemann-Pick C1 mouse with transferrin receptor-targeted Trojan horse liposomes. Sci Rep. 2020;10:13334.10.1038/s41598-020-70290-wSearch in Google Scholar PubMed PubMed Central

[216] Spagnou S, Miller AD, Keller M. Lipidic carriers of siRNA: Differences in the formulation, cellular uptake, and delivery with plasmid DNA. Biochemistry. 2004;43:13348–56.10.1021/bi048950aSearch in Google Scholar PubMed

[217] Xia C-F, Boado RJ, Zhang Y, Chu C, Pardridge WM. Intravenous glial-derived neurotrophic factor gene therapy of experimental Parkinson’s disease with Trojan horse liposomes and a tyrosine hydroxylase promoter. J Gene Med. 2008;10:306–15.10.1002/jgm.1152Search in Google Scholar PubMed

[218] Kumar B, Pandey M, Pottoo FH, Fayaz F, Sharma A, Sahoo PK. Liposomes: novel drug delivery approach for targeting parkinson’s disease. Curr Pharm Des. 2020;26:4721–37.10.2174/1381612826666200128145124Search in Google Scholar PubMed

[219] Zhang Y, Zhang YF, Bryant J, Charles A, Boado RJ, Pardridge WM. Intravenous RNA interference gene therapy targeting the human epidermal growth factor receptor prolongs survival in intracranial brain cancer. Clin Cancer Res. 2004;10:3667–77.10.1158/1078-0432.CCR-03-0740Search in Google Scholar PubMed

[220] Pardridge WM. Brain delivery of nanomedicines: trojan horse liposomes for plasmid DNA gene therapy of the brain. Front Med Technol. 2020;2:602236.10.3389/fmedt.2020.602236Search in Google Scholar PubMed PubMed Central

[221] Menina S, Eisenbeis J, Kamal MAM, Koch M, Bischoff M, Gordon S, et al. Bioinspired liposomes for oral delivery of colistin to combat intracellular infections by Salmonella enterica. Adv Healthc Mater. 2019;8:1900564.10.1002/adhm.201900564Search in Google Scholar PubMed

[222] Peine KJ, Gupta G, Brackman DJ, Papenfuss TL, Ainslie KM, Satoskar AR, et al. Liposomal resiquimod for the treatment of leishmania donovani infection. J Antimicrob Chemother. 2014;69:168–75.10.1093/jac/dkt320Search in Google Scholar PubMed PubMed Central

[223] Pinheiro M, Lúcio M, Lima JL, Reis S. Liposomes as drug delivery systems for the treatment of TB. Nanomedicine. 2011;6:1413–28.10.2217/nnm.11.122Search in Google Scholar PubMed

[224] Ye J, Yang Y, Dong W, Gao Y, Meng Y, Wang H, et al. Drug-free mannosylated liposomes inhibit tumor growth by promoting the polarization of tumor-associated macrophages. Int J Nanomed. 2019;14:3203–20.10.2147/IJN.S207589Search in Google Scholar PubMed PubMed Central

[225] Van Den Hoven JM, Van Tomme SR, Metselaar JM, Nuijen B, Beijnen JH, Storm G. Liposomal drug formulations in the treatment of rheumatoid arthritis. Mol Pharmaceutics. 2011;8:1002–15.10.1021/mp2000742Search in Google Scholar PubMed

[226] Qi Y, Yan X, Xia T, Liu S. Use of macrophage as a Trojan horse for cancer nanotheranostics. Mater Des. 2021;198:109388.10.1016/j.matdes.2020.109388Search in Google Scholar

[227] Foged C, Arigita C, Sundblad A, Jiskoot W, Storm G, Frokjaer S. Interaction of dendritic cells with antigen-containing liposomes: Effect of bilayer composition. Vaccine. 2004;22:1903–13.10.1016/j.vaccine.2003.11.008Search in Google Scholar PubMed

[228] Huang WC, Deng B, Seffouh A, Ortega J, Long CA, Suresh RV, et al. Antibody response of a particle-inducing, liposome vaccine adjuvant admixed with a Pfs230 fragment. npj Vaccines. 1 December 2020;5:23. 10.1038/s41541-020-0173-x.Search in Google Scholar PubMed PubMed Central

[229] Aldosari BN, Alfagih IM, Almurshedi AS. Lipid nanoparticles as delivery systems for RNA-based vaccines. Pharmaceutics. 2021;13:1–29.10.3390/pharmaceutics13020206Search in Google Scholar PubMed PubMed Central

[230] Espeseth AS, Cejas PJ, Citron MP, Wang D, DiStefano DJ, Callahan C, et al. Modified mRNA/lipid nanoparticle-based vaccines expressing respiratory syncytial virus F protein variants are immunogenic and protective in rodent models of RSV infection. npj Vaccines. 1 December 2020;5(1):16. 10.1038/s41541-020-0163-z.Search in Google Scholar PubMed PubMed Central

[231] Polack FP, Thomas SJ, Kitchin N, Absalon J, Gurtman A, Lockhart S, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med. 2020;383:2603–15.10.1056/NEJMoa2034577Search in Google Scholar PubMed PubMed Central

[232] Baden LR, El Sahly HM, Essink B, Kotloff K, Frey S, Novak R, et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N Engl J Med. 2021;384:403–16.10.1056/NEJMoa2035389Search in Google Scholar PubMed PubMed Central

[233] Schoenmaker L, Witzigmann D, Kulkarni JA, Verbeke R, Kersten G, Jiskoot W, et al. mRNA-lipid nanoparticle COVID-19 vaccines: Structure and stability. Int J Pharm. 2021;601:120586.10.1016/j.ijpharm.2021.120586Search in Google Scholar PubMed PubMed Central

[234] Chen H, Wang L, Zeng X, Schwarz H, Nanda HS, Peng X, et al. Exosomes, a new star for targeted delivery. Front Cell Dev Biol. 2021;9:2827.10.3389/fcell.2021.751079Search in Google Scholar PubMed PubMed Central

[235] Xiong H, Huang Z, Yang Z, Lin Q, Yang B, Fang X, et al. Recent progress in detection and profiling of cancer cell-derived exosomes. Small. 2021;17:2007971.10.1002/smll.202007971Search in Google Scholar PubMed

[236] Osaki M, Okada F. Exosomes and their role in cancer progression. Yonago Acta Med. 2019;62:182–90.10.33160/yam.2019.06.002Search in Google Scholar PubMed PubMed Central

[237] Yong T, Zhang X, Bie N, Zhang H, Zhang X, Li F, et al. Tumor exosome-based nanoparticles are efficient drug carriers for chemotherapy. Nat Commun. 2019 1012019;10:1–16.10.1038/s41467-019-11718-4Search in Google Scholar PubMed PubMed Central

[238] Xu M, Yang Q, Sun X, Wang Y. Recent advancements in the loading and modification of therapeutic exosomes. Front Bioeng Biotechnol. 2020;8:1173.10.3389/fbioe.2020.586130Search in Google Scholar PubMed PubMed Central

[239] Dai J, Su Y, Zhong S, Cong L, Liu B, Yang J, et al. Exosomes: key players in cancer and potential therapeutic strategy. Signal Transduct Target Ther. 2020;5:1–10.10.1038/s41392-020-00261-0Search in Google Scholar PubMed PubMed Central

[240] Ag Seleci D, Seleci M, Walter JG, Stahl F, Scheper T. Niosomes as nanoparticular drug carriers: Fundamentals and recent applications. J Nanomater. 2016;2016:1–13. 10.1155/2016/7372306.Search in Google Scholar

[241] Yeo PL, Lim CL, Chye SM, Kiong Ling AP, Koh RY. Niosomes: A review of their structure, properties, methods of preparation, and medical applications. Asian Biomed. 2017;11:301–13.10.1515/abm-2018-0002Search in Google Scholar

[242] Ge X, Wei M, He S, Yuan WE. Advances of non-ionic surfactant vesicles (Niosomes) and their application in drug delivery. Pharm. 2019;11:55.10.3390/pharmaceutics11020055Search in Google Scholar PubMed PubMed Central

[243] Bartelds R, Nematollahi MH, Pols T, Stuart MC, Pardakhty A, Asadikaram G, et al. Niosomes, an alternative for liposomal delivery. PLoS One. 1 April 2018;13(4):e0194179. 10.1371/JOURNAL.PONE.0194179.Search in Google Scholar

[244] Arzani G, Haeri A, Daeihamed M, Bakhtiari-Kaboutaraki H, Dadashzadeh S. Niosomal carriers enhance oral bioavailability of carvedilol: effects of bile salt-enriched vesicles and carrier surface charge. Int J Nanomed. 2015;10:4797–813.10.2147/IJN.S84703Search in Google Scholar PubMed PubMed Central

[245] Sultan AA, El-Gizawy SA, Osman MA, El Maghraby GM. Niosomes for oral delivery of nateglinide: in situ-in vivo correlation. J Liposome Res. 2018;28:209–17.10.1080/08982104.2017.1343835Search in Google Scholar PubMed

[246] Sita VG, Jadhav D, Vavia P. Niosomes for nose-to-brain delivery of bromocriptine: Formulation development, efficacy evaluation and toxicity profiling. J Drug Deliv Sci Technol. 2020;58:101791.10.1016/j.jddst.2020.101791Search in Google Scholar

[247] Li D, Martini N, Wu Z, Chen S, Falconer JR, Locke M, et al. Niosomal nanocarriers for enhanced dermal delivery of epigallocatechin gallate for protection against oxidative stress of the skin. Pharm. 2022;14:726.10.3390/pharmaceutics14040726Search in Google Scholar

[248] Durak S, Esmaeili Rad M, Alp Yetisgin A, Eda Sutova H, Kutlu O, Cetinel S, et al. Niosomal drug delivery systems for ocular disease – recent advances and future prospects. Nanomaterials. 2020;10:1–29.10.3390/nano10061191Search in Google Scholar

[249] Müller RH, Radtke M, Wissing SA. Solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) in cosmetic and dermatological preparations. Adv Drug Deliv Rev. 2002;54:S131–55.10.1016/S0169-409X(02)00118-7Search in Google Scholar

[250] Schwarz C, Mehnert W, Lucks JS, Müller RH. Solid lipid nanoparticles (SLN) for controlled drug delivery. I. Production, characterization and sterilization. J Control Rel. 1994;30:83–96.10.1016/0168-3659(94)90047-7Search in Google Scholar

[251] Tenchov R, Bird R, Curtze AE, Zhou Q. Lipid nanoparticles from liposomes to mRNA vaccine delivery, a landscape of research diversity and advancement. ACS Nano. 2021;15:16982–7015.10.1021/acsnano.1c04996Search in Google Scholar

[252] Mehnert W, Mäder K. Solid lipid nanoparticles: Production, characterization and applications. Adv Drug Deliv Rev. 2001;47:165–96.10.1016/S0169-409X(01)00105-3Search in Google Scholar

[253] Mishra V, Bansal K, Verma A, Yadav N, Thakur S, Sudhakar K, et al. Solid lipid nanoparticles: emerging colloidal nano drug delivery systems. Pharm. 2018;10:191.10.3390/pharmaceutics10040191Search in Google Scholar PubMed PubMed Central

[254] Abdellatif AAH, Mohammed HA, Khan RA, Singh V, Bouazzaoui A, Yusuf M, et al. Nano-scale delivery: A comprehensive review of nano-structured devices, preparative techniques, site-specificity designs, biomedical applications, commercial products, and references to safety, cellular uptake, and organ toxicity. Nanotechnol Rev. 2021;10:1493–559.10.1515/ntrev-2021-0096Search in Google Scholar

[255] Salah E, Abouelfetouh MM, Pan Y, Chen D, Xie S. Solid lipid nanoparticles for enhanced oral absorption: A review. Colloids Surf B Biointerfaces. 1 December 2020;196:111305. 10.1016/J.COLSURFB.2020.111305.Search in Google Scholar PubMed

[256] Wang H, Ding W, Peng L, Fan H, Yan C, Xu S, et al. Gadolinium-loaded solid lipid nanoparticles for colorectal tumor in MR colonography. J Biomed Nanotechnol. 2020;16:594–602.10.1166/jbn.2020.2922Search in Google Scholar PubMed

[257] Wang JL, Hanafy MS, Xu H, Leal J, Zhai Y, Ghosh D, et al. Aerosolizable siRNA-encapsulated solid lipid nanoparticles prepared by thin-film freeze-drying for potential pulmonary delivery. Int J Pharm. 1 March 2021;596:120215. 10.1016/J.IJPHARM.2021.120215.Search in Google Scholar PubMed

[258] Singh M, Guzman-Aranguez A, Hussain A, Srinivas CS, Kaur IP. Solid lipid nanoparticles for ocular delivery of isoniazid: evaluation, proof of concept and in vivo safety & kinetics. Nanomed (Lond). 2019;14:465–91.10.2217/nnm-2018-0278Search in Google Scholar PubMed

[259] Garcês A, Amaral MH, Sousa Lobo JM, Silva AC. Formulations based on solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) for cutaneous use: A review. Eur J Pharm Sci. 2018;112:159–67.10.1016/j.ejps.2017.11.023Search in Google Scholar PubMed

[260] Masiiwa WL, Gadaga LL. Intestinal permeability of artesunate-loaded solid lipid nanoparticles using the everted gut method. J Drug Deliv. 2018;2018:1–9.10.1155/2018/3021738Search in Google Scholar PubMed PubMed Central

[261] Lin CH, Chen CH, Lin ZC, Fang JY. Recent advances in oral delivery of drugs and bioactive natural products using solid lipid nanoparticles as the carriers. J Food Drug Anal. 2017;25:219–34.10.1016/j.jfda.2017.02.001Search in Google Scholar PubMed PubMed Central

[262] Zhang Z, Lu Y, Qi J, Wu W. An update on oral drug delivery via intestinal lymphatic transport. Acta Pharm Sin B. 2021;11:2449–68.10.1016/j.apsb.2020.12.022Search in Google Scholar PubMed PubMed Central

[263] Ganesan P, Ramalingam P, Karthivashan G, Ko YT, Choi DK. Recent developments in solid lipid nanoparticle and surface-modified solid lipid nanoparticle delivery systems for oral delivery of phyto-bioactive compounds in various chronic diseases. Int J Nanomed. 2018;13:1569–83.10.2147/IJN.S155593Search in Google Scholar PubMed PubMed Central

[264] Shah P, Chavda K, Vyas B, Patel S. Formulation development of linagliptin solid lipid nanoparticles for oral bioavailability enhancement: role of P-gp inhibition. Drug Deliv Transl Res. 2021;11:1166–85.10.1007/s13346-020-00839-9Search in Google Scholar PubMed

[265] Obinu A, Burrai GP, Cavalli R, Galleri G, Migheli R, Antuofermo E, et al. Transmucosal solid lipid nanoparticles to improve genistein absorption via intestinal lymphatic transport. Pharmaceutics. 2021;13:1–17.10.3390/pharmaceutics13020267Search in Google Scholar PubMed PubMed Central

[266] Li H, Qu X, Qian W, Song Y, Wang C, Liu W. Andrographolide-loaded solid lipid nanoparticles enhance anti-cancer activity against head and neck cancer and precancerous cells. Oral Dis. 2022;28:142–9.10.1111/odi.13751Search in Google Scholar PubMed

[267] Dudhipala N, Gorre T. Neuroprotective effect of ropinirole lipid nanoparticles enriched hydrogel for parkinson’s disease: in vitro, ex vivo, pharmacokinetic and pharmacodynamic evaluation. Pharmaceutics. 1 May 2020;12(5):448. 10.3390/PHARMACEUTICS12050448.Search in Google Scholar

[268] Shen MY, Liu TI, Yu TW, Kv R, Chiang WH, Tsai YC, et al. Hierarchically targetable polysaccharide-coated solid lipid nanoparticles as an oral chemo/thermotherapy delivery system for local treatment of colon cancer. Biomaterials. 2019;197:86–100.10.1016/j.biomaterials.2019.01.019Search in Google Scholar PubMed

[269] Khosa A, Reddi S, Saha RN. Nanostructured lipid carriers for site-specific drug delivery. Biomed Pharmacother. 2018;103:598–613.10.1016/j.biopha.2018.04.055Search in Google Scholar PubMed

[270] Jaiswal P, Gidwani B, Vyas A. Nanostructured lipid carriers and their current application in targeted drug delivery. 2014;44:27–40. 103109/216914012014909822.Search in Google Scholar

[271] Salvi VR, Pawar P. Nanostructured lipid carriers (NLC) system: A novel drug targeting carrier. J Drug Deliv Sci Technol. 2019;51:255–67.10.1016/j.jddst.2019.02.017Search in Google Scholar

[272] Costa CP, Moreira JN, Sousa Lobo JM, Silva AC. Intranasal delivery of nanostructured lipid carriers, solid lipid nanoparticles and nanoemulsions: A current overview of in vivo studies. Acta Pharm Sin B. 2021;11:925–40.10.1016/j.apsb.2021.02.012Search in Google Scholar PubMed PubMed Central

[273] Singh SK, Hidau MK, Gautam S, Gupta K, Singh KP, Singh SK, et al. Glycol chitosan functionalized asenapine nanostructured lipid carriers for targeted brain delivery: Pharmacokinetic and teratogenic assessment. Int J Biol Macromol. 2018;108:1092–100.10.1016/j.ijbiomac.2017.11.031Search in Google Scholar PubMed

[274] Pai RV, Vavia PR. Chitosan oligosaccharide enhances binding of nanostructured lipid carriers to ocular mucins: Effect on ocular disposition. Int J Pharm. 2020;577:119095.10.1016/j.ijpharm.2020.119095Search in Google Scholar PubMed

[275] Naderi N, Karponis D, Mosahebi A, Seifalian AM. Nanoparticles in wound healing; from hope to promise, from promise to routine. Front Biosci (Landmark Ed. 2018;23:1038–59.10.2741/4632Search in Google Scholar PubMed

[276] Vairo C, Collantes M, Quincoces G, Villullas S, Peñuelas I, Pastor M, et al. Preclinical safety of topically administered nanostructured lipid carriers (NLC) for wound healing application: biodistribution and toxicity studies. Int J Pharm. 2019;569:118484.10.1016/j.ijpharm.2019.118484Search in Google Scholar PubMed

[277] Chato-Astrain J, Chato-Astrain I, Sánchez-Porras D, García-García ÓD, Bermejo-Casares F, Vairo C, et al. Generation of a novel human dermal substitute functionalized with antibiotic-loaded nanostructured lipid carriers (NLCs) with antimicrobial properties for tissue engineering. J Nanobiotechnology. 2020;18:1–13.10.1186/s12951-020-00732-0Search in Google Scholar PubMed PubMed Central

[278] Zhu N, Wang D, Xie F, Qin M, Lin Z, Wang Y. Fabrication and characterization of calcium-phosphate lipid system for potential dental application. Front Chem. 2020;8:161.10.3389/fchem.2020.00161Search in Google Scholar PubMed PubMed Central

[279] Persano F, Gigli G, Leporatti S. Lipid-polymer hybrid nanoparticles in cancer therapy: current overview and future directions. Nano Express. 2021;2:012006.10.1088/2632-959X/abeb4bSearch in Google Scholar

[280] Sivadasan D, Sultan MH, Madkhali O, Almoshari Y, Thangavel N. Polymeric lipid hybrid nanoparticles (plns) as emerging drug delivery platform – A comprehensive review of their properties, preparation methods, and therapeutic applications. Pharm. 2021;13:1291.10.3390/pharmaceutics13081291Search in Google Scholar PubMed PubMed Central

[281] Mukherjee A, Waters AK, Kalyan P, Achrol AS, Kesari S, Yenugonda VM. Lipid–polymer hybrid nanoparticles as a next-generation drug delivery platform: state of the art, emerging technologies, and perspectives. Int J Nanomed. 2019;14:1937–52.10.2147/IJN.S198353Search in Google Scholar PubMed PubMed Central

[282] Khan MM, Madni A, Filipczak N, Pan J, Rehman M, Rai N, et al. Folate targeted lipid chitosan hybrid nanoparticles for enhanced anti-tumor efficacy. Nanomed Nanotechnol Biol Med. 2020;28:102228.10.1016/j.nano.2020.102228Search in Google Scholar PubMed

[283] Conte G, Costabile G, Baldassi D, Rondelli V, Bassi R, Colombo D, et al. Hybrid lipid/polymer nanoparticles to tackle the cystic fibrosis mucus barrier in siRNA delivery to the lungs: does pegylation make the difference? ACS Appl Mater Interfaces. 2022;14:7565–78.10.1021/acsami.1c14975Search in Google Scholar PubMed PubMed Central

[284] monirinasab H, Asadi H, Rostamizadeh K, Esmaeilzadeh A, Khodaei M, Fathi M. Novel lipid-polymer hybrid nanoparticles for siRNA delivery and IGF-1R gene silencing in breast cancer cells. J Drug Deliv Sci Technol. 2018;48:96–105.10.1016/j.jddst.2018.08.025Search in Google Scholar

[285] D'angelo I, Costabile G, Durantie E, Brocca P, Rondelli V, Russo A, et al. Hybrid lipid/polymer nanoparticles for pulmonary delivery of siRNA: Development and fate upon in vitro deposition on the human epithelial airway barrier. J Aerosol Med Pulm Drug Deliv. 2018;31:170–81.10.1089/jamp.2017.1364Search in Google Scholar PubMed

[286] Mekmene O, Quillard S, Rouillon T, Bouler JM, Piot M, Gaucheron F. Effects of pH and Ca/P molar ratio on the quantity and crystalline structure of calcium phosphates obtained from aqueous solutions. Dairy Sci Technol. 2009;89(3):301–16.10.1051/dst/2009019Search in Google Scholar

[287] Tang J, Li L, Howard CB, Mahler SM, Huang L, Xu ZP. Preparation of optimized lipid-coated calcium phosphate nanoparticles for enhanced in vitro gene delivery to breast cancer cells. J Mater Chem B. 2015;3:6805–12.10.1039/C5TB00912JSearch in Google Scholar PubMed PubMed Central

[288] Levingstone TJ, Herbaj S, Dunne NJ. Calcium phosphate nanoparticles for therapeutic applications in bone regeneration. Nanomaterials. 1 November 2019;9(11):1570. 10.3390/NANO9111570.Search in Google Scholar PubMed PubMed Central

[289] Bastari K, Arshath M, Ng ZH, Chia JH, Yow ZX, Sana B, et al. A controlled release of antibiotics from calcium phosphate-coated poly(lactic-co-glycolic acid) particles and their in vitro efficacy against Staphylococcus aureus biofilm. J Mater Sci Mater Med. 2014;25:747–57.10.1007/s10856-013-5125-9Search in Google Scholar PubMed

[290] Hadjicharalambous C, Kozlova D, Sokolova V, Epple M, Chatzinikolaidou M. Calcium phosphate nanoparticles carrying BMP-7 plasmid DNA induce an osteogenic response in MC3T3-E1 pre-osteoblasts. J Biomed Mater Res Part A. 2015;103:3834–42.10.1002/jbm.a.35527Search in Google Scholar PubMed

[291] Bolu B, Sanyal R, Sanyal A. Drug delivery systems from self-assembly of dendron-polymer conjugates. Molecules. 2018;23:1570.10.3390/molecules23071570Search in Google Scholar PubMed PubMed Central

[292] Malkoch M, García-Gallego S. Chapter 1: Introduction to dendrimers and other dendritic polymers. Monogr Supramol Chem. 2020-January;2020:1–20.10.1039/9781788012904-00001Search in Google Scholar

[293] Palmerston Mendes L, Pan J, Torchilin V. Dendrimers as nanocarriers for nucleic acid and drug delivery in cancer therapy. Molecules. 2017;22:1401.10.3390/molecules22091401Search in Google Scholar PubMed PubMed Central

[294] Barba AA, Cascone S, Caccavo D, Lamberti G, Chiarappa G, Abrami M, et al. Engineering approaches in siRNA delivery. Int J Pharm. 2017;525:343–58.10.1016/j.ijpharm.2017.02.032Search in Google Scholar PubMed

[295] Duncan R, Izzo L. Dendrimer biocompatibility and toxicity. Adv Drug Delivery Rev. 2005;57:2215–37.10.1016/j.addr.2005.09.019Search in Google Scholar PubMed

[296] Janaszewska A, Lazniewska J, Trzepiński P, Marcinkowska M, Klajnert-Maculewicz B. Cytotoxicity of dendrimers. Biomolecules. 1 August 2019;9(8):330. 10.3390/biom9080330.Search in Google Scholar PubMed PubMed Central

[297] Somani S, Laskar P, Altwaijry N, Kewcharoenvong P, Irving C, Robb G, et al. PEGylation of polypropylenimine dendrimers: Effects on cytotoxicity, DNA condensation, gene delivery and expression in cancer cells. Sci Rep. 2018;8:9410.10.1038/s41598-018-27400-6Search in Google Scholar PubMed PubMed Central

[298] Kannan R, Prabakaran P, Basu R, Pindi C, Senapati S, Muthuvijayan V, et al. Mechanistic study on the antibacterial activity of self-assembled poly(aryl ether)-based amphiphilic dendrimers. ACS Appl Bio Mater. 2019;2:3212–24.10.1021/acsabm.9b00140Search in Google Scholar PubMed

[299] Valikala V, Santhakumar I, Kannappan S. Synthesis and effect of pegylation on citric acid dendritic nano architectures anchored with cefotaxime sodium. J Photochem Photobiol B Biol. 2019;201:111683.10.1016/j.jphotobiol.2019.111683Search in Google Scholar PubMed

[300] Relaño-Rodríguez I, Espinar-Buitrago MS, Martín-Cañadilla V, Gómez-Ramirez R, Jiménez JL, Muñoz-Fernández MA. Nanotechnology against human cytomegalovirus in vitro: polyanionic carbosilane dendrimers as antiviral agents. J Nanobiotechnol. 2021;19:65.10.1186/s12951-021-00809-4Search in Google Scholar PubMed PubMed Central

[301] Günther SC, Maier JD, Vetter J, Podvalnyy N, Khanzhin N, Hennet T, et al. Antiviral potential of 3′-sialyllactose- and 6′-sialyllactose-conjugated dendritic polymers against human and avian influenza viruses. Sci Rep. 2020;10:1–9.10.1038/s41598-020-57608-4Search in Google Scholar PubMed PubMed Central

[302] Dzmitruk V, Apartsin E, Ihnatsyeu-Kachan A, Abashkin V, Shcharbin D, Bryszewska M. Dendrimers Show Promise for siRNA and microRNA therapeutics. Pharmaceutics. 2018;10:126.10.3390/pharmaceutics10030126Search in Google Scholar PubMed PubMed Central

[303] Mhlwatika Z, Aderibigbe B. Application of dendrimers for the treatment of infectious diseases. Molecules. 2018;23:2205.10.3390/molecules23092205Search in Google Scholar PubMed PubMed Central

[304] Martí Coma-Cros E, Lancelot A, San Anselmo M, Neves Borgheti-Cardoso L, Valle-Delgado JJ, Serrano JL, et al. Micelle carriers based on dendritic macromolecules containing bis-MPA and glycine for antimalarial drug delivery. Biomater Sci. 2019;7:1661–74.10.1039/C8BM01600CSearch in Google Scholar PubMed

[305] Mehrizi TZ, Ardestani MS, Khamesipour A, Hoseini M, Mosaffa N, Anissian A, et al. Reduction toxicity of Amphotericin B through loading into a novel nanoformulation of anionic linear globular dendrimer for improve treatment of leishmania major. J Mater Sci Mater Med. 1 August 2018;29(8):125. 10.1007/s10856-018-6122-9.Search in Google Scholar PubMed

[306] Ho MN, Bach LG, Nguyen TH, Ho MH, Nguyen DH, Nguyen CK, et al. PEGylated poly(amidoamine) dendrimers-based drug loading vehicles for delivering carboplatin in treatment of various cancerous cells. J Nanopart Res. 2019;21:1–12.10.1007/s11051-019-4486-5Search in Google Scholar

[307] Relaño-Rodríguez I, Muñoz-Fernández MÁ. Emergence of nanotechnology to fight HIV sexual transmission: The trip of G2-S16 polyanionic carbosilane dendrimer to possible pre-clinical trials. Int J Mol Sci. 2020;21:1–20.10.3390/ijms21249403Search in Google Scholar PubMed PubMed Central

[308] Guerrero-Beltrán C, Garcia-Heredia I, Ceña-Diez R, Rodriguez-Izquierdo I, Serramía MJ, Martinez-Hernandez F, et al. Cationic dendrimer g2-s16 inhibits herpes simplex type 2 infection and protects mice vaginal microbiome. Pharmaceutics. 2020 Jun 4;12(6):515. 10.3390/pharmaceutics12060515.Search in Google Scholar PubMed PubMed Central

[309] Lu Y, Han S, Zheng H, Ma R, Ping Y, Zou J, et al. A novel RGDyC/PEG co-modified PAMAM dendrimer-loaded arsenic trioxide of glioma targeting delivery system. Int J Nanomed. 2018;13:5937–52.10.2147/IJN.S175418Search in Google Scholar PubMed PubMed Central

[310] Fana M, Gallien J, Srinageshwar B, Dunbar GL, Rossignol J. PAMAM dendrimer nanomolecules utilized as drug delivery systems for potential treatment of glioblastoma: a systematic review. Int J Nanomed. 2020;15:2789–808.10.2147/IJN.S243155Search in Google Scholar PubMed PubMed Central

[311] Gorain B, Tekade M, Kesharwani P, Iyer AK, Kalia K, Tekade RK. The use of nanoscaffolds and dendrimers in tissue engineering. Drug Discovery Today. 2017;22:652–64.10.1016/j.drudis.2016.12.007Search in Google Scholar PubMed

[312] Pistone A, Iannazzo D, Celesti C, et al. Chitosan/PAMAM/Hydroxyapatite engineered drug release hydrogels with tunable rheological properties. Polym (Basel). 2020;12. 10.3390/polym12040754.Search in Google Scholar PubMed PubMed Central

[313] El-Say KM, El-Sawy HS. Polymeric nanoparticles: Promising platform for drug delivery. Int J Pharmaceutics. 2017;528:675–91.10.1016/j.ijpharm.2017.06.052Search in Google Scholar PubMed

[314] Zielińska A, Carreiró F, Oliveira AM, Neves A, Pires B, Venkatesh DN, et al. Polymeric nanoparticles: production, characterization, toxicology and ecotoxicology. Molecules. 2020;25:3731.10.3390/molecules25163731Search in Google Scholar PubMed PubMed Central

[315] Lee S, Lee K. Ph-sensitive folic acid conjugated alginate nanoparticle for induction of cancer-specific fluorescence imaging. Pharmaceutics. 2020;12:1–14.10.3390/pharmaceutics12060537Search in Google Scholar PubMed PubMed Central

[316] An FF, Zhang XH. Strategies for preparing albumin-based nanoparticles for multifunctional bioimaging and drug delivery. Theranostics. 2017;7:3667–89.10.7150/thno.19365Search in Google Scholar PubMed PubMed Central

[317] Radwan RR, Ali HE. Radiation-synthesis of chitosan/poly (acrylic acid) nanogel for improving the antitumor potential of rutin in hepatocellular carcinoma. Drug Deliv Transl Res. 2021;11:261–78.10.1007/s13346-020-00792-7Search in Google Scholar PubMed

[318] Spicer CD, Jumeaux C, Gupta B, Stevens MM. Peptide and protein nanoparticle conjugates: Versatile platforms for biomedical applications. Chem Soc Rev. 2018;47:3574–620.10.1039/C7CS00877ESearch in Google Scholar PubMed PubMed Central

[319] Liang H, Huang Q, Zhou B, He L, Lin L, An Y, et al. Cellulose nanoparticles: structure-morphology-rheology relationships. ACS Sustain Chem Eng. 2015;3:821–32.10.1021/acssuschemeng.5b00144Search in Google Scholar

[320] Mauro N, Campora S, Scialabba C, Adamo G, Licciardi M, Ghersi G, et al. Self-organized environment-sensitive inulin-doxorubicin conjugate with a selective cytotoxic effect towards cancer cells. RSC Adv. 2015;5(41):32421–30. 10.1039/c5ra00287g.Search in Google Scholar

[321] Azimi B, Nourpanah P, Rabiee M, Arbab S. Producing gelatin nanoparticles as delivery system for bovine serum albumin. Iran Biomed J. 2013;18:34–40.Search in Google Scholar

[322] Rezvantalab S, Drude NI, Moraveji MK, Güvener N, Koons EK, Shi Y, et al. PLGA-based nanoparticles in cancer treatment. Front Pharmacol. 2 November 2018;9:1260. 10.3389/fphar.2018.01260.Search in Google Scholar PubMed PubMed Central

[323] Silva AL, Soema PC, Slütter B, Ossendorp F, Jiskoot W. PLGA particulate delivery systems for subunit vaccines: Linking particle properties to immunogenicity. Hum Vaccines Immunotherapeutics. 2016;12:1056–69.10.1080/21645515.2015.1117714Search in Google Scholar PubMed PubMed Central

[324] Khalil IR, Burns AT, Radecka I, Kowalczuk M, Khalaf T, Adamus G, et al. Bacterial-derived polymer poly-γ-glutamic acid (γ-PGA)-based micro/nanoparticles as a delivery system for antimicrobials and other biomedical applications. Int J Mol Sci. 2 February 2017;18(2):313. 10.3390/ijms18020313.Search in Google Scholar PubMed PubMed Central

[325] Arif M, Dong QJ, Raja MA, Zeenat S, Chi Z, Liu CG. Development of novel pH-sensitive thiolated chitosan/PMLA nanoparticles for amoxicillin delivery to treat Helicobacter pylori. Mater Sci Eng C. 2018;83:17–24.10.1016/j.msec.2017.08.038Search in Google Scholar PubMed

[326] Mara D, Kaczmarek AM, Artizzu F, Abalymov A, Skirtach AG, Van Hecke K, et al. Luminescent PMMA films and PMMA@SiO2 nanoparticles with embedded Ln3+ complexes for highly sensitive optical thermometers in the physiological temperature range. Chem – A Eur J. 2021;27:6479–88.10.1002/chem.202004951Search in Google Scholar PubMed

[327] Tamsilian Y, Ramazani S.A. A, Shaban M, Ayatollahi S, Tomovska R. High molecular weight polyacrylamide nanoparticles prepared by inverse emulsion polymerization: reaction conditions-properties relationships. Colloid Polym Sci. 2016;294:513–25.10.1007/s00396-015-3803-5Search in Google Scholar

[328] Adamoa G, Grimaldib N, Camporaa S, Antonietta M, Sabatinob CD, Ghersia G. Glutathione-sensitive nanogels for drug release. Chem Eng Trans. 2014;38:457–62. 10.3303/CET1438077.Search in Google Scholar

[329] Li H, Palamoor M, Jablonski MM. Poly(ortho ester) nanoparticles targeted for chronic intraocular diseases: ocular safety and localization after intravitreal injection. Nanotoxicology. 2016;10:1152–9.10.1080/17435390.2016.1181808Search in Google Scholar PubMed PubMed Central

[330] Yin Y, Li Y, Wang S, Dong Z, Liang C, Sun J, et al. MSCs-engineered biomimetic PMAA nanomedicines for multiple bioimaging-guided and photothermal-enhanced radiotherapy of NSCLC. J Nanobiotechnol. 2021;19:80.10.1186/s12951-021-00823-6Search in Google Scholar PubMed PubMed Central

[331] Lanzalaco S, Campora S, Brucato V, Carfì Pavia F, Di Leonardo ER, Ghersi G, et al. Sterilization of macroscopic poly(l-lactic acid) porous scaffolds with dense carbon dioxide: Investigation of the spatial penetration of the treatment and of its effect on the properties of the matrix. J Supercrit Fluids. 2016;111:83–90. 10.1016/j.supflu.2016.01.014.Search in Google Scholar

[332] Beibei D, Tiantang F, Jiafeng L, Li G, Qin Z, Wuyou Y, et al. PLLA-grafted gelatin amphiphilic copolymer and its self-assembled nano carrier for anticancer drug delivery. Macromol Chem Phys. 2019;220:1800528.10.1002/macp.201800528Search in Google Scholar

[333] Carfì Pavia F, Conoscenti G, Greco S, La Carrubba V, Ghersi G, Brucato V. Preparation, characterization and in vitro test of composites poly-lactic acid/hydroxyapatite scaffolds for bone tissue engineering. Int J Biol Macromol. 2018;119:945–53.10.1016/j.ijbiomac.2018.08.007Search in Google Scholar PubMed

[334] Campora S, Mohsen R, Passaro D, Samir H, Ashraf H, Al-Mofty SE, et al. Functionalized poly(N-isopropylacrylamide)-based microgels in tumor targeting and drug delivery. Gels. 2021;7:203.10.3390/gels7040203Search in Google Scholar PubMed PubMed Central

[335] Banik BL, Fattahi P, Brown JL. Polymeric nanoparticles: The future of nanomedicine. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2016;8:271–99.10.1002/wnan.1364Search in Google Scholar PubMed

[336] Di Prima G, Saladino S, Bongiovì F, Adamo G, Ghersi G, Pitarresi G, et al. Novel inulin-based mucoadhesive micelles loaded with corticosteroids as potential transcorneal permeation enhancers. Eur J Pharm Biopharm. 2017;117:385–99.10.1016/j.ejpb.2017.05.005Search in Google Scholar PubMed

[337] Adamo G, Grimaldi N, Campora S, Bulone D, Bondì ML, Al-Sheikhly M, et al. Multi-functional nanogels for tumor targeting and redox-sensitive drug and siRNA delivery. Molecules. 2016;21(11):1594. 10.3390/molecules21111594.Search in Google Scholar PubMed PubMed Central

[338] Dispenza C, Sabatino MA, Grimaldi N<et al. Large-scale radiation manufacturing of hierarchically assembled nanogels. Chemical engineering transactions. Italian ASSOCIATION OF CHEMICAL ENGINeering – AIDIC. p. 229–34Search in Google Scholar

[339] 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. 10.3390/jfb10010004.Search in Google Scholar PubMed PubMed Central

[340] Carradori D, Balducci C, Re F, Brambilla D, Le Droumaguet B, Flores O, et al. Antibody-functionalized polymer nanoparticle leading to memory recovery in Alzheimer’s disease-like transgenic mouse model. Nanomed Nanotechnol Biol Med. 2018;14:609–18.10.1016/j.nano.2017.12.006Search in Google Scholar PubMed

[341] Tan JPK, Voo ZX, Lim S, Venkataraman S, Ng KM, Gao S, et al. Effective encapsulation of apomorphine into biodegradable polymeric nanoparticles through a reversible chemical bond for delivery across the blood–brain barrier. Nanomed Nanotechnol Biol Med. 2019;17:236–45.10.1016/j.nano.2019.01.014Search in Google Scholar PubMed

[342] Lu X, Zhang Y, Wang L, Li G, Gao J, Wang Y. Development of L-carnosine functionalized iron oxide nanoparticles loaded with dexamethasone for simultaneous therapeutic potential of blood brain barrier crossing and ischemic stroke treatment. Drug Deliv. 2021;28:380–9.10.1080/10717544.2021.1883158Search in Google Scholar PubMed PubMed Central

[343] Zamanlu M, Eskandani M, Barar J, Jaymand M, Pakchin PS, Farhoudi M. Enhanced thrombolysis using tissue plasminogen activator (tPA)-loaded PEGylated PLGA nanoparticles for ischemic stroke. J Drug Deliv Sci Technol. 1 October 2019;53. 10.1016/j.jddst.2019.101165.Search in Google Scholar

[344] Sokolova V, Kostka K, Shalumon KT, Prymak O, Chen JP, Epple M. Synthesis and characterization of PLGA/HAP scaffolds with DNA-functionalised calcium phosphate nanoparticles for bone tissue engineering. J Mater Sci Mater Med. 2020;31:1–12.10.1007/s10856-020-06442-1Search in Google Scholar PubMed PubMed Central

[345] Park JS, Yi SW, Kim HJ, Kim SM, Park KH. Regulation of cell signaling factors using PLGA nanoparticles coated/loaded with genes and proteins for osteogenesis of human mesenchymal stem cells. ACS Appl Mater Interfaces. 2016;8:30387–97.10.1021/acsami.6b08343Search in Google Scholar PubMed

[346] Gu P, Wusiman A, Zhang Y, Liu Z, Bo R, Hu Y, et al. Rational design of PLGA nanoparticle vaccine delivery systems to improve immune responses. Mol Pharm. 2019;16:5000–12.10.1021/acs.molpharmaceut.9b00860Search in Google Scholar PubMed

[347] Kaneko K, Miyaji EN, Gonçalves VM, Ferreira DM, Solórzano C, MacLoughlin R, et al. Evaluation of polymer choice on immunogenicity of chitosan coated PLGA NPs with surface-adsorbed pneumococcal protein antigen PspA4Pro. Int J Pharm. 2021;599:120407.10.1016/j.ijpharm.2021.120407Search in Google Scholar PubMed PubMed Central

[348] Adamo G, Campora S, Ghersi G. Functionalization of nanoparticles in specific targeting and mechanism release. Nanostructures for novel therapy; Bucharest, Romania: Synthesis, Characterization and Applications Micro and Nano Technologies. 2017. p. 57–80. 10.1016/B978-0-323-46142-9.00003-7.Search in Google Scholar

[349] Seaberg J, Montazerian H, Hossen MN, Bhattacharya R, Khademhosseini A, Mukherjee P. Hybrid nanosystems for biomedical applications. ACS Nano. 2021;15:2099–142.10.1021/acsnano.0c09382Search in Google Scholar PubMed

[350] Li YF, Zhang HT, Xin L. Hyaluronic acid-modified polyamidoamine dendrimer G5-entrapped gold nanoparticles delivering METase gene inhibits gastric tumor growth via targeting CD44+ gastric cancer cells. J Cancer Res Clin Oncol. 2018;144:1463–73.10.1007/s00432-018-2678-5Search in Google Scholar PubMed

[351] Zhuang J, Gong H, Zhou J, Zhang Q, Gao W, Fang RH, et al. Targeted gene silencing in vivo by platelet membrane-coated metal-organic framework nanoparticles. Sci Adv. 2020 Mar 27;613:eaaz6108. 10.1126/SCIADV.AAZ6108/ASSET/E4F34B18-E6CB-4714-BA45-3C49A38DDDAC/ASSETS/GRAPHIC/AAZ6108-F6.JPEG.Search in Google Scholar

[352] Abdellatif AAH, Alsowinea AF. Approved and marketed nanoparticles for disease targeting and applications in COVID-19. Nanotechnol Rev. 2021;2021(10):1941–77.10.1515/ntrev-2021-0115Search in Google Scholar

[353] Farjadian F, Ghasemi A, Gohari O, Roointan A, Karimi M, Hamblin MR. Nanopharmaceuticals and nanomedicines currently on the market: challenges and opportunities. Nanomed (Lond). 2019;14:93–126.10.2217/nnm-2018-0120Search in Google Scholar PubMed PubMed Central

[354] Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2020;20:101–24.10.1038/s41573-020-0090-8Search in Google Scholar PubMed PubMed Central

Received: 2021-11-03
Revised: 2022-04-15
Accepted: 2022-05-01
Published Online: 2022-07-11

© 2022 Simona Campora and Giulio Ghersi, published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Research Articles
  2. Theoretical and experimental investigation of MWCNT dispersion effect on the elastic modulus of flexible PDMS/MWCNT nanocomposites
  3. Mechanical, morphological, and fracture-deformation behavior of MWCNTs-reinforced (Al–Cu–Mg–T351) alloy cast nanocomposites fabricated by optimized mechanical milling and powder metallurgy techniques
  4. Flammability and physical stability of sugar palm crystalline nanocellulose reinforced thermoplastic sugar palm starch/poly(lactic acid) blend bionanocomposites
  5. Glutathione-loaded non-ionic surfactant niosomes: A new approach to improve oral bioavailability and hepatoprotective efficacy of glutathione
  6. Relationship between mechano-bactericidal activity and nanoblades density on chemically strengthened glass
  7. In situ regulation of microstructure and microwave-absorbing properties of FeSiAl through HNO3 oxidation
  8. Research on a mechanical model of magnetorheological fluid different diameter particles
  9. Nanomechanical and dynamic mechanical properties of rubber–wood–plastic composites
  10. Investigative properties of CeO2 doped with niobium: A combined characterization and DFT studies
  11. Miniaturized peptidomimetics and nano-vesiculation in endothelin types through probable nano-disk formation and structure property relationships of endothelins’ fragments
  12. N/S co-doped CoSe/C nanocubes as anode materials for Li-ion batteries
  13. Synergistic effects of halloysite nanotubes with metal and phosphorus additives on the optimal design of eco-friendly sandwich panels with maximum flame resistance and minimum weight
  14. Octreotide-conjugated silver nanoparticles for active targeting of somatostatin receptors and their application in a nebulized rat model
  15. Controllable morphology of Bi2S3 nanostructures formed via hydrothermal vulcanization of Bi2O3 thin-film layer and their photoelectrocatalytic performances
  16. Development of (−)-epigallocatechin-3-gallate-loaded folate receptor-targeted nanoparticles for prostate cancer treatment
  17. Enhancement of the mechanical properties of HDPE mineral nanocomposites by filler particles modulation of the matrix plastic/elastic behavior
  18. Effect of plasticizers on the properties of sugar palm nanocellulose/cinnamon essential oil reinforced starch bionanocomposite films
  19. Optimization of nano coating to reduce the thermal deformation of ball screws
  20. Preparation of efficient piezoelectric PVDF–HFP/Ni composite films by high electric field poling
  21. MHD dissipative Casson nanofluid liquid film flow due to an unsteady stretching sheet with radiation influence and slip velocity phenomenon
  22. Effects of nano-SiO2 modification on rubberised mortar and concrete with recycled coarse aggregates
  23. Mechanical and microscopic properties of fiber-reinforced coal gangue-based geopolymer concrete
  24. Effect of morphology and size on the thermodynamic stability of cerium oxide nanoparticles: Experiment and molecular dynamics calculation
  25. Mechanical performance of a CFRP composite reinforced via gelatin-CNTs: A study on fiber interfacial enhancement and matrix enhancement
  26. A practical review over surface modification, nanopatterns, emerging materials, drug delivery systems, and their biophysiochemical properties for dental implants: Recent progresses and advances
  27. HTR: An ultra-high speed algorithm for cage recognition of clathrate hydrates
  28. Effects of microalloying elements added by in situ synthesis on the microstructure of WCu composites
  29. A highly sensitive nanobiosensor based on aptamer-conjugated graphene-decorated rhodium nanoparticles for detection of HER2-positive circulating tumor cells
  30. Progressive collapse performance of shear strengthened RC frames by nano CFRP
  31. Core–shell heterostructured composites of carbon nanotubes and imine-linked hyperbranched polymers as metal-free Li-ion anodes
  32. A Galerkin strategy for tri-hybridized mixture in ethylene glycol comprising variable diffusion and thermal conductivity using non-Fourier’s theory
  33. Simple models for tensile modulus of shape memory polymer nanocomposites at ambient temperature
  34. Preparation and morphological studies of tin sulfide nanoparticles and use as efficient photocatalysts for the degradation of rhodamine B and phenol
  35. Polyethyleneimine-impregnated activated carbon nanofiber composited graphene-derived rice husk char for efficient post-combustion CO2 capture
  36. Electrospun nanofibers of Co3O4 nanocrystals encapsulated in cyclized-polyacrylonitrile for lithium storage
  37. Pitting corrosion induced on high-strength high carbon steel wire in high alkaline deaerated chloride electrolyte
  38. Formulation of polymeric nanoparticles loaded sorafenib; evaluation of cytotoxicity, molecular evaluation, and gene expression studies in lung and breast cancer cell lines
  39. Engineered nanocomposites in asphalt binders
  40. Influence of loading voltage, domain ratio, and additional load on the actuation of dielectric elastomer
  41. Thermally induced hex-graphene transitions in 2D carbon crystals
  42. The surface modification effect on the interfacial properties of glass fiber-reinforced epoxy: A molecular dynamics study
  43. Molecular dynamics study of deformation mechanism of interfacial microzone of Cu/Al2Cu/Al composites under tension
  44. Nanocolloid simulators of luminescent solar concentrator photovoltaic windows
  45. Compressive strength and anti-chloride ion penetration assessment of geopolymer mortar merging PVA fiber and nano-SiO2 using RBF–BP composite neural network
  46. Effect of 3-mercapto-1-propane sulfonate sulfonic acid and polyvinylpyrrolidone on the growth of cobalt pillar by electrodeposition
  47. Dynamics of convective slippery constraints on hybrid radiative Sutterby nanofluid flow by Galerkin finite element simulation
  48. Preparation of vanadium by the magnesiothermic self-propagating reduction and process control
  49. Microstructure-dependent photoelectrocatalytic activity of heterogeneous ZnO–ZnS nanosheets
  50. Cytotoxic and pro-inflammatory effects of molybdenum and tungsten disulphide on human bronchial cells
  51. Improving recycled aggregate concrete by compression casting and nano-silica
  52. Chemically reactive Maxwell nanoliquid flow by a stretching surface in the frames of Newtonian heating, nonlinear convection and radiative flux: Nanopolymer flow processing simulation
  53. Nonlinear dynamic and crack behaviors of carbon nanotubes-reinforced composites with various geometries
  54. Biosynthesis of copper oxide nanoparticles and its therapeutic efficacy against colon cancer
  55. Synthesis and characterization of smart stimuli-responsive herbal drug-encapsulated nanoniosome particles for efficient treatment of breast cancer
  56. Homotopic simulation for heat transport phenomenon of the Burgers nanofluids flow over a stretching cylinder with thermal convective and zero mass flux conditions
  57. Incorporation of copper and strontium ions in TiO2 nanotubes via dopamine to enhance hemocompatibility and cytocompatibility
  58. Mechanical, thermal, and barrier properties of starch films incorporated with chitosan nanoparticles
  59. Mechanical properties and microstructure of nano-strengthened recycled aggregate concrete
  60. Glucose-responsive nanogels efficiently maintain the stability and activity of therapeutic enzymes
  61. Tunning matrix rheology and mechanical performance of ultra-high performance concrete using cellulose nanofibers
  62. Flexible MXene/copper/cellulose nanofiber heat spreader films with enhanced thermal conductivity
  63. Promoted charge separation and specific surface area via interlacing of N-doped titanium dioxide nanotubes on carbon nitride nanosheets for photocatalytic degradation of Rhodamine B
  64. Elucidating the role of silicon dioxide and titanium dioxide nanoparticles in mitigating the disease of the eggplant caused by Phomopsis vexans, Ralstonia solanacearum, and root-knot nematode Meloidogyne incognita
  65. An implication of magnetic dipole in Carreau Yasuda liquid influenced by engine oil using ternary hybrid nanomaterial
  66. Robust synthesis of a composite phase of copper vanadium oxide with enhanced performance for durable aqueous Zn-ion batteries
  67. Tunning self-assembled phases of bovine serum albumin via hydrothermal process to synthesize novel functional hydrogel for skin protection against UVB
  68. A comparative experimental study on damping properties of epoxy nanocomposite beams reinforced with carbon nanotubes and graphene nanoplatelets
  69. Lightweight and hydrophobic Ni/GO/PVA composite aerogels for ultrahigh performance electromagnetic interference shielding
  70. Research on the auxetic behavior and mechanical properties of periodically rotating graphene nanostructures
  71. Repairing performances of novel cement mortar modified with graphene oxide and polyacrylate polymer
  72. Closed-loop recycling and fabrication of hydrophilic CNT films with high performance
  73. Design of thin-film configuration of SnO2–Ag2O composites for NO2 gas-sensing applications
  74. Study on stress distribution of SiC/Al composites based on microstructure models with microns and nanoparticles
  75. PVDF green nanofibers as potential carriers for improving self-healing and mechanical properties of carbon fiber/epoxy prepregs
  76. Osteogenesis capability of three-dimensionally printed poly(lactic acid)-halloysite nanotube scaffolds containing strontium ranelate
  77. Silver nanoparticles induce mitochondria-dependent apoptosis and late non-canonical autophagy in HT-29 colon cancer cells
  78. Preparation and bonding mechanisms of polymer/metal hybrid composite by nano molding technology
  79. Damage self-sensing and strain monitoring of glass-reinforced epoxy composite impregnated with graphene nanoplatelet and multiwalled carbon nanotubes
  80. Thermal analysis characterisation of solar-powered ship using Oldroyd hybrid nanofluids in parabolic trough solar collector: An optimal thermal application
  81. Pyrene-functionalized halloysite nanotubes for simultaneously detecting and separating Hg(ii) in aqueous media: A comprehensive comparison on interparticle and intraparticle excimers
  82. Fabrication of self-assembly CNT flexible film and its piezoresistive sensing behaviors
  83. Thermal valuation and entropy inspection of second-grade nanoscale fluid flow over a stretching surface by applying Koo–Kleinstreuer–Li relation
  84. Mechanical properties and microstructure of nano-SiO2 and basalt-fiber-reinforced recycled aggregate concrete
  85. Characterization and tribology performance of polyaniline-coated nanodiamond lubricant additives
  86. Combined impact of Marangoni convection and thermophoretic particle deposition on chemically reactive transport of nanofluid flow over a stretching surface
  87. Spark plasma extrusion of binder free hydroxyapatite powder
  88. An investigation on thermo-mechanical performance of graphene-oxide-reinforced shape memory polymer
  89. Effect of nanoadditives on the novel leather fiber/recycled poly(ethylene-vinyl-acetate) polymer composites for multifunctional applications: Fabrication, characterizations, and multiobjective optimization using central composite design
  90. Design selection for a hemispherical dimple core sandwich panel using hybrid multi-criteria decision-making methods
  91. Improving tensile strength and impact toughness of plasticized poly(lactic acid) biocomposites by incorporating nanofibrillated cellulose
  92. Green synthesis of spinel copper ferrite (CuFe2O4) nanoparticles and their toxicity
  93. The effect of TaC and NbC hybrid and mono-nanoparticles on AA2024 nanocomposites: Microstructure, strengthening, and artificial aging
  94. Excited-state geometry relaxation of pyrene-modified cellulose nanocrystals under UV-light excitation for detecting Fe3+
  95. Effect of CNTs and MEA on the creep of face-slab concrete at an early age
  96. Effect of deformation conditions on compression phase transformation of AZ31
  97. Application of MXene as a new generation of highly conductive coating materials for electromembrane-surrounded solid-phase microextraction
  98. A comparative study of the elasto-plastic properties for ceramic nanocomposites filled by graphene or graphene oxide nanoplates
  99. Encapsulation strategies for improving the biological behavior of CdS@ZIF-8 nanocomposites
  100. Biosynthesis of ZnO NPs from pumpkin seeds’ extract and elucidation of its anticancer potential against breast cancer
  101. Preliminary trials of the gold nanoparticles conjugated chrysin: An assessment of anti-oxidant, anti-microbial, and in vitro cytotoxic activities of a nanoformulated flavonoid
  102. Effect of micron-scale pores increased by nano-SiO2 sol modification on the strength of cement mortar
  103. Fractional simulations for thermal flow of hybrid nanofluid with aluminum oxide and titanium oxide nanoparticles with water and blood base fluids
  104. The effect of graphene nano-powder on the viscosity of water: An experimental study and artificial neural network modeling
  105. Development of a novel heat- and shear-resistant nano-silica gelling agent
  106. Characterization, biocompatibility and in vivo of nominal MnO2-containing wollastonite glass-ceramic
  107. Entropy production simulation of second-grade magnetic nanomaterials flowing across an expanding surface with viscidness dissipative flux
  108. Enhancement in structural, morphological, and optical properties of copper oxide for optoelectronic device applications
  109. Aptamer-functionalized chitosan-coated gold nanoparticle complex as a suitable targeted drug carrier for improved breast cancer treatment
  110. Performance and overall evaluation of nano-alumina-modified asphalt mixture
  111. Analysis of pure nanofluid (GO/engine oil) and hybrid nanofluid (GO–Fe3O4/engine oil): Novel thermal and magnetic features
  112. Synthesis of Ag@AgCl modified anatase/rutile/brookite mixed phase TiO2 and their photocatalytic property
  113. Mechanisms and influential variables on the abrasion resistance hydraulic concrete
  114. Synergistic reinforcement mechanism of basalt fiber/cellulose nanocrystals/polypropylene composites
  115. Achieving excellent oxidation resistance and mechanical properties of TiB2–B4C/carbon aerogel composites by quick-gelation and mechanical mixing
  116. Microwave-assisted sol–gel template-free synthesis and characterization of silica nanoparticles obtained from South African coal fly ash
  117. Pulsed laser-assisted synthesis of nano nickel(ii) oxide-anchored graphitic carbon nitride: Characterizations and their potential antibacterial/anti-biofilm applications
  118. Effects of nano-ZrSi2 on thermal stability of phenolic resin and thermal reusability of quartz–phenolic composites
  119. Benzaldehyde derivatives on tin electroplating as corrosion resistance for fabricating copper circuit
  120. Mechanical and heat transfer properties of 4D-printed shape memory graphene oxide/epoxy acrylate composites
  121. Coupling the vanadium-induced amorphous/crystalline NiFe2O4 with phosphide heterojunction toward active oxygen evolution reaction catalysts
  122. Graphene-oxide-reinforced cement composites mechanical and microstructural characteristics at elevated temperatures
  123. Gray correlation analysis of factors influencing compressive strength and durability of nano-SiO2 and PVA fiber reinforced geopolymer mortar
  124. Preparation of layered gradient Cu–Cr–Ti alloy with excellent mechanical properties, thermal stability, and electrical conductivity
  125. Recovery of Cr from chrome-containing leather wastes to develop aluminum-based composite material along with Al2O3 ceramic particles: An ingenious approach
  126. Mechanisms of the improved stiffness of flexible polymers under impact loading
  127. Anticancer potential of gold nanoparticles (AuNPs) using a battery of in vitro tests
  128. Review Articles
  129. Proposed approaches for coronaviruses elimination from wastewater: Membrane techniques and nanotechnology solutions
  130. Application of Pickering emulsion in oil drilling and production
  131. The contribution of microfluidics to the fight against tuberculosis
  132. Graphene-based biosensors for disease theranostics: Development, applications, and recent advancements
  133. Synthesis and encapsulation of iron oxide nanorods for application in magnetic hyperthermia and photothermal therapy
  134. Contemporary nano-architectured drugs and leads for ανβ3 integrin-based chemotherapy: Rationale and retrospect
  135. State-of-the-art review of fabrication, application, and mechanical properties of functionally graded porous nanocomposite materials
  136. Insights on magnetic spinel ferrites for targeted drug delivery and hyperthermia applications
  137. A review on heterogeneous oxidation of acetaminophen based on micro and nanoparticles catalyzed by different activators
  138. Early diagnosis of lung cancer using magnetic nanoparticles-integrated systems
  139. Advances in ZnO: Manipulation of defects for enhancing their technological potentials
  140. Efficacious nanomedicine track toward combating COVID-19
  141. A review of the design, processes, and properties of Mg-based composites
  142. Green synthesis of nanoparticles for varied applications: Green renewable resources and energy-efficient synthetic routes
  143. Two-dimensional nanomaterial-based polymer composites: Fundamentals and applications
  144. Recent progress and challenges in plasmonic nanomaterials
  145. Apoptotic cell-derived micro/nanosized extracellular vesicles in tissue regeneration
  146. Electronic noses based on metal oxide nanowires: A review
  147. Framework materials for supercapacitors
  148. An overview on the reproductive toxicity of graphene derivatives: Highlighting the importance
  149. Antibacterial nanomaterials: Upcoming hope to overcome antibiotic resistance crisis
  150. Research progress of carbon materials in the field of three-dimensional printing polymer nanocomposites
  151. A review of atomic layer deposition modelling and simulation methodologies: Density functional theory and molecular dynamics
  152. Recent advances in the preparation of PVDF-based piezoelectric materials
  153. Recent developments in tensile properties of friction welding of carbon fiber-reinforced composite: A review
  154. Comprehensive review of the properties of fly ash-based geopolymer with additive of nano-SiO2
  155. Perspectives in biopolymer/graphene-based composite application: Advances, challenges, and recommendations
  156. Graphene-based nanocomposite using new modeling molecular dynamic simulations for proposed neutralizing mechanism and real-time sensing of COVID-19
  157. Nanotechnology application on bamboo materials: A review
  158. Recent developments and future perspectives of biorenewable nanocomposites for advanced applications
  159. Nanostructured lipid carrier system: A compendium of their formulation development approaches, optimization strategies by quality by design, and recent applications in drug delivery
  160. 3D printing customized design of human bone tissue implant and its application
  161. Design, preparation, and functionalization of nanobiomaterials for enhanced efficacy in current and future biomedical applications
  162. A brief review of nanoparticles-doped PEDOT:PSS nanocomposite for OLED and OPV
  163. Nanotechnology interventions as a putative tool for the treatment of dental afflictions
  164. Recent advancements in metal–organic frameworks integrating quantum dots (QDs@MOF) and their potential applications
  165. A focused review of short electrospun nanofiber preparation techniques for composite reinforcement
  166. Microstructural characteristics and nano-modification of interfacial transition zone in concrete: A review
  167. Latest developments in the upconversion nanotechnology for the rapid detection of food safety: A review
  168. Strategic applications of nano-fertilizers for sustainable agriculture: Benefits and bottlenecks
  169. Molecular dynamics application of cocrystal energetic materials: A review
  170. Synthesis and application of nanometer hydroxyapatite in biomedicine
  171. Cutting-edge development in waste-recycled nanomaterials for energy storage and conversion applications
  172. Biological applications of ternary quantum dots: A review
  173. Nanotherapeutics for hydrogen sulfide-involved treatment: An emerging approach for cancer therapy
  174. Application of antibacterial nanoparticles in orthodontic materials
  175. Effect of natural-based biological hydrogels combined with growth factors on skin wound healing
  176. Nanozymes – A route to overcome microbial resistance: A viewpoint
  177. Recent developments and applications of smart nanoparticles in biomedicine
  178. Contemporary review on carbon nanotube (CNT) composites and their impact on multifarious applications
  179. Interfacial interactions and reinforcing mechanisms of cellulose and chitin nanomaterials and starch derivatives for cement and concrete strength and durability enhancement: A review
  180. Diamond-like carbon films for tribological modification of rubber
  181. Layered double hydroxides (LDHs) modified cement-based materials: A systematic review
  182. Recent research progress and advanced applications of silica/polymer nanocomposites
  183. Modeling of supramolecular biopolymers: Leading the in silico revolution of tissue engineering and nanomedicine
  184. Recent advances in perovskites-based optoelectronics
  185. Biogenic synthesis of palladium nanoparticles: New production methods and applications
  186. A comprehensive review of nanofluids with fractional derivatives: Modeling and application
  187. Electrospinning of marine polysaccharides: Processing and chemical aspects, challenges, and future prospects
  188. Electrohydrodynamic printing for demanding devices: A review of processing and applications
  189. Rapid Communications
  190. Structural material with designed thermal twist for a simple actuation
  191. Recent advances in photothermal materials for solar-driven crude oil adsorption
Downloaded on 1.10.2025 from https://www.degruyterbrill.com/document/doi/10.1515/ntrev-2022-0148/html?lang=en&srsltid=AfmBOopeo_hFMUxVNB6q9oGJGTLwlS8EB3CqNmM236HAV6YViW8CnXSq
Scroll to top button