v
Search
Advanced

Publications > Journals > Journal of Exploratory Research in Pharmacology> Article Full Text

  • OPEN ACCESS

The Combination of Cutting-edge Strategies in Nano-delivery Systems to Overcome Drawbacks for Malignant Tumor Treatment

  • Robson Roney Bernardo1,2,
  • Luiz Augusto Sousa de Oliveira1,2,
  • Grazielle Silva Paz1 and
  • Janaina Fernandes1,* 
 Author information 

Abstract

The advent of nanoparticle technology has transformed oncology therapeutics through its capacity for accurate drug delivery and regulated pharmaceutical release, boosting treatment effectiveness while minimizing adverse reactions. Various nanostructures, including polymeric carriers, liposomal formulations, and metal-based nanoparticles, can be engineered with tumor-specific targeting molecules to facilitate cellular uptake in malignant cells. Despite these advancements, issues such as production scalability, potential chronic toxicity, and regulatory approval processes still need to be addressed. Viral nanoparticles and virus-like particles (VLPs) represent innovative tools in nanotechnology and biomedicine, offering exceptional potential for targeted therapies, immune modulation, and diagnostic applications. Their natural biocompatibility, precise structural organization, and capacity for surface modification make them highly suitable for developing strategies to treat malignant tumors. Alongside VLP development, other approaches have also been investigated, such as magnetic hyperthermia, where magnetic nanoparticles are used to generate localized heat under an external magnetic field, selectively destroying cancer cells while sparing healthy tissue. This paper presents a brief review of nanocarriers in drug delivery systems and discusses the integration of nanoparticles, viral nanoparticles, and VLPs. Additionally, we explore the challenges and propose cutting-edge solutions, offering a forward-looking perspective on how the combination of these advanced technologies could transform oncology.

Keywords

Drug-delivery, Virus-like particles, Viral nanoparticles, Magnetic hyperthermia, Malignant tumor, Merged strategies

Introduction

Nanotechnology applies engineering principles such as electronics and materials, as well as physical science, to fabricating materials at the molecular level, which can be devices, systems, or supramolecular structures with dimensions ranging from 0.1 to 100 nm. Nanotechnology has brought significant advances in medical applications, gene therapy, drug delivery, imaging, and techniques for new approaches to drug therapies.1,2 In terms of drug delivery, several nano-delivery systems for small molecules, proteins, and DNA have been developed. Physicochemical properties of nanostructures allow them to cross cellular and tissue barriers, making them promising materials for biomedical applications.3 In the field of cancer research, the problems associated with therapeutic agents involve bioavailability, biodistribution, degradation, elimination, and elimination of the biological activity of nanoparticles and their structures.4

In addition, classical and target-directed drugs can kill both healthy and malignant cells, leading to strong side effects.5 Drug delivery mediated by nanoparticles represents an improvement over conventional methods, enhancing the selective delivery to the target cancer cells, which has already been achieved for several chemotherapeutic drugs.6 There are still challenges related to nanodelivery of antitumor drugs concerning instability in the biological tract and loss of drugs due to rapid degradation. The tumor-targeting efficiency of nanoparticles is related to several physicochemical, biochemical, and biological features, and the interaction with the cellular surface of the target cells is one of those.

The cellular uptake of nanoparticles is divided into endocytosis-mediated internalization and direct intracellular entry. The first strategy involves endocytosis mediated by clathrin, caveolin, phagocytosis, and macropinocytosis,7 while the direct intracellular entry involves lipid fusion and translocation.8 In addition, artificial strategies such as electroporation and microinjection achieve limited use due to induced deformation of the membrane structure and its destruction.9 Receptor-mediated endocytosis involves the interaction between surface receptor proteins of the cell with target molecules present on the nanoparticle surface. This interaction leads to the activation of the endocytic pathway, with the nucleation of cytosolic proteins within the nanoparticle entry site, generating a coated pit, followed by an invagination process that will create an intracellular vesicle carrying the nanoparticles (Fig. 1). There are five types of receptor-mediated endocytosis: clathrin-dependent endocytosis, clathrin-independent endocytosis (endophilin-mediated/dynamin-dependent and glycosylphosphatidylinositol-anchored protein-enriched endocytic compartment (GEEC), macropinocytosis, phagocytosis, and caveolin-dependent endocytosis.7 Clathrin is a protein involved in the formation of a polyhedral lattice in the cell membrane, which forms a coated cavity as the membrane invaginates to create a vesicle during specific endocytic pathways.10 In clathrin-dependent endocytosis, the nanoparticles bind to cell membrane receptors, e.g., epidermal growth factor receptors. After the formation of the clathrin-coated cavity consisting of receptor-bound nanoparticles within the cavity and the invagination of the cell membrane, the cell membrane’s breakdown forms an intracellular vesicle able to entrap nanoparticles of ∼100 nm in size.11 The endocytosed nanoparticles are then extracted from the vesicles, where they can reach their target.

Nanoparticles endocytosis – In receptor-mediated endocytosis NPs ligands are recognized by cell receptors activating the endocytic pathway, what occurs for clathrin, caveolin, fast endophilin mediated endocytosis, CLIC/GEEC and phagocytosis, while macropinocytosis is not initiated by the binding of specific cargo molecules to receptors on the cell surface.
Fig. 1  Nanoparticles endocytosis – In receptor-mediated endocytosis NPs ligands are recognized by cell receptors activating the endocytic pathway, what occurs for clathrin, caveolin, fast endophilin mediated endocytosis, CLIC/GEEC and phagocytosis, while macropinocytosis is not initiated by the binding of specific cargo molecules to receptors on the cell surface.

Instead, it involves the non-selective uptake of extracellular fluid and its contents through the formation of large membrane structures and subsequent engulfment. After internalization the NPścargo can be released through endosomal or lysosomal escape. CLIC, clathrin-independent carriers; GEEC, glycosylphosphatidylinositol-anchored protein-enriched endocytic compartment; NPs, nanoparticles.

In caveolin-mediated endocytosis, this protein that generates the coated vesicles also follows receptor-mediated binding of nanoparticles, generating flask-shaped membrane invaginations known as caveolae.12 Both clathrin- and caveolin-mediated endocytosis need to constrict and cut the plasma membrane to generate the intracellular vesicles. Dynamin, a GTPase, is thought to perform this role by forming spiral polymers. Even though there are several works showing that this is true for clathrin,13 serious doubts have been raised regarding dynamin’s role in caveolin-mediated endocytosis.14

Clathrin- and caveolin-independent endocytosis (Fig. 1) utilizes lipid rafts, structures composed of cholesterol and sphingolipids found in the cell membrane, which are capable of being endocytosed.15 Endophilin-mediated endocytosis is initiated by cell surface ligand-receptor interaction and is modulated by endophilin A2 recruitment and actin polymerization. The cutting of the membrane neck to generate the vesicles is also dependent on dynamin.11 GEEC endocytosis, on the other hand, is clathrin- and dynamin-independent, utilizing extracellular galectin proteins, glycoproteins, and glycolipids for vesicle formation and loading. GEEC endocytic vesicles are capable of transporting cargo up to 100 nm in size, whereas vesicles formed during the endophilin-mediated/dynamin-dependent endocytosis pathway transport cargo approximately 60–80 nm in size.11

Phagocytosis of nanoparticles can be instigated through interaction with receptors of phagocytic cells such as polymorphonuclear neutrophils, monocytes, and macrophages, but also by non-professional phagocytes, such as Fc receptors and complement receptors.16,17 Phagocytosis can be utilized for the cellular uptake of larger cargoes, typically exceeding 200 nm, and can therefore facilitate the uptake of larger nanoparticles.11 Components of the immune system, such as immunoglobulins and complement proteins, are responsible for identifying and removing nanoparticles by phagocytes through opsonization and adsorption onto the nanoparticle surface. When nanoparticles are phagocytosed, they are contained in phagosome vesicles that, in turn, fuse with a lysosome, thus forming the phagolysosome. On the other hand, macropinocytosis is not initiated by the binding of specific cargo molecules to receptors on the cell surface. Instead, it involves the non-selective uptake of extracellular fluid and its contents through the formation of large membrane structures mediated by actin filaments and subsequent engulfment of the molecules interacting with the cell membrane.18

Once nanoparticles are endocytosed, the endocytic vesicle becomes the early endosomal compartment that matures into a late endosome that fuses with the lysosome. Due to the enzymatic nature of the lysosomal content, this may lead to degradation of the nanoparticle cargo, reducing therapeutic efficiency. To reduce this effect and ensure the delivery of active therapeutic cargo, a common strategy is an event called lysosomal escape, which uses the proton sponge effect, where nanoparticles induce an influx of ions into the lysosome, leading to swelling and rupture.19 Other strategies include pH-sensitive linkers that degrade under the acidic conditions of the lysosome, favoring the release of therapeutics into the cytoplasm; and the addition of peptides that promote fusion with the lysosomal membrane and subsequent release of the drug.20 On the other hand, if the cargo is released from the endosome into the cytoplasm (endosomal escape), the release of the cargo from nanoparticles in the cytosol results in intact therapeutic release, maintaining its bioactivity.21

In addition to the engineered synthetic nanoparticles that possess their own advantages and drawbacks (Fig. 2), this technology also takes advantage of nanoparticles of natural origin, such as viral nanoparticles (VNPs). As with synthetic nanoparticles, VNPs are also subject to endocytic pathway entry. In particular, virus-like particles (VLPs) (a subgroup of VNPs) have become a versatile platform that can be explored to solve a variety of problems that arise with the use of synthetic nanocarriers, including biocompatibility and sharper targeting of tumor cells. This class of nanoparticles also has its own issues regarding drug delivery, encapsulation, scalability, stability, and immune response. Studies on the application of both synthetic and natural nanoparticles to treat tumors where conventional and targeted therapy have failed have been increasing at an exponential rate. This review addresses the combination of several nanocarriers with different therapeutic strategies to improve the anticancer response of patients, discusses the main drawbacks, and the innovative solutions to achieve maximum efficiency.

Types of nanoparticles (NPs) for use in drug delivery, as well as its advantages and disadvantages.
Fig. 2  Types of nanoparticles (NPs) for use in drug delivery, as well as its advantages and disadvantages.

Cancer

Cancer is a major public health problem worldwide and is one of the most prevalent malignant diseases that cause morbidity and mortality.22 Traditional methods of cancer treatment include surgical resection, chemotherapy, and radiotherapy. Immunotherapy and photothermal therapy have also emerged recently.23,24 Chemotherapeutic drugs have several types of mechanisms, including affecting the chemical structure of DNA, inhibiting nucleic acid synthesis, acting on nucleic acid transcription and DNA replication, and interfering with mitotic tubulin synthesis.25 However, these drugs do not distinguish between healthy and malignant cells, leading to side effects during treatment. In addition, conventional drugs for this type of treatment have several problems, such as drug solubility in aqueous media, biodistribution, short half-life, and the resistance of these drugs over time.26

Several types of nanoparticles, including liposomes, polymeric nanoparticles (PNPs), solid lipid nanoparticles (SLNs), dendrimers, silica-based materials, and magnetic nanoparticles, constitute an improvement in drug delivery to cancer cells, increasing treatment efficacy and minimizing side effects.27 Thus, the binding of these drugs to nanoparticles currently presents great advantages, such as targeting the drug to the proper tumor cell (active targeting). Active targeting utilizes molecular recognition: ligands, such as aptamers or monoclonal antibodies, are conjugated to nanocarriers, enabling them to specifically bind to receptors that are overexpressed on the surface of cancer cells. This effect enables the administration of the medication to be more precise and reduces unwanted side effects. On the other hand, passive targeting leverages the enhanced permeability and retention effect,28 a consequence of the fact that tumor vasculature tends to be leaky and lacks adequate lymphatic drainage, allowing nanoparticles, such as liposomes or polymeric micelles, to accumulate more easily in tumor tissues than in healthy tissues. We can combine both strategies, with passive targeting ensuring that nanocarriers reach the general tumor area, while active targeting enhances targeting down to the cellular level.28 We have several examples, such as in the case of achieving selective drug accumulation in tumor tissues through antibody-mediated binding, like monoclonal antibodies (trastuzumab) that can guide nanoparticles to human epidermal growth factor receptor-2 (HER2)-positive breast cancer cells,29 or even the evolution of cancer-targeting therapies (the development of Accurins), which are programmable nanoparticles containing a therapeutic load, grafted with a ligand targeting the extracellular domain of prostate-specific membrane antigen, designed to target tumors at three levels: tissue, cellular, and molecular.30 There are approximately 14 drugs with nanoparticles approved by the U.S. Food and Drug Administration for clinical use, the majority of which are in the liposome matrix. These nanoparticles are more efficient vehicles for transport and targeted delivery in a hydrophobic manner without provoking an immune response.31

Anticancer drugs are generally lipophilic and behave well within the trapped vesicles, being protected from external reactions.32 Nanoparticles associated with anticancer drugs act on angiogenesis mechanisms, uncontrolled cell proliferation, and increased tumor mass,33 affecting only tumor tissues and reducing multidrug resistance.34 The issue of endocytosis of these tumor cells by these drugs through nanoparticles is an important factor because it reduces the dispersion of the drug outside the cancer cell and can increase the therapeutic potential of the drug.35 In addition to drugs, nanoparticles can be loaded with peptides, proteins, nucleic acids, and antibodies, improving their pharmacokinetics.36 There are several challenges for nanoparticles in cancer therapy; among them are the proper delivery and accumulation at the target site and decreased accumulation at off-target sites, problems addressed by those who study the improvement of drug delivery systems.

Nanocarriers in drug-delivery systems for cancer treatment

In the development of nanocarriers (biodegradable or non-biodegradable), the drugs are enveloped, adsorbed, or encapsulated in the nanoparticle matrix and can be prepared in several ways with distinct drug-release properties and characteristics (Fig. 2).

Liposomes

These nanocarriers were the first to be tested as drug carriers. They are spherical vesicles composed of phospholipids and steroids. They may have a bilayer or even surfactants formed when some lipids are dispersed in an aqueous medium and can be prepared by sonication.37 Liposomes increase the solubility of drugs and improve their pharmacokinetic properties, such as the therapeutic index of chemotherapeutic agents, rapid metabolism, and reduction of harmful side effects.38 The drug is incorporated into the liposome via an encapsulation process, and its release depends on several factors, such as the composition of the liposome, pH, and osmotic gradient.38 The interactions between liposomes and cells are mediated by adsorption, fusion, and endocytosis. Several liposomal formulations are available for anticancer drugs such as cisplatin,38 as well as for other conditions, which use liposomal formulations with neurotransmitters,39 antibiotics,40 anti-inflammatories,41 and antirheumatics.42

Nanoparticles based on solid lipids

SLNs, nanostructured lipid carriers, and lipid drug conjugates (LDCs) are carrier systems based on a solid lipid matrix.43 They have been used in dermal,44 parenteral,45 ocular,46 pulmonary,47 and rectal administration.48 SLNs are particles made of solid lipids, e.g., highly purified triglycerides, complex mixtures of glycerides, or waxes stabilized by various surfactants.38,49 They are characterized by good physical stability, drug-degradation protection, and controlled drug release. However, they have a low drug-loading capacity due to low drug solubility in the lipid. Nanostructured lipid carriers and LDCs are modifications of lipid-based nanoparticles that were developed to overcome the limitations of conventional SLNs, such as low drug-loading capacity, and LDCs were developed for lipophilic drug delivery.43 SLN has been extensively studied for application in the treatment of triple-negative breast cancer using docetaxel.50,51

Polymeric nanoparticles

PNPs are structures with a diameter ranging from 10 to 100 nm. They are obtained from various synthetic polymers, such as polyacrylamide and polyacrylate,52,53 or even from natural polymers, for example, albumin,54 DNA,55 chitosan,55 and gelatin.56 PNPs can be classified as biodegradable—poly(L-lactide),57 and polyglycolide,58 and non-biodegradable, such as polyurethane.59 PNPs have a structure coated with non-ionic surfactants to minimize immunological interactions, as well as molecular interactions between surface chemical groups such as van der Waals forces and hydrogen bonds.60 Drugs can be immobilized on the surface of PNPs or encapsulated and released into the target tissue by diffusion or desorption.60–62 PNPs have been evaluated to improve delivery to treat resistant tumors.63,64

Dendrimer nanocarriers

They were discovered in 1978 and are macromolecular compounds with a series of branches around an internal core.65 These systems are interesting for drug delivery because of their nanometric size range, ease of preparation and functionalization, and ability to display multiple copies of surface groups for biological reorganization processes.55,66 In the structure of the dendrimer, in contrast to the linear polymer, the following elements can be distinguished: a core, dendrons, and surface active groups. The core is a single atom or molecule (only if it has at least two identical functional groups) to which the dendrons are attached. The dendrons (dendrimer arms) are monomer molecules attached to the core that form layers in successive generations. The biocompatibility and physicochemical properties of dendrimers are determined by the presence of surface functional groups.67 Due to their globular shape and the presence of internal cavities, they have interesting properties, such as encapsulating drugs inside macromolecules or attaching them to surface groups.68 The encapsulation of drugs inside macromolecules is used when they are toxic, unstable, or poorly soluble. On the surface of dendrimers, the amount of drugs can be controlled through the number of covalent bonds.58 The surface of dendrimers provides a surface for the binding of specific ligands, which may include folic acid,69 antibodies,70 peptides,71 selective adenosine A3 receptors,72 antimicrobial agents of silver salt complexes,73 and poly(ethylene glycol).74 Among the anticancer drugs conjugated in dendrimers are doxorubicin, camptothecin, cisplatin, and paclitaxel.75

Silica materials

Silica materials are used in drug delivery as xerogels through the sol-gel method.76 This manufacturing process involves temperature, agitation, the proportion of reagents, and drying of this material during the reaction,77 allowing the formation of an amorphous and porous material with high surface area and biocompatibility that can carry drugs such as doxorubicin and cisplatin.78,79

Carbon nanomaterials

Carbon nanocarriers used in drug delivery systems are differentiated into nanotubes and nanohorns.3,80,81 Carbon nanotubes are formed by rolling single (single-walled carbon nanotubes) or multi (multi-walled carbon nanotubes) layers of graphite with an enormous surface area and excellent electronic and thermal conductivity,81 and their biocompatibility can be implemented with dendrimer anchoring.81,82 Single-walled carbon nanotubes have been used to improve the properties of other carriers, such as polymeric or non-polymeric composites.81–83 There are three ways of immobilizing drugs in carbon nanocarriers: encapsulation of a drug in a carbon nanotube,84,85 chemical adsorption on the surface or in the spaces between the nanotubes (by electrostatic, hydrophobic interactions, and hydrogen bonds),86,87 and fixation of active agents in functionalized carbon nanotubes. The application of carbon nanomaterials in cancer treatment has expanded to a variety of architectures combined with photodynamic therapy,88 gene therapy, immunotherapy, and chemotherapy, and has been demonstrated in gastric cancer,89 liver cancer,90 pancreatic cancer,91 ovarian cancer,92 among others.93

Magnetic nanoparticles

Magnetic nanoparticles are highly promising materials that can be used in several areas of nanotechnology. They are relatively easy to manufacture, low-cost, biocompatible, and can be directed by an external magnetic field. They can be visualized by magnetic resonance imaging (MRI).3,94 Magnetic nanoparticles can be manufactured using family 1B metals with empty orbitals (3d), such as iron, manganese, zinc, cobalt, and nickel. Iron magnetic nanoparticles have been approved for clinical use by the U.S. Food and Drug Administration as an MRI agent and to treat iron deficiency.95 In the synthesis of magnetic nanoparticles using iron, cobalt, nickel, and zinc, a core can be formed, and various types of coatings can be applied, such as polymers,96 dendrimers,97 or silanes.98

Nanocarriers and their challenges

In the current pharmaceutical industry, one issue concerns drug delivery systems and the pharmaceutical technologies used, which are less developed than the production of drugs, whether they are of natural or synthetic origin. The industry is seeking new delivery systems, which has shifted the pharmaceutical market in this direction. The therapeutic arsenal of drugs is extensive, and often the application of these delivery systems is minimal, requiring investment and a change in perspective on drugs that are usually already established but whose effectiveness in some diseases is entirely compromised. From a technological point of view, nanocarriers have interesting characteristics, including their surface-to-mass ratio—much higher than that of other particles, their quantum properties, and their ability to absorb and transport drugs to different biological systems. Although nanoparticles are defined as 1–100 nm in size, this size limits the adsorption of drugs, and often, in a disease, a larger quantity of the transported drug is needed. Consequently, the size range (1–100 nm) is, in this case, relative.

Furthermore, differences in size can influence distribution and bioavailability.99–103 In liposomes with sizes above 100 nm, the clearance rate by the phagocytic system increases with liposome size,104 whereas liposomes smaller than 100 nm show a more effective charge.105 However, this varies with nanoparticle composition. The size, composition, surface charge, and shape of the nanoparticles are equally crucial for their performance.106 Factors such as heating and light can cause or even increase their therapeutic effects, such as cell death or drug release at the drug site. Doxorubicin is an example, as it exhibits increased cytotoxicity when the in vitro temperature is 42°C.107

The composition of these nanocarriers can be of natural or synthetic origin; in this case, it is essential to consider their biocompatibility. It is important not only to transport nanoparticles to the tissue or organ site and release the drug from the nanoparticle but also to ensure biocompatibility with the biological system and reduce toxicity.3,108–110

Formulation challenges

The ideal condition in drug administration is to deliver the drug to the tumor site in the body while minimizing its harmful effects on unaffected tissues, which is a challenge considering the development of metastasis, a feature of resistant tumors. Nanoparticles improve local action related to the affected organ or tissue, increasing the amount of drug at the site and allowing penetration into cell membranes and lysosomal escape after endocytosis.111 There are several reviews on nanometric formulations of chemotherapeutics.112–114 In addition to size, the chemistry of the particle surface is crucial for the absorption, distribution, stability, and effects of the particles. However, one problem is that nanoparticles can become trapped in the liver and spleen,115,116 which can be positive for the treatment of liver tumors. On the other hand, it reduces the circulation of these nanoparticles to target other organs. In the case of nanoparticles for oral administration, this can be critical, as some nanoparticles will be metabolized in the liver (first-pass effect), and much of the carrier and drug will be lost. The ideal use would perhaps be local or intravenous administration to minimize drug loss along with the carrier.117 At present, several liposomes and PNPs are approved for use in anticancer treatment (Table 1).118–139

Table 1

Approved cancer drugs using nanotechnology

Nanocarrier typeDrugsDrug product nameAgency and year of approvalTreatment indicationReference
LiposomesMitoxantroneNovantroneEMA (2016)Lymphoma and Breast cancer118
LiposomesIrinotecan liposome injectionOnivydeFDA (2015)Metastatic pancreatic cancer119,120
LiposomesEribulin mesylateHalavenFDA (2012), EMA (2011)Liposarcoma and Breast neoplasms121
LiposomesLiposome vincristine sulfateMarqiboFDA (2012)Anticancer alkaloid that binds to tubulin and interferes with cell division122
LiposomesMifamurtideMepactEMA (2009)Osteosarcoma123
LiposomesPaclitaxel Genexal/CynviloqPaclitaxel Genexal/CynviloqKorea (2007)Breast cancer and Non-small cell lung cancer124
LiposomesDoxorubicin nonpegylated liposomalMyocetFDA (2000)Breast cancer125,126
LiposomesLiposomal cytarabineDepocytFDA (2007)Lymphomatous meningitis/ antineoplasic agent/ inhibit DNA polymerase127,128
LiposomesDiphtheria toxinOntakFDA (1999)T-cell lymphoma129,130
LiposomesLiposomal daunorubicinDaunoXomeFDA (1996)HIV-associated Kaposi’s Sarcoma (KS) as a chemotherapy drug131
LiposomesLiposomal DoxorubicinDoxil/Caelyx/LipidoxFDA (1995)Treatment of diferent cancers ranging from metastatic ovarian cancer132,133
LiposomesDaunorubicin and cytarabine encapsulated in liposomesVyxeosFDA (2017)Acute myeloid leukemia134,135
Polymeric NPsPEGylated-L asparaginaseOncasparFDA (1994)Acute lymphoblastic leukemia and chronic myelogenous leukemia136,137
Polymeric NPsAlbumin-NPs bound to paclitaxelAbraxaneFDA (2005/2012/2013)Metastatic breast cancer, lung cancer and metastatic pancreatic adenocarcinoma138
Polymeric NPsLeuprolide acetateEligardFDA (2002)Prostate cancer139

Challenge of the brain and its blood-brain barrier (BBB)

The brain poses specific challenges regarding drug delivery. The BBB is well known as the body’s best shield against exogenous substances.140 Pharmaceuticals, including most small molecules, generally do not cross the BBB. The endothelial barrier is particularly narrow at the interface with brain astrocytes and, under normal conditions, can be crossed only using endogenous BBB transporters, resulting in transporter-mediated active efflux transport and/or receptor-mediated transport. However, the barrier properties can be compromised, either intentionally or unintentionally, by drug treatment, allowing the passage of nanoparticles.141–144 Several studies analyze the toxic effects of nanoparticles on brain endothelial cells (nanoparticles > 200 nm),145 but contradictory results have been reported in other studies with similar nanoparticles.142 When nanoparticles with different surface characteristics were evaluated, it was found that neutral nanoparticles and low concentrations of anionic nanoparticles had no effect on BBB integrity, whereas high concentrations of anionic and cationic nanoparticles were toxic to the BBB. The extent of brain uptake by anionic nanoparticles at lower concentrations was higher than that by neutral or cationic formulations at the same concentration. Therefore, the surface charges of nanoparticles should be considered when determining their toxicity and brain distribution profiles.143 In particular, coating nanoparticles with surfactants (e.g., Tween) has resulted in drug transport across the BBB. The transport mechanism is suggested to be endocytosis through the low-density lipoprotein receptor of endothelial cells after the adsorption of lipoproteins from blood plasma onto the nanoparticles.142 The role of apolipoprotein E has been observed in drug transport across the BBB, whereas apolipoprotein E variants that do not recognize lipoprotein receptors fail to transport the drug across the BBB.146

It has been suggested that recognition and interaction with lipoprotein receptors on brain capillary endothelial cells are responsible for drug uptake by the brain. BBB passage can also be achieved by masking certain drug characteristics, thereby preventing or limiting binding to cellular efflux systems such as P-glycoprotein, a cellular transporter associated with drug removal from cells. P-glycoprotein is an adenosine triphosphate (ATP)-dependent efflux transporter that plays an important physiological role in limiting drug entry into the brain.147–149 Other routes to reach the brain, bypassing the BBB, include migration along the olfactory or trigeminal nerve endings after deposition in the olfactory mucosa in the nasal region.150 Solid nanoparticles, such as manganese oxide, have also been shown to be translocated to the brain via the olfactory pathway,151 based on measurements of manganese in different parts of the brain. Due to the BBB, a number of brain treatments for cancer with nanoparticles involve intracranial injections, but the search for a non-invasive route to the brain has become a priority among investigators.152

Hyperthermia-based anti-cancer treatment

Hyperthermia-based treatment dates back to ancient civilizations such as the Egyptians, Greeks, and Romans, who recognized the healing properties of heat, using thermal baths and saunas not only for relaxation but also to alleviate pain and treat various ailments.

In the late 19th century, medical practitioners began to observe that cancer patients who experienced high fevers occasionally saw their tumors regress. This observation led to the hypothesis that heat could have a direct therapeutic effect on diseases. This marked one of the earliest scientific acknowledgments of the potential of heat in treating cancer.153

By the 1970s, technological advancements allowed for the development of precise equipment, such as microwave and ultrasound devices, enabling controlled heat application. Consequently, in 1975, during the first International Hyperthermia Congress held in Washington, scientists unveiled groundbreaking findings highlighting the effectiveness of combining heat therapy with conventional treatment methods for combating tumors. This announcement marked a pivotal moment in cancer research, establishing hyperthermia as a promising complementary approach to traditional therapies.154

In the 21st century, hyperthermia gained recognition as an adjuvant therapy in cancer treatment, particularly when combined with radiotherapy, chemotherapy, and immunotherapy. Ongoing clinical studies and research continue to explore its potential in treating various types of tumors and other diseases. Heat causes direct damage to cancer cells by denaturing proteins and damaging cell membranes, leading to cell death. Cancer cells are particularly vulnerable to this damage due to their less efficient repair systems. Additionally, hyperthermia enhances the effectiveness of other treatments by increasing blood flow to tumors, thereby improving the delivery of chemotherapy drugs and sensitizing cancer cells to radiation. It also stimulates the immune system, activating cells such as lymphocytes and macrophages,155,156 which promote a more robust antitumor response (Fig. 3).

Overview of viral nanoparticle-based drug delivery and magnetic hyperthermia.
Fig. 3  Overview of viral nanoparticle-based drug delivery and magnetic hyperthermia.

This figure shows the various mechanisms of action of VNPs and how they can be combined with magnetic nanoparticles for tumor targeting and drug release. The figure also highlights the role of magnetic hyperthermia in enhancing therapeutic efficacy by localizing heat to the tumor site. HT, hyperthermia; NPs, nanoparticles; VNPs, viral nanoparticles.

Modern hyperthermia techniques vary depending on the application. Local hyperthermia targets superficial or accessible tumors using radio waves, microwaves, or ultrasound. Regional hyperthermia is used for larger areas, such as organs or body cavities, often involving the perfusion of heated fluids or radiofrequency devices. Whole-body hyperthermia, which consists of heating the entire body, is typically reserved for cases of metastatic cancer and can be induced using thermal blankets, hot water baths, or specialized devices.157 Despite its promising potential, hyperthermia is not without challenges and risks. Precise temperature control is crucial, as excessively high temperatures can damage healthy tissues and cause burns, swelling, or pain in the treated area. In cases of whole-body and regional hyperthermia, for example, there is a risk of overheating and systemic complications, leading to severe side effects like gastrointestinal symptoms and cardiac complications.157,158

As an advanced evolution of the technique, magnetic hyperthermia involves injecting magnetic nanoparticles into tumors, which generate localized heat when exposed to an alternating magnetic field (AMF). This approach is highly efficient, noninvasive, and minimizes damage to normal tissues. It is also cost-effective, offers excellent tissue penetration, and effectively destroys cancer cells.

Magnetic hyperthermia

Magnetic hyperthermia has emerged as a revolutionary approach in cancer treatment, offering a unique blend of precision, efficiency, and minimal invasiveness. When exposed to an AMF, this innovative technique leverages magnetic nanoparticles to generate localized heat within tumors. By delivering these nanoparticles directly to the tumor site—either through passive targeting, which exploits the leaky vasculature of tumors through what is known as the enhanced permeability and retention effect,159 or active targeting using tumor-specific ligands—magnetic hyperthermia ensures that heat is concentrated precisely where needed. When the AMF is applied, two phenomena emerge: Néel relaxation, due to the internal reorientation of the magnetic moments within the magnetic nanoparticle, and Brownian relaxation, due to the rotation of the entire nanoparticle within a fluid caused by Brownian motion. These processes produce heat that raises the local temperature to therapeutic levels, typically between 42–46°C. This localized heating disrupts cancer cell membranes, denatures proteins, and impairs mitochondrial function, leading to cell death or halted proliferation while sparing surrounding healthy tissues.160

The development of advanced magnetic nanoparticles has been a driving force behind the success of magnetic hyperthermia. Traditional iron oxide nanoparticles (Fe3O4 and/or γ-Fe2O3) have been widely used due to their biocompatibility and magnetic properties.161 These nanoparticles can be tailored to optimize their performance, ensuring they accumulate effectively in tumors and generate sufficient heat under the influence of the AMF.

One of the most compelling aspects of magnetic hyperthermia is its ability to combine this thermal therapy with other treatment modalities.162 Magnetic nanoparticles can be functionalized to carry chemotherapy drugs or be encapsulated within thermo-responsive materials that release therapeutic agents when heated. This dual approach enhances the direct cytotoxic effects of heat and allows for targeted drug delivery, reducing systemic side effects. Additionally, it can activate thermo-responsive promoters to trigger the expression of therapeutic genes, further amplifying its anticancer effects. The versatility of magnetic hyperthermia extends to its compatibility with existing treatments like chemotherapy and radiotherapy, as the heat generated can sensitize tumor cells, making them more susceptible to these therapies.

Established therapies combined with magnetic hyperthermia

Magnetic hyperthermia exerts its antitumor effects through various interconnected mechanisms. These mechanisms include direct cell damage, structural changes in cells, DNA damage, repair inhibition, apoptosis induction, and modulation of heat shock proteins (HSPs). Together, these processes halt tumor growth and enhance the effectiveness of other cancer therapies.163,164

Direct cell damage is the primary mechanism of hyperthermia and depends on the temperature and duration of heat exposure. At moderate temperatures (40–45°C), prolonged exposure of 30–60 min is required to cause irreversible cell damage.165 At higher temperatures (above 60°C), proteins rapidly denature, leading to immediate cell death through coagulative necrosis. This destabilizes the cytoskeleton and cell membrane structure, impairing motility and intracellular signaling. Mitochondria are particularly sensitive to heat, with high temperatures causing proton leakage, swelling, and structural changes that contribute to cell death. Additionally, heat inhibits DNA replication by denaturing essential enzymes, such as DNA polymerase alpha, further compromising cell survival.166–170 Hyperthermia also induces significant changes in cellular structure, particularly in the cytoskeleton, which is essential for maintaining cell shape and function.

Another important mechanism is DNA damage and repair inhibition. Hyperthermia directly causes DNA breaks and the formation of apurinic/apyrimidinic sites while promoting the generation of reactive oxygen species that further damage DNA.171 Heat limits the activity of DNA polymerases, impairing replication and increasing DNA breaks. It also promotes the formation of γH2AX foci, markers of double-strand breaks, and modulates the activity of proteins such as ATM and ATR, which are involved in the DNA damage response. Additionally, hyperthermia disrupts multiple DNA repair mechanisms, including base excision repair, nucleotide excision repair, mismatch repair, and homologous recombination. By inhibiting these repair mechanisms, hyperthermia increases the susceptibility of cancer cells to other treatments, such as chemotherapy and radiotherapy.172–176

Finally, hyperthermia also triggers apoptosis, or programmed cell death, and modulates the expression and function of HSPs, which play a dual role in the tumor response. On the one hand, HSPs are overexpressed in response to heat stress, helping cells cope with protein denaturation and preventing irreversible damage. On the other hand, extracellular HSPs released by necrotic tumor cells act as signaling molecules, activating the immune system. Proteins such as HSP70 and HMGB1 enhance antigen presentation and dendritic cell activation, promoting an immune response against the tumor. HSP70, in particular, forms complexes with tumor antigens recognized by immune cells, further amplifying antitumor immunity.

Hyperthermia in clinical applications

The clinical applications of hyperthermia are extensive. It is widely studied in cancer treatment, particularly in combination with radiotherapy, chemotherapy, and immunotherapy (Fig. 3). It has shown promise in treating breast, prostate, head and neck, and melanoma cancers, among others. Clinical studies have demonstrated that hyperthermia can increase tumor response rates and improve patient survival. A summary of clinical studies evaluating the efficacy and outcomes of hyperthermia combined with radiotherapy, chemoradiotherapy, and immunology across various cancer types is presented in Table 2.177–190

Table 2

Summary of clinical studies evaluating the efficacy and outcomes of hyperthermia combined with radiotherapy, chemoradiotherapy and immunology across various cancer types. Missing values indicate unreported or inapplicable data

Cancer typeTreatment protocolSample sizeKey outcomespCR/CR rate3–5 years OS (%)Main conclusionStudy (Ref.)
Rectal cancerRT + CT + Magnetic HT7814% pCR; 50% Dworak 3–4; 3-yr OS: 94%, DFS: 81%1494Magnetic HT is feasible, enhances regression, and maintains QoL177
LACC (IB2–IVA)RT (46–50.4 Gy) + BT + Weekly RHT378CR: 77%; 5-yr LC: 53%, DSS: 47%, Late Toxicity: 12%77-RHT improves outcomes; supports use in LACC as alternative to chemoradiation178
Locally advanced prostate cancerEBRT (70 Gy) + Regional/Interstitial HT263-yr bNED: 70% overall; 79% (regional), 57% (interstitial)-100Combined therapy favorable; minimal toxicity179
Bladder, cervical, rectal cancerRT ± HT (median dose: 65 Gy)358CR: 55% (RT+HT) vs. 39% (RT); 3-yr OS: 51% vs. 27%5551HT improves CR and LC; especially effective in cervical cancer180
Advanced prostate cancer (C2–D1)EBRT (median 68 Gy) + 1–2 HT sessions265-yr OS: 73%; bNED: 35%; PSA nadir significant-73HT feasible, but limited outcome improvement181
HNSCC (recurrent)Re-irradiation + Weekly FRWBH10No excess toxicity; partial protocol adherence-86FRWBH potentially beneficial, but challenging to implement182
Locally advanced rectal cancerPreop RCT (RT + 5-FU/Leucovorin) + Weekly HT37pCR: 14%; R0 resection: 86%; 38-mo OS: 86%14-HRCT is effective, safe, and improves locoregional control183
Prostate cancer (T2b–T3b)EBRT ± Androgen Therapy + 2 TRUS HT sessions30Rectal toxicity correlated with Tmax; GI toxicity ≤ Grade 2--Rectal wall temperature strongly predicts acute toxicity184
BladderCHT (MMC) vs. BCG (1-year regimen)19024-mo RFS (PP): 81.8% (CHT) vs. 64.8% (BCG); p = 0.02--CHT demonstrated superior RFS in PP analysis; both had <2% progression. Study underpowered but supports CHT as viable alternative.185
BreastLow-dose ICB (nivolumab, ipilimumab), regional HT, systemic fever-range HT (IL-2)Case reportComplete remission of pulmonary metastases100alive at 27 moCombined ICB + HT + IL-2 induced durable remission with minimal toxicity; promising protocol for further TNBC research.186
Breast, ovary, colon, prostateLocoregional & whole-body HT, IL-2, low-dose ipilimumab + nivolumab131ORR: 31.3%, median PFS: 10 mo, OS: 36.6% at 24 mo; Grade 3–4 irAEs: 8.4%31.336.6Novel ICI-HT combination therapy showed favorable safety and efficacy; further optimization with viral/bacterial agents suggested.187
High-risk Soft Tissue SarcomaNeoadjuvant CT + Regional HT341HR for local PFS = 0.65 (p=0.002); HR for OS = 0.73 (p=0.04); 10-yr OS: 52.6% (HT) vs. 42.7% (CT)Not reported62.7% (HT) vs. 51.3% (control)Regional HT significantly improves local PFS and OS in high-risk sarcoma.188
Colorectal Peritoneal MetastasesCRS ± Oxaliplatin-based HIPEC265No OS benefit (HR=1.00, p=0.99); Grade ≥3 AEs higher at 60 days in HIPEC group (26% vs. 15%)Not reportedno improvementHIPEC did not improve OS; CRS alone should remain standard for peritoneal metastases.189
Stage III Epithelial Ovarian CancerCRS ± Cisplatin-based HIPEC245RFS: 14.2 mo (HIPEC) vs. 10.7 mo (CRS); HR for death = 0.67 (p=0.02); No significant increase in Grade 3–4 AEsCR not specified, but improved RFS45.7 mo (HIPEC) vs. 33.9 moHIPEC added to interval CRS significantly improves RFS and OS without increasing toxicity.190

Magnetic hyperthermia in clinical applications

In 2003, the first clinical feasibility study on magnetic nanoparticle hyperthermia was conducted with 14 glioblastoma multiforme patients, including two with primary tumors and 12 with recurrences.191 Magnetic fluid was injected into the tumors using neuro-navigation guidance, ensuring a nearly atraumatic procedure with no complications. Slow injection prevented increased intracranial pressure. Patients underwent four to ten thermotherapy sessions (median: 6.5), each lasting 1 h and administered twice weekly, combined with external beam radiation. The median injected fluid volume was 3 mL (range: 1.0–5.5 mL), corresponding to 0.1–0.7 mL per mL of tumor volume. Treatment was well tolerated at magnetic field strengths of 3.8 to 13.5 kA/m (median: 8.5 kA/m), with only minor side effects reported. Intratumoral temperatures reached 42.4–49.5°C, with 90% of tumor volumes achieving 39.3–45.5°C (median: 40.5°C). Approximately 55% of tumor volumes exceeded 42°C, and the median CEM43 T90 was 7.7 min (range: 3.2–502 min). These results demonstrate the feasibility and tolerability of magnetic nanoparticle hyperthermia in treating glioblastoma multiforme.

A prospective feasibility study, launched in February 2004, included 22 patients with recurrent or residual tumors (non-resectable and pre-treated, such as prostate and cervix carcinoma, and soft tissue sarcoma). All patients received additional radiotherapy and/or chemotherapy in conjunction with the experimental treatment. All patients tolerated the nanoparticle instillation well, although pre-irradiated tumor tissue in some cases posed mechanical resistance to injection and fluid diffusion. Median infiltration volumes were 3 mL (range: 1.5–5 mL) for chemotherapy/transrectal ultrasound (CT/TRUS)-guided procedures and 8.5 mL (range: 6–12.5 mL) for TRUS-guided prostate treatments, equating to approximately 0.3–0.4 mL of magnetic fluid per mL of tumor volume. Intraoperative infiltration volumes averaged 7 mL per patient (range: 2.3–10 mL). Magnetic field strengths were limited by patient discomfort (e.g., at skin folds or bone surfaces) to 3–5 kA/m in the pelvic region and up to 8.5 kA/m in the upper thorax, resulting in a median CEM43 T90 of 10.5 min (range: 1–106 min). Treatments were generally well tolerated, with minor to moderate side effects such as sensations of heat, superficial skin burns, increased pulse rate, and elevated blood pressure. Two patients experienced grade 1 to 2 perineal pain lasting up to 4 months. Overall, the study demonstrated the feasibility and tolerability of magnetic nanoparticle hyperthermia in treating recurrent and residual tumors.191

The first clinical use of interstitial hyperthermia with magnetic nanoparticles in human cancer was performed by Johannsen et al.192 A pilot study investigated the potential of magnetic hyperthermia as a minimally invasive treatment for locally recurrent prostate cancer. Treatment planning involved CT imaging to determine the optimal number and placement of magnetic fluid depots in the prostate, ensuring adequate heat delivery while protecting the rectum and urethra. Magnetic nanoparticles were injected transperineally under ultrasound and fluoroscopy guidance, and treatments were administered using a magnetic field applicator operating at 100 kHz with a field strength ranging from 0 to 18 kA/m. Invasive temperature measurements were taken during the first and last of six weekly 60-min magnetic hyperthermia sessions. CT scans were repeated to track nanoparticle distribution and the positioning of temperature probes. The nanoparticles remained in the prostate throughout the six-week treatment period, allowing for non-invasive temperature estimation using AMIRA software, which correlated well with direct invasive measurements. A cooling device was used to ensure patient comfort without the need for anesthesia. In the first patient treated, intra-prostatic temperatures ranged from 40.0°C to 48.5°C at a field strength of 4.0–5.0 kA/m, demonstrating effective and controlled heating. These encouraging findings prompted the initiation of a phase I study to evaluate the feasibility, toxicity, and impact on quality of life in patients with biopsy-confirmed local recurrence following radiotherapy.192

Viruses as nanocarriers

Viruses are infectious agents ranging in size from 17 to 1,500 nanometers.193 Since they are obligatory intracellular parasites incapable of generating energy, they do not feed or grow. Their fundamental structure includes a viral genome, which can be either DNA or RNA, but not both simultaneously, and a protein capsid, which protects this genetic material. In addition to the capsid, some viruses may possess a lipid envelope derived from the host cell membrane. Virus-encoded proteins at the surface of the capsid or envelope are used for the recognition and infection of host cells through several pathways, including the endocytic pathway or direct injection of the genetic material into the host cell.194 The life cycle of a virus comprises several stages: attachment to the cell surface, cell entry, uncoating (release of genetic material into the cell), expression of the early genes, replication of the viral genome, expression of late genes (virus structural proteins), viral assembly (formation of new virion particles), and release from the host cell, leading to or not leading to host cell lysis.194

Compared to cellular organisms, viruses are structurally simple, allowing them to be highly efficient at replicating and generating viral burst values, which refers to the number of viruses released by an infected cell during its life cycle.195 These values can range from 50 to 100 viruses per infected cell in bacteriophages to 50,000 particles per cell in HIV.196–198 Much of this efficiency is related to how viruses assemble their structures and escape from the cell. In terms of morphology, viruses can present icosahedral symmetry or helical symmetry. Non-enveloped icosahedral viruses have higher viral bursts.199 This efficiency is also related to self-assembly, the process that allowed the development of natural nanocarriers of viral origin.

Self-assembly of viral capsids

The viral particles assemble spontaneously from their protein subunits and nucleic acid genomes in a process known as self-assembly, defined as “The autonomous organization of individual components into patterns or structures without human intervention”.200 The viral genome encodes the information that determines their three-dimensional structures in a way that the protein subunits bind to each other. The length of the viral genome, and consequently the number of unique proteins it can encode, is limited by the necessity of being enclosed within its capsid; thus, the capsid must be composed of a large number of identical protein subunits to avoid exhausting the coding capacity of the genome.201 While for some viruses, the capsid grows around the genome, others build an empty capsid, and the genome is later packed using packaging motors dependent on ATP hydrolysis.202

The interactions between neighboring subunits are specific. With all subunits on the capsid surface identical, their interactions will be nearly identical, resulting in a compact, closed, and symmetrical structure.203 Viral capsids are supramolecular structures, whose assembly results from various noncovalent interactions, including van der Waals forces, electrostatic and hydrophobic interactions, hydrogen bonding, among others.204 As a consequence of the self-assembly strategy, non-infective virus particles (lacking viral genome) may be naturally produced, generating empty shells. This phenomenon was observed in 1968 by Bayer and collaborators.205 Since then, it has been observed that these characteristics of viral capsids could make them suitable for the delivery of drugs and other therapeutic alternatives, leading to the development of what is now known as VNPs and VLPs.

VNPs and VLPs

As mentioned in previous sections, synthetic nanocarriers offer several benefits but also some drawbacks, such as toxicity, pharmacokinetics, and lack of tissue specificity. VNPs are defined as nanomaterials derived from plants, bacteriophages, and mammalian viruses. VNPs are the version that contains the viral genome,206 while VLPs constitute a subgroup of VNPs, whose structures are composed of natural viruses lacking their genomes, making them incapable of replication and non-infectious, thus safe for use in vaccines, diagnostics, and research.207 Since the obligatory parasite features of the virus are essential to its propagation strategy, their components present intrinsic biocompatibility and biodegradability, an advantage over synthetic nanocarriers.208 Furthermore, co-evolution with their hosts generates tissue specificity, defining the viral tropism toward tissues and organs.

VNPs are made by expressing viral proteins in host systems like bacteria, yeast, insects, or mammalian cells, where the proteins self-assemble into nanoparticles. These particles are then purified and often engineered for specific tasks, such as displaying targeting ligands, encapsulating drugs, or carrying genetic material.209 In VNPs, the viral genome remains inside the capsid, and the resulting product can be either replication-competent or replication-defective vectors. Examples include adenovirus nanoparticles for gene therapy, where specific regions essential for gene expression of its double-stranded DNA genome are replaced by a therapeutic gene that, when expressed, can lead to defect corrections (gene therapy).210 In adenovirus vaccine applications, the genes inserted are from antigens that stimulate the immune system.211 In cancer gene therapy, genes related to the activation of the cell death pathway are activated to induce tumor cell death.212 Plant virus nanoparticles for cancer therapy have been extensively studied, also due to their safety, since, with or without their genomes, plant viruses do not replicate in mammalian cells.213

While for some viruses the capsid can self-assemble with or without the presence of its genome, other VNPs are derived from viruses that can only assemble in the presence of their genomes. Thus, those with this limitation need to carry their loads exclusively at the surface of the VNP, such as the potato virus X.214 VNPs are highly versatile due to their uniform size, self-assembly properties, and ability to be modified for targeted applications.

Chimeric VNPs combine components from different viruses or are engineered to display foreign peptides or proteins.215 Functionalized VNPs involve modification with chemical groups,216 targeting ligands,217 or imaging agents for specific applications, such as the use of cowpea mosaic virus particles functionalized with fluorescent dyes for imaging (Fig. 4).218

Comparison between virus-like particles (VLPs) and viral nanoparticles (VNPs).
Fig. 4  Comparison between virus-like particles (VLPs) and viral nanoparticles (VNPs).

VLPs is a subgroup of VNPs that lacks genetic material and therefore is non-infectious. Generally, is composed by only structural proteins and used as empty shells. On the other hand, the viral genome of the VNPs may be present, inactivated or modified and thus, retains partially its infectivity.

The generation of VLPs involves the use of yeast-based (among others) expression systems, since yeast can efficiently express and assemble viral proteins. The process begins by cloning the gene(s) encoding the viral structural protein(s) into a yeast expression vector, which is then introduced into yeast cells like Saccharomyces cerevisiae or Pichia pastoris.219 The yeast cells are cultured under conditions that induce the expression of the viral proteins, which then self-assemble into VLPs. These VLPs can be harvested from yeast cells or the culture medium and purified using techniques like centrifugation or chromatography. Yeast is particularly advantageous for VLP production due to its cost-effective, scalable nature and its capacity for performing post-translational modifications necessary for proper protein folding and assembly.220 Examples of VLP vaccines produced in yeast include the hepatitis B vaccine and the human papillomavirus (HPV) vaccine.221,222 Despite some challenges, such as differences in glycosylation patterns compared to mammalian cells and the production of VLPs derived from enveloped viruses,223 yeast remains a powerful and widely used platform for VLP production (Fig. 5). The development of VNPs and VLPs to treat cancer is related, among other things, to enhancing drug targeting against tumor cells. Thus, the efficiency of each model (VNPs or VLPs) depends on the targeted tumor.

Steps of virus-like particles (VLPs) production.
Fig. 5  Steps of virus-like particles (VLPs) production.

1. Production and expression: the viral structural genes are cloned, and after the viral proteins are expressed in different expression systems, including yeast, bacteria, plant cells, mammalian cells, and insect cells. 2. Purification of VLPs: to obtain VLPs with high purity and integrity, techniques like chromatography and ultracentrifugation are employed to purify the VLPs. 3. Formulation: in this last step, adjuvants and excipients are added to VLPs, enabling the production of VLPs vaccine, which will be administered to patients.

The use of VLPs in cancer research

Regarding the use of VLPs in cancer therapeutics, several approaches have been developed to both prevent and treat tumors. In the first case, VLPs are used in cancer vaccines to stimulate strong immune responses and long-term immunological memory. When injected, VLPs are taken up by immune cells, such as dendritic cells, activating both adaptive and innate immune responses (Fig. 6).224 In this respect, preventive vaccines based on VLPs have been developed for hepatitis B virus (HBV) and HPV.225 When VLPs are generated with structural proteins from HPV, eliciting strong immune responses against HPV infection, preventing the development of cervical cancer. These vaccines were approved by international regulatory agencies for human use, with names such as Engerix-B, Recombivax HB, Euvax, among others (for HBV),226 and Cervarix, Gardasil, and Gardasil 9 (for HPV).227 In breast cancer, HER2 is overexpressed in 20–30% of invasive breast tumors. This epitope is used to generate an immune response against HER2, inducing prophylactic vaccination so as to reduce the proliferation of tumors when already established.228 Other cancer vaccines using VLPs are those developed to treat pancreatic cancer,229 melanoma,230 and brain tumors.231 Another approach involves the generation of VLPs to display tumor-specific antigens, which train the immune system to recognize and attack established tumors, reducing their mass and contributing to overcoming resistance to conventional therapies. Challenges include identifying effective antigens, overcoming immune suppression in tumors, and the development of neo-antigens – antigens derived from tumor mutations.232

VLPs-inducing immunity.
Fig. 6  VLPs-inducing immunity.

Upon administration, VLPs are uptake by APC, like dendritic cells. VLPs can be presented by MHC-I or MHC-II, which are recognized by CD8+ and CD4+ T cells, respectively. For cellular response, CD8+ cells differentiate in cytotoxic T cells, releasing IFN-γ to exert their cytotoxicity activity in cancer cells. In humoral response, CD4+ cells differentiate into T helper cells (TH1/TH2), TH1 maintains the activity of cytotoxic T cells, and TH2 releases IL-4 and IL-10, inducing B cell activation. Once activated, B cells release IgG antibodies, enabling their antineoplasic effects. APC, antigen-presenting cells; IFN- γ, interferon-gamma; IgG, immunoglobulin G; IL-4/-10, interleukin-4/-10; MHC-I/II, major histocompatibility complex class I/II; TH, T helper cells; TNF-α, tumor necrosis factor-α; VLP, virus-like particles.

In addition, a widely studied approach is the use of VLPs loaded with chemotherapeutic drugs using the VLP’s surface specificity, which can be engineered to target tumoral tissue instead of normal tissue, also reducing systemic toxicity. Several works have demonstrated the efficiency of VLPs loaded with doxorubicin, showing enhanced efficiency against glioblastoma multiforme, among others.233,234 Monomethyl auristatin (MMAE) belongs to the group of the Auristatins, synthetic analogs of dolastatin 10 (D10), a highly cytotoxic antineoplastic agent derived from Dolabella auricularia. MMAE is usually conjugated with antibodies to treat lymphoma due to its high systemic toxicity,235 being administered as a pro-drug, Val-Cit linked MMAE. Shukla and co-workers evaluated the potato virus X VLPs conjugated with Val-Cit linked MMAE in vivo and in vitro, observing enhanced cytotoxicity in vitro and improved percent survival rates in vivo.236

Cisplatin, a platinum derivative used in chemotherapy for the last 30 years, was tested as cargo for VLPs by Hu and Steinmetz. In their work, VLPs of the physalis mottle virus were conjugated with maleimide-functionalized cisplatin, a prodrug that, upon cellular pH changes, generates cisplatin. They linked the prodrug to both the internal and external surfaces of physalis mottle virus-derived VLPs, and with this approach, they were able to carry cisplatin into cancer cells.237

Merging strategies

Most of the strategies discussed have their own advantages and drawbacks. To leverage the strengths of each, they have been combined to maximize their benefits, enhancing delivery efficiency and improving the overall treatment outcomes.

Synergistic combination of VLPs and hyperthermia

VLPs, hyperthermia, and magnetic hyperthermia represent cutting-edge technologies in the fields of nanomedicine and cancer therapy. Their combination offers a promising approach for targeted drug delivery, imaging, and thermal ablation of tumors.

Magnetic nanoparticles, typically composed of iron oxides like magnetite (Fe3O4) or maghemite (γ-Fe2O3), are injected into the tumor site and activated by an external AMF.160 The controlled heating induces apoptosis or necrosis in cancer cells while sparing surrounding healthy tissue.

Several groups have explored combinations of both techniques by adding one approach to another. Hoopes and co-workers evaluated the effect of intra-tumoral magnetic nanoparticle hyperthermia in an in vivo model for MTG-B mammary adenocarcinoma. They treated the tumor with iron oxide nanoparticles, applying an AMF, and added a modified version of the cowpea mosaic virus VLPs to induce a stronger immune response. In fact, this combined treatment enhanced the expression of pro-inflammatory cytokines like tumor necrosis factor-alpha, interleukin-6, enhanced the immunogenicity against the tumor, and reduced the tumor mass through cell death.238

The generation of VLPs with a magnetic core can be used in a series of applications, such as targeted drug delivery,239 magnetic bioseparation,240 and MRI contrast agents,241 among others.242 The use of superparamagnetic nanoparticles is preferred due to their reversible magnetization, which avoids NP aggregation.

It was already well established that synthetic nanocarriers can be thermally responsive, leading to chemotherapy drug delivery controlled by temperature,243 but in 2019, Thong and co-workers developed a multifunctional nanovehicle based on the VLP of Macrobrachium rosenbergii nodavirus. In this work, they covalently conjugated folic acid to lysine residues located on the surface of Macrobrachium rosenbergii nodavirus, while doxorubicin was loaded inside the VLP. The objective was to deliver the cargo (doxorubicin) to tumor cells (HT29 colorectal cancer) rich in folic acid receptor (FR) using hyperthermia. They also tested two other cell lines with lower FR expression, and they concluded that, in fact, in the cells with higher FR receptors, the VLP uptake was higher.244

Intranasal delivery using different combinations for brain tumors

Primary brain tumors are classified as primary central nervous system (CNS) lymphoma, malignant ependymomas, meningiomas, lower-grade gliomas (I-III), and glioblastomas (Grade IV glioma). In addition, secondary brain tumors derived from metastasis from lung and breast tumors can also pose a threat to the patient’s life. Glioblastoma is the most aggressive brain tumor, characterized by necrosis, microvascular proliferation, and rapid, infiltrative growth. Glioblastoma can arise as a primary tumor or develop secondarily from lower-grade gliomas. Treatment for patients with good performance status includes surgery, radiation, and chemotherapy, and even with the development of targeted therapy, the prognosis remains poor, with a median survival of 14–16 months and a five-year overall survival rate of 9.8%.245

Developing drugs whose action occurs in the brain is challenging, largely because of the BBB: endothelial cells in capillaries and other microvasculature of cerebral tissue, which function to protect the CNS against xenobiotics and maintain homeostasis, but impair the satisfactory delivery of pharmacological agents to the CNS.246 A promising solution is intranasal drug delivery, which bypasses this barrier noninvasively by transporting drugs directly to the brain via the olfactory and trigeminal nerves.

The olfactory neurons extend axons through the cribriform plate. These axons are wrapped in the olfactory ensheathing cells and neural fibroblasts that form a protective sheath connected to the brain’s protective layers (meninges), allowing drugs to move directly into the brain. These neurons are unmyelinated and bipolar, meaning they have two extensions. They can carry drugs to the CNS through the intracellular pathway, where drugs are taken up by olfactory sensory cells, transported along their axons to the olfactory bulb in the brain, and then spread to other brain regions through a process called transynaptic transport; and the extracellular pathway, where the drugs move through the spaces between cells in the nasal epithelium, then travel along the outside of nerve fibers to reach the brain’s cerebrospinal fluid.247

Intranasal delivery is a promising method for treating brain-related conditions because it bypasses the BBB, a major obstacle for many drugs. It is non-invasive and allows drugs to reach the brain directly. Furthermore, other benefits include bypassing liver drug metabolism, reducing systemic toxicity.248

Problems with intranasal administration

Despite these advantages, intranasal administration of drugs also faces drawbacks, such as the reduction of drug penetration within the nasal epithelial membrane in the nasal chamber due to efflux transporters (P-glycoprotein),249 metabolic enzymes, and mucociliary clearance.250 Other side effects include sneezing, bleeding, among others.251 Thus, alternatives for delivering drugs to the brain via the intranasal route using nanotechnology have been investigated.

Viral oncolysis through intranasal delivery

While in VLPs, natural viruses lacking genomes (replication-incompetent) are used, in viral oncolysis (VO), natural or engineered viruses that maintain their replication capacity are used. They have an intrinsic or induced capacity to replicate only in tumor tissue, leaving normal cells intact. This feature is related to the defective antiviral response of tumor cells (e.g., type I interferon response), dysfunctional tumor suppressor proteins (e.g., p53 or pRb), and overexpression of tumor survival factors in cancer cells. Normal cells are spared because their antiviral defense systems and tumor suppressor pathways remain intact.252 There are a few naturally occurring oncolytic viruses; in addition, pathogenic viruses can be genetically modified to enhance tumor selectivity by removing virulence factors critical for infecting normal cells. Viral infection relies on cell surface receptors and intracellular interactions, with tumor-specific tropism. Once infected, these cells undergo strong proliferation, inducing tumor cell lysis. In normal cells, interferon activation inhibits viral replication, providing an additional layer of selectivity.253 Therefore, this therapeutic modality depends on the ability of the virus to maintain its intact replication capacity.

Several studies have shown the capacity of viral strains to infect and kill brain tumors in vivo, in vitro, and in clinical studies.254 Oncolytic viruses already studied for brain tumors include oncolytic H-1 parvovirus, herpes simplex virus-1, Reovirus, among others, with non-standardized delivery methods, including intratumoral injection, intravenous,255 and also intranasal routes, using vesicular stomatitis virus.256 However, this approach has a limited effect as monotherapy.257

To improve the efficiency of viral delivery for viral oncolysis, this method was combined with intranasal cellular delivery of oncolytic viruses. This promising approach involves the use of mesenchymal stem cells (MSCs) as anticancer therapeutics, as they are able to deliver proteins, genes, or oncolytic vectors.258 This strategy is based on the observation that MSCs can cross the cribriform plate and migrate through the olfactory bulb to other parts of the brain, representing a non-invasive method for cell delivery to the CNS (Fig. 7).259 Thus, this strategy uses MSCs loaded with oncolytic viruses to directly reach the brain,260,261 avoiding the side effects related to direct intranasal drug delivery.

Representation of viral oncolysis via intranasal pathway (IN).
Fig. 7  Representation of viral oncolysis via intranasal pathway (IN).

Intranasal inoculation of the oncolytic virus leads to olfactory nerve transport into the brain, infecting and killing tumors, while leaving normal cells intact, reinforcing the viral selectivity against tumor.

Using neurotropic viruses to generate VLPs

The VLPs under investigation to treat brain tumors mostly use the intravenous route or intratumoral injection, but data on the intranasal route through olfactory neurons have not been explored, as evidenced by the lack of literature data. Even though VLPs derived from non-neurotropic viruses can be modified to target neural cells,262 one way to improve the specificity of these VLPs is to use viruses that have natural neurotropism.

In 2018, Chao and collaborators developed VLPs derived from the neurotropic JC polyomavirus, which infects glial cells and oligodendrocytes and causes fatal progressive multifocal leukoencephalopathy in patients with AIDS. They investigated the feasibility of a gene therapy strategy for glioblastoma using JC polyomavirus VLPs as a gene delivery vector.263

In contrast to non-replicating VLPs, oncolytic viruses are replicating viruses designed to destroy tumor cells. Examples include adenoviruses, herpes simplex virus, and rotaviruses, some of which are approved or under clinical trials for cancer therapy.264 Notably, the Zika virus (ZIKV) has emerged as a promising oncolytic virus for treating glioblastoma due to its neurotropic nature and ability to infect glioma stem cells, which are resistant to conventional therapies like chemotherapy and radiation. ZIKV’s specificity for glioma stem cells is linked to the expression of the AXL receptor, SOX2, and αVβ5 integrin, which are highly expressed in these cells.265 Additionally, the Musashi-1 protein, which is overexpressed in tumors but not in most healthy tissues, enhances ZIKV replication in cancer cells while limiting side effects in patients due to restricted viral replication in normal tissues.

VLPs combined with other nanocarriers

Even though VNPs and VLPs represent an innovation in cancer research and treatment, there are some issues that limit their efficiency. Among the limitations of VLPs regarding drug delivery are: payload capacity, pre-existing immunity, difficulty in crossing biological barriers, penetrating solid tumors, and physical instability. As can be seen below, and in Table 3, some combinations of VLPs with other nanocarriers have the potential to solve some of these problems.206,266–273

Table 3

Limitations and cutting-edge solutions achieved with the combinations of different nanocarriers with VLPs

StrategyProblems solvedRemainder challengesRef
VLP + DrugEnhanced drug specificity towards the tumoral tissue.Payload capacity, Low cell uptake, rapid clearance, pre-existing immunity, difficulty in cross biological barriers (BBB) and penetrate solid tumors and physical instability.206
VLP + micromotorsEnhanced distribution and delivery of the payload.NP Agregation266
VLPs of plant originEvade immune neutralization, cost- effective and environmentally sustainableWeak and transient immunological responses (IgG and IgE)267
VLP + GoldNpEnhanced photothermal efficiency using plasmon nanobubles (potential to induce transient BBB opening)N.D268
VLP + drug + GoldNpEnhanced drug deliveryN.D270
VLP + Metalic NPsEnhanced physical stability of the VLPsN.D269
VLP + Magnetic NPEnhanced payload capacity; Enhanced biocompatibility of magnetic NPsIrregular aggregates and a large fraction of empty capsids271,272
VLP + Silica NP (biomimetic)Enhanced silica NP´cell uptake, improved immune Responses against CancerN.D.273

Payload capacity

VLPs may present low payload capacity. They have limited internal space, restricting the amount of drugs they can carry. Among the recent solutions to improve the delivery of VLPs’ payload, Mg-based micromotors have been designed. This strategy utilizes the autonomous motion of biocompatible micromotors to actively deliver Qβ VLPs’ payloads within the tumor area, enhancing the local distribution and retention time.266 These micromotors utilize a core made of biocompatible and biodegradable Mg microparticle ‘engine’ covered with gold. The gold layer allows the micromotor chemical reaction with the water fuel, generating hydrogen bubbles in biological media. This method was evaluated to treat ovarian cancer in vivo.266

Pre-existing immunity

For the VLP to successfully reach the target tissues, it needs to avoid immune neutralization. The immunological response towards VLPs in cancer can reduce their efficiency, since those used for the prevention of cancer development (vaccines against oncoviruses) mostly use VLPs built of human viruses (such as HBV or HPV VLPs).274 For the treatment of established tumors such as breast cancer and melanoma, human viruses are also used. In both cases, this can lead to the development of antibodies against VLP components, leading to neutralization and reducing the therapeutic response.275 A solution in this case is the use of plant-based VLPs for targeting human tumor tissues; they can be targeted to the tumor tissue, but since they are derived from a plant virus, there’s no off-target binding or immunological clearance. In addition, compared to the generation of synthetic or animal-derived nanoparticles, the synthesis of these plant-based VLPs is scalable, cost-effective, and environmentally sustainable. Even though a weak immunological response was observed, no signs of allergy were detected.267

Low permeability in the BBB

Improving the permeability of the BBB is critical for the treatment of brain tumors, and several cutting-edge strategies have been designed to achieve this goal. Parsamian et al.268 engineered gold nanoparticles conjugated to Qβ VLPs, generating a polyvalent VLP-gold nanosystem, where the VLPs are decorated with gold nanoparticles to generate photothermal-induced nanobubbles. The transient nanobubble cavitation and collapse can temporarily induce openings in the blood–brain barrier.268 Without conjugation with VLPs, gold nanostructures can induce tissue damage. This combination enhanced the efficiency of photothermal therapy.

VLPs’ physical instability

Among the strategies where VLPs are used, there is antigen presentation. The size of the inserts in viral surface proteins is associated with increased instability.276 Different viruses need different optimization protocols to reduce VLP instability.277 More recent approaches to reduce VLP instability use metallic nanoparticles coated with VLPs,269 also enhancing drug delivery.270

Magnetic nanoparticles coated with VLPs

Magnetic nanoparticles have been developed for MRI contrast agents and cancer treatment, but among the issues detected was dispersibility in human fluids such as serum and blood.271 They also have the potential to enhance payload delivery.278 Coating magnetic nanoparticles with VLPs improved the dispersibility, and the conjugation with tumor-specific ligands (such as epidermal growth factor) efficiently targets tumor cells.272

Topology mimicking strategy

The topology of organic VLPs led researchers to develop methods to synthesize inorganic VLPs (viral-mimicking topography), to enhance internalization and efficiency of silica nanoparticles.279 Kim et al.273 developed a method where organic VLPs are used as a template to build silica nanoparticles. In their work, they produced VLPs made of HPV16 structural protein, encapsulated doxorubicin, and then performed controlled silicification of HPV16 VLPs, creating a silica nanocage.273 Their results showed higher biocompatibility and cellular uptake than conventional mesoporous silica nanoparticles. However, the long-term toxicity of inorganic nanoparticles caused by accumulation in the reticuloendothelial system remains a challenge for clinical application.280

Limitations of this review

While this review provides an overview of the advancements in combining conventional anticancer therapies (drugs and antibodies) with nanoparticles, magnetic hyperthermia, and VLPs, it has certain limitations. We focused on innovative approaches and challenges but did not extensively cover all possible combinations of these therapies, potentially omitting some emerging or niche approaches. Given the fast evolution of this field, some recent studies or alternative viewpoints might not be included.

Future directions

Technological progress has allowed the integration of multiple strategies that individually enhance anti-tumor treatment efficacy. Synthetic nanoparticles, VNPs, and VLPs, when combined with chemotherapy and hyperthermia, improve targeting accuracy and minimize drug exposure to healthy tissues, particularly in resistant cancers. Intranasal drug delivery, which bypasses the BBB, also offers a promising approach for treating brain tumors. Despite these advancements, cancer remains a critical global health challenge, and widespread access to these innovative therapies continues to be a major barrier.

Magnetic hyperthermia is a promising technique for treating cancer, but it still faces several important limitations that hinder its widespread clinical use. One of the main challenges is the efficacy of delivering magnetic nanoparticles specifically to the tumor site. It is difficult to ensure that enough particles reach the tumor without affecting healthy tissue and to ensure uniform distribution. This uneven distribution can cause inconsistent heating, which reduces the effectiveness of the treatment and increases the risk of damage to nearby healthy tissue. Controlling and monitoring the temperature during treatment is also a major issue. Since heating depends on the concentration and distribution of nanoparticles, it is difficult to predict or regulate the temperature rise. In addition, accurately measuring the internal temperature in real time during treatment remains a technical challenge. There are also concerns about the materials used. Some nanoparticles can be toxic, especially if they are not biocompatible or if they remain in the body for a long time. Their long-term effects are not fully understood, and there is still no standard for what types of particles, coatings, or doses should be used. Another limitation is efficiency. Not all nanoparticles produce sufficient heat, especially when limited by the safe range of magnetic fields that can be used in humans. Stronger fields can increase heating, but they risk causing harm, such as nerve stimulation or tissue damage. From a clinical and practical perspective, magnetic hyperthermia is not yet widely available. The equipment is expensive, clinical trials are limited, and few nanoparticle formulations have been approved for routine use. It is also primarily suited for more localized tumors, which limits its use in the treatment of disseminated or metastatic cancers.

Despite its challenges and limitations, the technique offers a fundamentally different mechanism of action than biochemical or molecular approaches. This makes it particularly useful for overcoming certain forms of drug resistance and for treating tumors that are less responsive to traditional therapies. As demonstrated, a major advantage lies in its strong potential for synergistic use with other treatment modalities. When combined with chemotherapy or radiotherapy, magnetic hyperthermia can enhance therapeutic outcomes by sensitizing tumor cells, increasing drug uptake, improving tissue oxygenation, and activating temperature-sensitive drug delivery systems. This multimodal capability makes it a versatile tool in personalized cancer therapy. In addition, the repeatability of magnetic hyperthermia treatments, made possible by the permanent presence of nanoparticles and the non-invasive nature of magnetic field application, adds a practical dimension to its appeal. Patients can undergo multiple treatment sessions without the need for additional invasive procedures or systemic drug administration.

In summary, while magnetic hyperthermia has real potential, issues related to targeting, control, safety, efficiency, and clinical readiness need to be addressed before it becomes a standard cancer therapy.

The application of VLPs faces several challenges, such as the complexity of large-scale production with high purity and stability. The variation in the immunogenicity of VLPs may affect their efficacy, while improvements in surface modifications, with special focus on tumor neo-antigens, may be considered to ensure efficient delivery to cancer cells, minimizing uptake by healthy tissues. Among magnetic hyperthermia challenges, there are difficulties in controlling heat distribution in target tissues, which can lead to damage to healthy cells, tumor heterogeneity and variations in nanoparticle concentration, and the development of nanoparticles with optimized magnetic properties, capable of generating sufficient heat in clinically acceptable magnetic fields.

Conclusions

Nanoparticles-based anticancer drug delivery improves cancer treatment, but tumors such as gliomas, imposes challenges that require alternative approaches. Even though intratumoral injection of nanoparticles to deliver drugs, therapeutic molecules or induce hyperthermia, improved the response to treatment, variable drug distribution within the tumor and potential leakage into surrounding non-tumoral tissue, uneven heat distribution (for hyperthermia), may reduce efficiency and increase toxicity.

The targeting capacity of VNPs and VLPs dramatically reduced toxicity and improved the antitumor response. The approval of VLPs for prophylactic vaccines and for cancer treatment, such as melanoma and breast cancer, has proven the success of this approach.

However, VLPs as monotherapy are less effective than when combined with conventional immunotherapy. Furthermore, the complex production of VLPs limits its scalability. Drug-loading efficiency, expensive cell culture systems, purification steps, batch-to-batch variability, and structural stability remain significant issues.

Thus, associating the targeting capacity of VNPs and VLPs with the efficiency of traditional drugs, immunotherapy, photothermal, gene therapies, and other nanocarriers may improve the results, leading to a concentrated effort to optimize VNPs and VLPs production, enhancing technical and economic viability, even though most of the optimization studies and their outcomes remain in the pre-clinical stage.

Declarations

Acknowledgement

None.

Funding

The present study was supported by the Research Support Foundation of Rio de Janeiro, grant No. E-26/201.591/2021.

Conflict of interest

Dr. Janaina Fernandes has been an editorial board member of the Journal of Exploratory Research in Pharmacology since November 2021. The authors have no other conflicts of interest to declare.

Authors’ contributions

Study concept and design (RRB, LASO, JF), acquisition of the data (RRB, LASO, GSP, JF), assay performance and data analysis (RRB, LASO, GSP, JF), drafting of the manuscript (RRB, LASO, GSP, JF), and critical revision of the manuscript (RRB, LASO, JF). All authors have approved the final version and publication of the manuscript.

References

  1. Wilkinson JM. Nanotechnology applications in medicine. Med Device Technol 2003;14(5):29-31 PubMed/NCBI
  2. Roco MC. Nanotechnology: convergence with modern biology and medicine. Curr Opin Biotechnol 2003;14(3):337-346 View Article PubMed/NCBI
  3. Wilczewska AZ, Niemirowicz K, Markiewicz KH, Car H. Nanoparticles as drug delivery systems. Pharmacol Rep 2012;64(5):1020-1037 View Article PubMed/NCBI
  4. Gavas S, Quazi S, Karpiński TM. Nanoparticles for Cancer Therapy: Current Progress and Challenges. Nanoscale Res Lett 2021;16(1):173 View Article PubMed/NCBI
  5. Guggina LM, Choi AW, Choi JN. EGFR Inhibitors and Cutaneous Complications: A Practical Approach to Management. Oncol Ther 2017;5(2):135-148 View Article
  6. Jarmila P, Veronika M, Peter M. Advances in the delivery of anticancer drugs by nanoparticles and chitosan-based nanoparticles. Int J Pharm X 2024;8:100281 View Article PubMed/NCBI
  7. Manzanares D, Ceña V. Endocytosis: The Nanoparticle and Submicron Nanocompounds Gateway into the Cell. Pharmaceutics 2020;12(4):371 View Article PubMed/NCBI
  8. Li Z, Zhang Y, Zhu D, Li S, Yu X, Zhao Y, et al. Transporting carriers for intracellular targeting delivery via non-endocytic uptake pathways. Drug Deliv 2017;24(sup1):45-55 View Article PubMed/NCBI
  9. Meacham JM, Durvasula K, Degertekin FL, Fedorov AG. Physical methods for intracellular delivery: practical aspects from laboratory use to industrial-scale processing. J Lab Autom 2014;19(1):1-18 View Article PubMed/NCBI
  10. Kaksonen M, Roux A. Mechanisms of clathrin-mediated endocytosis. Nat Rev Mol Cell Biol 2018;19(5):313-326 View Article PubMed/NCBI
  11. Rennick JJ, Johnston APR, Parton RG. Key principles and methods for studying the endocytosis of biological and nanoparticle therapeutics. Nat Nanotechnol 2021;16(3):266-276 View Article PubMed/NCBI
  12. Wang Z, Tiruppathi C, Cho J, Minshall RD, Malik AB. Delivery of nanoparticle: complexed drugs across the vascular endothelial barrier via caveolae. IUBMB Life 2011;63(8):659-667 View Article PubMed/NCBI
  13. Cheng X, Chen K, Dong B, Yang M, Filbrun SL, Myoung Y, et al. Dynamin-dependent vesicle twist at the final stage of clathrin-mediated endocytosis. Nat Cell Biol 2021;23(8):859-869 View Article PubMed/NCBI
  14. Parton RG, Taraska JW, Lundmark R. Is endocytosis by caveolae dependent on dynamin?. Nat Rev Mol Cell Biol 2024;25(7):511-512 View Article PubMed/NCBI
  15. Lajoie P, Nabi IR. Regulation of raft-dependent endocytosis. J Cell Mol Med 2007;11(4):644-653 View Article PubMed/NCBI
  16. Niedergang F, Grinstein S. How to build a phagosome: new concepts for an old process. Curr Opin Cell Biol 2018;50:57-63 View Article PubMed/NCBI
  17. Chen F, Wang G, Griffin JI, Brenneman B, Banda NK, Holers VM, et al. Complement proteins bind to nanoparticle protein corona and undergo dynamic exchange in vivo. Nat Nanotechnol 2017;12(4):387-393 View Article PubMed/NCBI
  18. Su MJ, Parayath NN, Amiji MM. Exosome-Mediated Communication in the Tumor Microenvironment. In: Amiji M, Ramesh R (eds). Diagnostic and Therapeutic Applications of Exosomes in Cancer. Cambridge, MA: Academic Press; 2018:187-218 View Article
  19. Sharma A, Vaghasiya K, Ray E, Verma RK. Lysosomal targeting strategies for design and delivery of bioactive for therapeutic interventions. J Drug Target 2018;26(3):208-221 View Article PubMed/NCBI
  20. Wei H, Hao Y, Zhang J, Qi Y, Feng C, Zhang C. Advances in lysosomal escape mechanisms for gynecological cancer nano-therapeutics. J Pharm Anal 2024;14(12):101119 View Article PubMed/NCBI
  21. Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov 2021;20(2):101-124 View Article PubMed/NCBI
  22. Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024;74(3):229-263 View Article PubMed/NCBI
  23. Fu L, Ma X, Liu Y, Xu Z, Sun Z. Applying nanotechnology to boost cancer immunotherapy by promoting immunogenic cell death. Chinese Chem Lett 2022;33(4):1718-1728 View Article
  24. Dai H, Cheng Z, Zhang T, Wang W, Shao J, Wang W, et al. Boron difluoride formazanate dye for high-efficiency NiR-ii fluorescence imaging-guided cancer photo thermal therapy. Chin Chem Lett 2022;33(5):2501-2506 View Article
  25. Gouw AM, Kumar V, Resendez A, Alvina FB, Liu NS, Margulis K, et al. Azapodophyllotoxin Causes Lymphoma and Kidney Cancer Regression by Disrupting Tubulin and Monoglycerols. ACS Med Chem Lett 2022;13(4):615-622 View Article PubMed/NCBI
  26. Zafar A, Khatoon S, Khan MJ, Abu J, Naeem A. Advancements and limitations in traditional anti-cancer therapies: a comprehensive review of surgery, chemotherapy, radiation therapy, and hormonal therapy. Discov Oncol 2025;16(1):607 View Article PubMed/NCBI
  27. Wang H, Yu J, Lu X, He X. Nanoparticle systems reduce systemic toxicity in cancer treatment. Nanomedicine (Lond) 2016;11(2):103-106 View Article PubMed/NCBI
  28. Sanna V, Sechi M. Therapeutic Potential of Targeted Nanoparticles and Perspective on Nanotherapies. ACS Med Chem Lett 2020;11(6):1069-1073 View Article PubMed/NCBI
  29. Caputo TM, Barisciano G, Mulè C, Cusano AM, Aliberti A, Muccillo L, et al. Development of High-Loading Trastuzumab PLGA Nanoparticles: A Powerful Tool Against HER2 Positive Breast Cancer Cells. Int J Nanomedicine 2023;18:6999-7020 View Article PubMed/NCBI
  30. Hrkach J, Von Hoff D, Mukkaram Ali M, Andrianova E, Auer J, Campbell T, et al. Preclinical development and clinical translation of a PSMA-targeted docetaxel nanoparticle with a differentiated pharmacological profile. Sci Transl Med 2012;4(128):128ra39 View Article PubMed/NCBI
  31. Sharma A, Sharma US. Liposomes in drug delivery: Progress and limitations. Int J Pharm 1997;154(2):123-140 View Article
  32. Fenske DB, Cullis PR. Liposomal nanomedicines. Expert Opin Drug Deliv 2008;5(1):25-44 View Article PubMed/NCBI
  33. Byrne JD, Betancourt T, Brannon-Peppas L. Active targeting schemes for nanoparticle systems in cancer therapeutics. Adv Drug Deliv Rev 2008;60(15):1615-1626 View Article PubMed/NCBI
  34. Brannon-Peppas L, Blanchette JO. Nanoparticle and targeted systems for cancer therapy. Adv Drug Deliv Rev 2004;56(11):1649-1659 View Article PubMed/NCBI
  35. Aliabadi HM, Lavasanifar A. Polymeric micelles for drug delivery. Expert Opin Drug Deliv 2006;3(1):139-162 View Article PubMed/NCBI
  36. Kontermann RE. Immunoliposomes for cancer therapy. Curr Opin Mol Ther 2006;8(1):39-45 PubMed/NCBI
  37. Silva R, Ferreira H, Cavaco-Paulo A. Sonoproduction of liposomes and protein particles as templates for delivery purposes. Biomacromolecules 2011;12(10):3353-3368 View Article PubMed/NCBI
  38. dos Santos Giuberti C, de Oliveira Reis EC, Ribeiro Rocha TG, Leite EA, Lacerda RG, Ramaldes GA, et al. Study of the pilot production process of long-circulating and pH-sensitive liposomes containing cisplatin. J Liposome Res 2011;21(1):60-69 View Article PubMed/NCBI
  39. Afergan E, Epstein H, Dahan R, Koroukhov N, Rohekar K, Danenberg HD, et al. Delivery of serotonin to the brain by monocytes following phagocytosis of liposomes. J Control Release 2008;132(2):84-90 View Article PubMed/NCBI
  40. Turkova A, Roilides E, Sharland M. Amphotericin B in neonates: deoxycholate or lipid formulation as first-line therapy - is there a ‘right’ choice?. Curr Opin Infect Dis 2011;24(2):163-171 View Article PubMed/NCBI
  41. Paavola A, Kilpeläinen I, Yliruusi J, Rosenberg P. Controlled release injectable liposomal gel of ibuprofen for epidural analgesia. Int J Pharm 2000;199(1):85-93 View Article PubMed/NCBI
  42. 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 Pharm 2011;8(4):1002-1015 View Article PubMed/NCBI
  43. Wissing SA, Kayser O, Müller RH. Solid lipid nanoparticles for parenteral drug delivery. Adv Drug Deliv Rev 2004;56(9):1257-1272 View Article PubMed/NCBI
  44. Abdel-Mottaleb MM, Neumann D, Lamprecht A. Lipid nanocapsules for dermal application: a comparative study of lipid-based versus polymer-based nanocarriers. Eur J Pharm Biopharm 2011;79(1):36-42 View Article PubMed/NCBI
  45. Nayak AP, Tiyaboonchai W, Patankar S, Madhusudhan B, Souto EB. Curcuminoids-loaded lipid nanoparticles: novel approach towards malaria treatment. Colloids Surf B Biointerfaces 2010;81(1):263-273 View Article PubMed/NCBI
  46. Attama AA, Schicke BC, Paepenmüller T, Müller-Goymann CC. Solid lipid nanodispersions containing mixed lipid core and a polar heterolipid: characterization. Eur J Pharm Biopharm 2007;67(1):48-57 View Article PubMed/NCBI
  47. Lahiri A, Balamuralidhara V. Chapter 7 - Solid lipid nanoparticles: A potential drug carrier in pulmonary system. In: Faiyazuddin MD, Ali H, Akbar MD, Iqbal B (eds). Lipids in Pulmonary Drug Delivery. Academic Press; 2025:121-132 View Article
  48. Sznitowska M, Gajewska M, Janicki S, Radwanska A, Lukowski G. Bioavailability of diazepam from aqueous-organic solution, submicron emulsion and solid lipid nanoparticles after rectal administration in rabbits. Eur J Pharm Biopharm 2001;52(2):159-163 View Article PubMed/NCBI
  49. Kovacevic A, Savic S, Vuleta G, Müller RH, Keck CM. Polyhydroxy surfactants for the formulation of lipid nanoparticles (SLN and NLC): effects on size, physical stability and particle matrix structure. Int J Pharm 2011;406(1-2):163-172 View Article PubMed/NCBI
  50. da Rocha MCO, da Silva PB, Radicchi MA, Andrade BYG, de Oliveira JV, Venus T, et al. Docetaxel-loaded solid lipid nanoparticles prevent tumor growth and lung metastasis of 4T1 murine mammary carcinoma cells. J Nanobiotechnology 2020;18(1):43 View Article PubMed/NCBI
  51. Llaguno-Munive M, Vazquez-Lopez MI, Garcia-Lopez P. Solid Lipid Nanoparticles, an Alternative for the Treatment of Triple-Negative Breast Cancer. Int J Mol Sci 2024;25(19):10712 View Article PubMed/NCBI
  52. Bai J, Li Y, Du J, Wang S, Zheng J, Yang Q, et al. One-pot synthesis of polyacrylamide-gold nanocomposite. Mater Chem Phys 2007;106(2-3):412-415 View Article
  53. Turos E, Shim JY, Wang Y, Greenhalgh K, Reddy GS, Dickey S, et al. Antibiotic-conjugated polyacrylate nanoparticles: new opportunities for development of anti-MRSA agents. Bioorg Med Chem Lett 2007;17(1):53-56 View Article PubMed/NCBI
  54. Martínez A, Iglesias I, Lozano R, Teijón JM, Blanco MD. Synthesis and characterization of thiolated alginate-albumin nanoparticles stabilized by disulfide bonds. Evaluation as drug delivery systems. Carbohydr Polym 2011;83(3):1311-1321 View Article
  55. Mao HQ, Roy K, Troung-Le VL, Janes KA, Lin KY, Wang Y, et al. Chitosan-DNA nanoparticles as gene carriers: synthesis, characterization and transfection efficiency. J Control Release 2001;70(3):399-421 View Article PubMed/NCBI
  56. Saraogi GK, Gupta P, Gupta UD, Jain NK, Agrawal GP. Gelatin nanocarriers as potential vectors for effective management of tuberculosis. Int J Pharm 2010;385(1-2):143-149 View Article PubMed/NCBI
  57. Mainardes RM, Khalil NM, Gremião MP. Intranasal delivery of zidovudine by PLA and PLA-PEG blend nanoparticles. Int J Pharm 2010;395(1-2):266-271 View Article PubMed/NCBI
  58. Park J, Fong PM, Lu J, Russell KS, Booth CJ, Saltzman WM, et al. PEGylated PLGA nanoparticles for the improved delivery of doxorubicin. Nanomedicine 2009;5(4):410-418 View Article PubMed/NCBI
  59. Fritzen-Garcia MB, Zanetti-Ramos BG, de Oliveira CS, Soldi V, Pasa AA, Creczynski-Pasa TB. Atomic force microscopy imaging of polyurethane nanoparticles onto different solid substrates. Mater Sci Eng C 2009;29(2):405-409 View Article
  60. Torchilin V. Multifunctional Pharmaceutical Nanocarriers: Development of the Concept. In: Torchilin V (ed). Multifunctional Pharmaceutical Nanocarriers. New York, NY: Springer New York; 2008:1-32 View Article
  61. Luo G, Yu X, Jin C, Yang F, Fu D, Long J, et al. LyP-1-conjugated nanoparticles for targeting drug delivery to lymphatic metastatic tumors. Int J Pharm 2010;385(1-2):150-156 View Article PubMed/NCBI
  62. Mora-Huertas CE, Fessi H, Elaissari A. Polymer-based nanocapsules for drug delivery. Int J Pharm 2010;385(1-2):113-142 View Article PubMed/NCBI
  63. Lee WH, Loo CY, Leong CR, Young PM, Traini D, Rohanizadeh R. The achievement of ligand-functionalized organic/polymeric nanoparticles for treating multidrug resistant cancer. Expert Opin Drug Deliv 2017;14(8):937-957 View Article PubMed/NCBI
  64. Caraway CA, Gaitsch H, Wicks EE, Kalluri A, Kunadi N, Tyler BM. Polymeric Nanoparticles in Brain Cancer Therapy: A Review of Current Approaches. Polymers (Basel) 2022;14(14):2963 View Article PubMed/NCBI
  65. Tomalia DA, Baker H, Dewald J, Hall M, Kallos G, Martin S, et al. A New Class of Polymers: Starburst-Dendritic Macromolecules. Polym J 1985;17(1):117-132 View Article
  66. Quintana A, Raczka E, Piehler L, Lee I, Myc A, Majoros I, et al. Design and function of a dendrimer-based therapeutic nanodevice targeted to tumor cells through the folate receptor. Pharm Res 2002;19(9):1310-1316 View Article PubMed/NCBI
  67. Caminade AM, Laurent R, Majoral JP. Characterization of dendrimers. Adv Drug Deliv Rev 2005;57(15):2130-2146 View Article PubMed/NCBI
  68. Kihara F, Arima H, Tsutsumi T, Hirayama F, Uekama K. Effects of structure of polyamidoamine dendrimer on gene transfer efficiency of the dendrimer conjugate with alpha-cyclodextrin. Bioconjug Chem 2002;13(6):1211-1219 View Article PubMed/NCBI
  69. Singh P, Gupta U, Asthana A, Jain NK. Folate and folate-PEG-PAMAM dendrimers: synthesis, characterization, and targeted anticancer drug delivery potential in tumor bearing mice. Bioconjug Chem 2008;19(11):2239-2252 View Article PubMed/NCBI
  70. Wängler C, Moldenhauer G, Eisenhut M, Haberkorn U, Mier W. Antibody-dendrimer conjugates: the number, not the size of the dendrimers, determines the immunoreactivity. Bioconjug Chem 2008;19(4):813-820 View Article PubMed/NCBI
  71. Waite CL, Roth CM. PAMAM-RGD conjugates enhance siRNA delivery through a multicellular spheroid model of malignant glioma. Bioconjug Chem 2009;20(10):1908-1916 View Article PubMed/NCBI
  72. Tosh DK, Yoo LS, Chinn M, Hong K, Kilbey SM, Barrett MO, et al. Polyamidoamine (PAMAM) dendrimer conjugates of “clickable” agonists of the A3 adenosine receptor and coactivation of the P2Y14 receptor by a tethered nucleotide. Bioconjug Chem 2010;21(2):372-384 View Article PubMed/NCBI
  73. Balogh L, Swanson DR, Tomalia DA, Hagnauer GL, McManus AT. Dendrimer−Silver Complexes and Nanocomposites as Antimicrobial Agents. Nano Lett 2001;1(1):18-21 View Article
  74. Lopez AI, Reins RY, McDermott AM, Trautner BW, Cai C. Antibacterial activity and cytotoxicity of PEGylated poly(amidoamine) dendrimers. Mol Biosyst 2009;5(10):1148-1156 View Article PubMed/NCBI
  75. Arora V, Abourehab MAS, Modi G, Kesharwani P. Dendrimers as prospective nanocarrier for targeted delivery against lung cancer. Eur Polym J 2022;180:111635 View Article
  76. Czarnobaj K. Preparation and characterization of silica xerogels as carriers for drugs. Drug Deliv 2008;15(8):485-492 View Article PubMed/NCBI
  77. Hench LL, West JK. The sol-gel process. Chem Rev 1990;90(1):33-72 View Article
  78. Prokopowicz M, Łukasiak J. Synthesis and in vitro characterization of freeze-dried doxorubicin-loaded silica/PEG composite. J Non CrystSolids 2010;356(33-34):1711-1720 View Article
  79. Czarnobaj K, Lukasiak J. In vitro release of cisplatin from sol-gel processed organically modified silica xerogels. J Mater Sci Mater Med 2007;18(10):2041-2044 View Article PubMed/NCBI
  80. Hilder TA, Hill JM. Modeling the loading and unloading of drugs into nanotubes. Small 2009;5(3):300-308 View Article PubMed/NCBI
  81. Beg S, Rizwan M, Sheikh AM, Hasnain MS, Anwer K, Kohli K. Advancement in carbon nanotubes: basics, biomedical applications and toxicity. J Pharm Pharmacol 2011;63(2):141-163 View Article PubMed/NCBI
  82. Zhang B, Chen Q, Tang H, Xie Q, Ma M, Tan L, et al. Characterization of and biomolecule immobilization on the biocompatible multi-walled carbon nanotubes generated by functionalization with polyamidoamine dendrimers. Colloids Surf B Biointerfaces 2010;80(1):18-25 View Article PubMed/NCBI
  83. Shin US, Yoon IK, Lee GS, Jang WC, Knowles JC, Kim HW. Carbon nanotubes in nanocomposites and hybrids with hydroxyapatite for bone replacements. J Tissue Eng 2011;2011:674287 View Article PubMed/NCBI
  84. Arsawang U, Saengsawang O, Rungrotmongkol T, Sornmee P, Wittayanarakul K, Remsungnen T, et al. How do carbon nanotubes serve as carriers for gemcitabine transport in a drug delivery system?. J Mol Graph Model 2011;29(5):591-596 View Article PubMed/NCBI
  85. Di Crescenzo A, Velluto D, Hubbell JA, Fontana A. Biocompatible dispersions of carbon nanotubes: a potential tool for intracellular transport of anticancer drugs. Nanoscale 2011;3(3):925-928 View Article PubMed/NCBI
  86. Chen Z, Pierre D, He H, Tan S, Pham-Huy C, Hong H, et al. Adsorption behavior of epirubicin hydrochloride on carboxylated carbon nanotubes. Int J Pharm 2011;405(1-2):153-161 View Article PubMed/NCBI
  87. Zhang D, Pan B, Wu M, Wang B, Zhang H, Peng H, et al. Adsorption of sulfamethoxazole on functionalized carbon nanotubes as affected by cations and anions. Environ Pollut 2011;159(10):2616-2621 View Article PubMed/NCBI
  88. Mohan H, Fagan A, Giordani S. Carbon Nanomaterials (CNMs) in Cancer Therapy: A Database of CNM-Based Nanocarrier Systems. Pharmaceutics 2023;15(5):1545 View Article PubMed/NCBI
  89. Wang C, Bao C, Liang S, Fu H, Wang K, Deng M, et al. RGD-conjugated silica-coated gold nanorods on the surface of carbon nanotubes for targeted photoacoustic imaging of gastric cancer. Nanoscale Res Lett 2014;9(1):264 View Article PubMed/NCBI
  90. Usman MS, Hussein MZ, Fakurazi S, Masarudin MJ, Ahmad Saad FF. A bimodal theranostic nanodelivery system based on [graphene oxide-chlorogenic acid-gadolinium/gold] nanoparticles. PLoS One 2018;13(7):e0200760 View Article PubMed/NCBI
  91. Wójcik B, Sawosz E, Szczepaniak J, Strojny B, Sosnowska M, Daniluk K, et al. Effects of Metallic and Carbon-Based Nanomaterials on Human Pancreatic Cancer Cell Lines AsPC-1 and BxPC-3. Int J Mol Sci 2021;22(22):12100 View Article PubMed/NCBI
  92. Chattaraj A, Mishra V, Mishra Y. Carbon Nanotubes in the Diagnosis and Treatment of Ovarian Cancer. Indian J Microbiol 2025;65(1):538-553 View Article PubMed/NCBI
  93. Tang L, Li J, Pan T, Yin Y, Mei Y, Xiao Q, et al. Versatile carbon nanoplatforms for cancer treatment and diagnosis: strategies, applications and future perspectives. Theranostics 2022;12(5):2290-2321 View Article PubMed/NCBI
  94. Amiri S, Shokrollahi H. The role of cobalt ferrite magnetic nanoparticles in medical science. Mater Sci Eng C Mater Biol Appl 2013;33(1):1-8 View Article PubMed/NCBI
  95. Sakashita M, Nangaku M. Ferumoxytol: an emerging therapeutic for iron deficiency anemia. Expert Opin Pharmacother 2023;24(2):171-175 View Article PubMed/NCBI
  96. Chomoucka J, Drbohlavova J, Huska D, Adam V, Kizek R, Hubalek J. Magnetic nanoparticles and targeted drug delivering. Pharmacol Res 2010;62(2):144-149 View Article PubMed/NCBI
  97. Pan BF, Gao F, Gu HC. Dendrimer modified magnetite nanoparticles for protein immobilization. J Colloid Interface Sci 2005;284(1):1-6 View Article PubMed/NCBI
  98. Chang JH, Kang KH, Choi J, Jeong YK. High efficiency protein separation with organosilane assembled silica coated magnetic nanoparticles. Superlattice Microst 2008;44(4-5):442-448 View Article
  99. Saez A, Guzmán M, Molpeceres J, Aberturas MR. Freeze-drying of polycaprolactone and poly(D,L-lactic-glycolic) nanoparticles induce minor particle size changes affecting the oral pharmacokinetics of loaded drugs. Eur J Pharm Biopharm 2000;50(3):379-387 View Article PubMed/NCBI
  100. Fishbein I, Chorny M, Banai S, Levitzki A, Danenberg HD, Gao J, et al. Formulation and delivery mode affect disposition and activity of tyrphostin-loaded nanoparticles in the rat carotid model. Arterioscler Thromb Vasc Biol 2001;21(9):1434-1439 View Article PubMed/NCBI
  101. Shim J, Seok Kang H, Park WS, Han SH, Kim J, Chang IS. Transdermal delivery of mixnoxidil with block copolymer nanoparticles. J Control Release 2004;97(3):477-484 View Article PubMed/NCBI
  102. Zhang L, Hu Y, Jiang X, Yang C, Lu W, Yang YH. Camptothecin derivative-loaded poly(caprolactone-co-lactide)-b-PEG-b-poly(caprolactone-co-lactide) nanoparticles and their biodistribution in mice. J Control Release 2004;96(1):135-148 View Article PubMed/NCBI
  103. Fang C, Shi B, Pei YY, Hong MH, Wu J, Chen HZ. In vivo tumor targeting of tumor necrosis factor-alpha-loaded stealth nanoparticles: effect of MePEG molecular weight and particle size. Eur J Pharm Sci 2006;27(1):27-36 View Article PubMed/NCBI
  104. Senior J, Gregoriadis G. Is half-life of circulating liposomes determined by changes in their permeability?. FEBS Lett 1982;145(1):109-114 View Article PubMed/NCBI
  105. Senior J, Delgado C, Fisher D, Tilcock C, Gregoriadis G. Influence of surface hydrophilicity of liposomes on their interaction with plasma protein and clearance from the circulation: studies with poly(ethylene glycol)-coated vesicles. Biochim Biophys Acta 1991;1062(1):77-82 View Article PubMed/NCBI
  106. Mohanraj VJ, Chen Y. Nanoparticles - A review. Trop J Pharm Res 2006;5(1):561-573 View Article
  107. Na K, Lee KH, Lee DH, Bae YH. Biodegradable thermo-sensitive nanoparticles from poly(L-lactic acid)/poly(ethylene glycol) alternating multi-block copolymer for potential anti-cancer drug carrier. Eur J Pharm Sci 2006;27(2-3):115-122 View Article PubMed/NCBI
  108. De Jong WH, Borm PJ. Drug delivery and nanoparticles:applications and hazards. Int J Nanomedicine 2008;3(2):133-149 View Article PubMed/NCBI
  109. Patra JK, Das G, Fraceto LF, Campos EVR, Rodriguez-Torres MDP, Acosta-Torres LS, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnology 2018;16(1):71 View Article PubMed/NCBI
  110. Chakraborty C, Pal S, Doss GP, Wen ZH, Lin CS. Nanoparticles as ‘smart’ pharmaceutical delivery. Front Biosci (Landmark Ed) 2013;18(3):1030-1050 View Article PubMed/NCBI
  111. Desai N, Rana D, Salave S, Benival D, Khunt D, Prajapati BG. Achieving Endo/Lysosomal Escape Using Smart Nanosystems for Efficient Cellular Delivery. Molecules 2024;29(13):3131 View Article PubMed/NCBI
  112. Crommelin DJ, Storm G. Liposomes: from the bench to the bed. J Liposome Res 2003;13(1):33-36 View Article PubMed/NCBI
  113. Metselaar JM, Storm G. Liposomes in the treatment of inflammatory disorders. Expert Opin Drug Deliv 2005;2(3):465-476 View Article PubMed/NCBI
  114. Minko T, Pakunlu RI, Wang Y, Khandare JJ, Saad M. New generation of liposomal drugs for cancer. Anticancer Agents Med Chem 2006;6(6):537-552 View Article PubMed/NCBI
  115. Gibaud S, Demoy M, Andreux JP, Weingarten C, Gouritin B, Couvreur P. Cells involved in the capture of nanoparticles in hematopoietic organs. J Pharm Sci 1996;85(9):944-950 View Article PubMed/NCBI
  116. Demoy M, Gibaud S, Andreux JP, Weingarten C, Gouritin B, Couvreur P. Splenic trapping of nanoparticles: complementary approaches for in situ studies. Pharm Res 1997;14(4):463-468 View Article PubMed/NCBI
  117. Lotfipour F, Shahi S, Farjami A, Salatin S, Mahmoudian M, Dizaj SM. Safety and Toxicity Issues of Therapeutically Used Nanoparticles from the Oral Route. Biomed Res Int 2021;2021:9322282 View Article PubMed/NCBI
  118. Gao Y, Huang Y, Zhang Q, Yang H, Li Y, Li Y, et al. Liposomal mitoxantrone monotherapy in patients with relapsed or refractory mature T-cell and natural killer-cell neoplasms: A phase 2, multicenter, open-label, single-arm trial. Cancer 2025;131(1):e35672 View Article PubMed/NCBI
  119. Passero FC, Grapsa D, Syrigos KN, Saif MW. The safety and efficacy of Onivyde (irinotecan liposome injection) for the treatment of metastatic pancreatic cancer following gemcitabine-based therapy. Expert Rev Anticancer Ther 2016;16(7):697-703 View Article PubMed/NCBI
  120. Gan J, Juang V, Wang K, Xia Z, Ackermann R, Yu M, et al. Reverse engineering of Onivyde® - Irinotecan liposome injection. Int J Pharm 2025;669:125000 View Article PubMed/NCBI
  121. Jain S, Vahdat LT. Eribulin mesylate. Clin Cancer Res 2011;17(21):6615-6622 View Article PubMed/NCBI
  122. Harrison TS, Lyseng-Williamson KA. Vincristine sulfate liposome injection: a guide to its use in refractory or relapsed acute lymphoblastic leukemia. BioDrugs 2013;27(1):69-74 View Article PubMed/NCBI
  123. Anderson PM, Tomaras M, McConnell K. Mifamurtide in osteosarcoma—a practical review. Drugs Today (Barc) 2010;46(5):327-337 View Article PubMed/NCBI
  124. Sofias AM, Dunne M, Storm G, Allen C. The battle of “nano” paclitaxel. Adv Drug Deliv Rev 2017;122:20-30 View Article PubMed/NCBI
  125. Balazsovits JA, Mayer LD, Bally MB, Cullis PR, McDonell M, Ginsberg RS, et al. Analysis of the effect of liposome encapsulation on the vesicant properties, acute and cardiac toxicities, and antitumor efficacy of doxorubicin. Cancer Chemother Pharmacol 1989;23(2):81-86 View Article PubMed/NCBI
  126. Kanter PM, Bullard GA, Pilkiewicz FG, Mayer LD, Cullis PR, Pavelic ZP. Preclinical toxicology study of liposome encapsulated doxorubicin (TLC D-99): comparison with doxorubicin and empty liposomes in mice and dogs. In Vivo 1993;7(1):85-95 PubMed/NCBI
  127. Pillai G, Ceballos-Coronel ML. Science and technology of the emerging nanomedicines in cancer therapy: A primer for physicians and pharmacists. SAGE Open Med 2013;1:2050312113513759 View Article PubMed/NCBI
  128. Galmarini CM, Thomas X, Calvo F, Rousselot P, Rabilloud M, El Jaffari A, et al. In vivo mechanisms of resistance to cytarabine in acute myeloid leukaemia. Br J Haematol 2002;117(4):860-868 View Article PubMed/NCBI
  129. Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R. FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist 2007;12(10):1247-1252 View Article PubMed/NCBI
  130. Manoukian G, Hagemeister F. Denileukin diftitox: a novel immunotoxin. Expert Opin Biol Ther 2009;9(11):1445-1451 View Article PubMed/NCBI
  131. Gill PS, Wernz J, Scadden DT, Cohen P, Mukwaya GM, von Roenn JH, et al. Randomized phase III trial of liposomal daunorubicin versus doxorubicin, bleomycin, and vincristine in AIDS-related Kaposi’s sarcoma. J Clin Oncol 1996;14(8):2353-2364 View Article PubMed/NCBI
  132. Lasic DD. Doxorubicin in sterically stabilized liposomes. Nature 1996;380(6574):561-562 View Article PubMed/NCBI
  133. Andreopoulou E, Gaiotti D, Kim E, Downey A, Mirchandani D, Hamilton A, et al. Pegylated liposomal doxorubicin HCL (PLD; Caelyx/Doxil): experience with long-term maintenance in responding patients with recurrent epithelial ovarian cancer. Ann Oncol 2007;18(4):716-721 View Article PubMed/NCBI
  134. Krauss AC, Gao X, Li L, Manning ML, Patel P, Fu W, et al. FDA Approval Summary: (Daunorubicin and Cytarabine) Liposome for Injection for the Treatment of Adults with High-Risk Acute Myeloid Leukemia. Clin Cancer Res 2019;25(9):2685-2690 View Article PubMed/NCBI
  135. Allen C. Why I’m Holding onto Hope for Nano in Oncology. Mol Pharm 2016;13(8):2603-2604 View Article PubMed/NCBI
  136. Harris JM, Chess RB. Effect of pegylation on pharmaceuticals. Nat Rev Drug Discov 2003;2(3):214-221 View Article PubMed/NCBI
  137. Dinndorf PA, Gootenberg J, Cohen MH, Keegan P, Pazdur R. FDA drug approval summary: pegaspargase (oncaspar) for the first-line treatment of children with acute lymphoblastic leukemia (ALL). Oncologist 2007;12(8):991-998 View Article PubMed/NCBI
  138. Miele E, Spinelli GP, Miele E, Tomao F, Tomao S. Albumin-bound formulation of paclitaxel (Abraxane ABI-007) in the treatment of breast cancer. Int J Nanomedicine 2009;4:99-105 View Article PubMed/NCBI
  139. Berges R. Eligard®: Pharmacokinetics, Effect on Testosterone and PSA Levels and Tolerability. Eur Urol Suppl 2005;4(5):20-25 View Article
  140. Pardridge WM. Blood-brain barrier delivery. Drug Discov Today 2007;12(1-2):54-61 View Article PubMed/NCBI
  141. Olivier JC, Fenart L, Chauvet R, Pariat C, Cecchelli R, Couet W. Indirect evidence that drug brain targeting using polysorbate 80-coated polybutylcyanoacrylate nanoparticles is related to toxicity. Pharm Res 1999;16(12):1836-1842 View Article PubMed/NCBI
  142. Kreuter J. Influence of the surface properties on nanoparticle-mediated transport of drugs to the brain. J Nanosci Nanotechnol 2004;4(5):484-488 View Article PubMed/NCBI
  143. Lockman PR, Koziara JM, Mumper RJ, Allen DD. Nanoparticle surface charges alter blood-brain barrier integrity and permeability. J Drug Target 2004;12(9-10):635-641 View Article PubMed/NCBI
  144. Koziara JM, Lockman PR, Allen DD, Mumper RJ. The blood-brain barrier and brain drug delivery. J Nanosci Nanotechnol 2006;6(9-10):2712-2735 View Article PubMed/NCBI
  145. Sánchez-Cano F, Hernández-Kelly LC, Ortega A. Silica Nanoparticles Decrease Glutamate Uptake in Blood-Brain Barrier Components. Neurotox Res 2024;42(2):20 View Article PubMed/NCBI
  146. Michaelis K, Hoffmann MM, Dreis S, Herbert E, Alyautdin RN, Michaelis M, et al. Covalent linkage of apolipoprotein e to albumin nanoparticles strongly enhances drug transport into the brain. J Pharmacol Exp Ther 2006;317(3):1246-1253 View Article PubMed/NCBI
  147. Girardin F. Membrane transporter proteins: a challenge for CNS drug development. Dialogues Clin Neurosci 2006;8(3):311-321 View Article PubMed/NCBI
  148. Mahringer A, Ott M, Reimold I, Reichel V, Fricker G. The ABC of the blood-brain barrier - regulation of drug efflux pumps. Curr Pharm Des 2011;17(26):2762-2770 View Article PubMed/NCBI
  149. Koziara JM, Lockman PR, Allen DD, Mumper RJ. Paclitaxel nanoparticles for the potential treatment of brain tumors. J Control Release 2004;99(2):259-269 View Article PubMed/NCBI
  150. Oberdörster E. Manufactured nanomaterials (fullerenes, C60) induce oxidative stress in the brain of juvenile largemouth bass. Environ Health Perspect 2004;112(10):1058-1062 View Article PubMed/NCBI
  151. Elder A, Gelein R, Silva V, Feikert T, Opanashuk L, Carter J, et al. Translocation of inhaled ultrafine manganese oxide particles to the central nervous system. Environ Health Perspect 2006;114(8):1172-1178 View Article PubMed/NCBI
  152. Formica ML, Real DA, Picchio ML, Catlin E, Donnelly RF, Paredes AJ. On a highway to the brain: A review on nose-to-brain drug delivery using nanoparticles. Appl Mater Today 2022;29:101631 View Article
  153. Seegenschmiedt MH, Vernon CC. A Historical Perspective on Hyperthermia in Oncology. In: Seegenschmiedt MH, Fessenden P, Vernon CC (eds). Thermoradiotherapy and Thermochemotherapy: Biology, Physiology, Physics. Berlin, Heidelberg: Springer Berlin Heidelberg; 1995:3-44 View Article
  154. Hübner J, Käsmann L, Liebl CM, Dörfler J, Kutschan S. A Review of the Effect of Hyperthermia in the Treatment of Various Types of Cancer. Clin Cancer Investig J 2024;13(1):40-45 View Article
  155. Ahlers O, Boehnke T, Kerner T, Deja M, Keh D, Löffel J, et al. Induced hyperthermia causes significant changes in lymphocytes. Crit Care 1998;2(Suppl 1):P002 View Article
  156. Kozłowski HM, Sobocińska J, Jędrzejewski T, Maciejewski B, Dzialuk A, Wrotek S. Fever-Range Hyperthermia Promotes Macrophage Polarization towards Regulatory Phenotype M2b. Int J Mol Sci 2023;24(24):17574 View Article PubMed/NCBI
  157. Liebl CM, Kutschan S, Dörfler J, Käsmann L, Hübner J. Systematic review about complementary medical hyperthermia in oncology. Clin Exp Med 2022;22(4):519-565 View Article PubMed/NCBI
  158. Szwed M, Marczak A. Application of Nanoparticles for Magnetic Hyperthermia for Cancer Treatment-The Current State of Knowledge. Cancers (Basel) 2024;16(6):1156 View Article PubMed/NCBI
  159. Wu J. The Enhanced Permeability and Retention (EPR) Effect: The Significance of the Concept and Methods to Enhance Its Application. J Pers Med 2021;11(8):771 View Article PubMed/NCBI
  160. Salunkhe AB, Khot VM, Pawar SH. Magnetic hyperthermia with magnetic nanoparticles: a status review. Curr Top Med Chem 2014;14(5):572-594 View Article PubMed/NCBI
  161. Verçoza BR, Bernardo RR, Pentón-Madrigal A, Sinnecker JP, Rodrigues JC, S de Oliveira LA. Therapeutic potential of low-cost nanocarriers produced by green synthesis: macrophage uptake of superparamagnetic iron oxide nanoparticles. Nanomedicine (Lond) 2019;14(17):2293-2313 View Article PubMed/NCBI
  162. Phung DC, Nguyen HT, Phuong Tran TT, Jin SG, Yong CS, Truong DH, et al. Combined hyperthermia and chemotherapy as a synergistic anticancer treatment. J Pharm Investig 2019;49(5):519-526 View Article
  163. Horsman MR, Overgaard J. Hyperthermia: a potent enhancer of radiotherapy. Clin Oncol (R Coll Radiol) 2007;19(6):418-426 View Article PubMed/NCBI
  164. Issels RD. Hyperthermia adds to chemotherapy. Eur J Cancer 2008;44(17):2546-2554 View Article PubMed/NCBI
  165. O’Neill KL, Fairbairn DW, Smith MJ, Poe BS. Critical parameters influencing hyperthermia-induced apoptosis in human lymphoid cell lines. Apoptosis 1998;3(5):369-375 View Article PubMed/NCBI
  166. Calderwood SK, Ciocca DR. Heat shock proteins: stress proteins with Janus-like properties in cancer. Int J Hyperthermia 2008;24(1):31-39 View Article PubMed/NCBI
  167. Peer AJ, Grimm MJ, Zynda ER, Repasky EA. Diverse immune mechanisms may contribute to the survival benefit seen in cancer patients receiving hyperthermia. Immunol Res 2010;46(1-3):137-154 View Article PubMed/NCBI
  168. Rossi A, Ciafrè S, Balsamo M, Pierimarchi P, Santoro MG. Targeting the heat shock factor 1 by RNA interference: a potent tool to enhance hyperthermochemotherapy efficacy in cervical cancer. Cancer Res 2006;66(15):7678-7685 View Article PubMed/NCBI
  169. Skitzki JJ, Repasky EA, Evans SS. Hyperthermia as an immunotherapy strategy for cancer. Curr Opin Investig Drugs 2009;10(6):550-558 PubMed/NCBI
  170. Biswas AK, Mitchell DL, Johnson DG. E2F1 responds to ultraviolet radiation by directly stimulating DNA repair and suppressing carcinogenesis. Cancer Res 2014;74(12):3369-3377 View Article PubMed/NCBI
  171. Warters RL, Henle KJ. DNA degradation in chinese hamster ovary cells after exposure to hyperthermia. Cancer Res 1982;42(11):4427-4432 PubMed/NCBI
  172. Jorritsma JB, Burgman P, Kampinga HH, Konings AW. DNA polymerase activity in heat killing and hyperthermic radiosensitization of mammalian cells as observed after fractionated heat treatments. Radiat Res 1986;105(3):307-319 PubMed/NCBI
  173. Paull TT, Rogakou EP, Yamazaki V, Kirchgessner CU, Gellert M, Bonner WM. A critical role for histone H2AX in recruitment of repair factors to nuclear foci after DNA damage. Curr Biol 2000;10(15):886-895 View Article PubMed/NCBI
  174. Takahashi A, Matsumoto H, Nagayama K, Kitano M, Hirose S, Tanaka H, et al. Evidence for the involvement of double-strand breaks in heat-induced cell killing. Cancer Res 2004;64(24):8839-8845 View Article PubMed/NCBI
  175. Wyman C, Kanaar R. DNA double-strand break repair: all’s well that ends well. Annu Rev Genet 2006;40:363-383 View Article PubMed/NCBI
  176. Robertson AB, Klungland A, Rognes T, Leiros I. DNA repair in mammalian cells: Base excision repair: the long and short of it. Cell Mol Life Sci 2009;66(6):981-993 View Article PubMed/NCBI
  177. Gani C, Lamprecht U, Ziegler A, Moll M, Gellermann J, Heinrich V, et al. Deep regional hyperthermia with preoperative radiochemotherapy in locally advanced rectal cancer, a prospective phase II trial. Radiother Oncol 2021;159:155-160 View Article PubMed/NCBI
  178. Franckena M, Lutgens LC, Koper PC, Kleynen CE, van der Steen-Banasik EM, Jobsen JJ, et al. Radiotherapy and hyperthermia for treatment of primary locally advanced cervix cancer: results in 378 patients. Int J Radiat Oncol Biol Phys 2009;73(1):242-250 View Article PubMed/NCBI
  179. Van Vulpen M, De Leeuw AA, Raaymakers BW, Van Moorselaar RJ, Hofman P, Lagendijk JJ, et al. Radiotherapy and hyperthermia in the treatment of patients with locally advanced prostate cancer: preliminary results. BJU Int 2004;93(1):36-41 View Article PubMed/NCBI
  180. van der Zee J, González González D, van Rhoon GC, van Dijk JD, van Putten WL, Hart AA. Comparison of radiotherapy alone with radiotherapy plus hyperthermia in locally advanced pelvic tumours: a prospective, randomised, multicentre trial. Dutch Deep Hyperthermia Group. Lancet 2000;355(9210):1119-1125 View Article PubMed/NCBI
  181. Algan O, Fosmire H, Hynynen K, Dalkin B, Cui H, Drach G, et al. External beam radiotherapy and hyperthermia in the treatment of patients with locally advanced prostate carcinoma. Cancer 2000;89(2):399-403 View Article PubMed/NCBI
  182. Zschaeck S, Weingärtner J, Ghadjar P, Wust P, Mehrhof F, Kalinauskaite G, et al. Fever range whole body hyperthermia for re-irradiation of head and neck squamous cell carcinomas: Final results of a prospective study. Oral Oncol 2021;116:105240 View Article PubMed/NCBI
  183. Rau B, Wust P, Hohenberger P, Löffel J, Hünerbein M, Below C, et al. Preoperative hyperthermia combined with radiochemotherapy in locally advanced rectal cancer: a phase II clinical trial. Ann Surg 1998;227(3):380-389 View Article PubMed/NCBI
  184. Hurwitz MD, Kaplan ID, Hansen JL, Prokopios-Davos S, Topulos GP, Wishnow K, et al. Association of rectal toxicity with thermal dose parameters in treatment of locally advanced prostate cancer with radiation and hyperthermia. Int J Radiat Oncol Biol Phys 2002;53(4):913-918 View Article PubMed/NCBI
  185. Arends TJ, Nativ O, Maffezzini M, de Cobelli O, Canepa G, Verweij F, et al. Results of a Randomised Controlled Trial Comparing Intravesical Chemohyperthermia with Mitomycin C Versus Bacillus Calmette-Guérin for Adjuvant Treatment of Patients with Intermediate- and High-risk Non-Muscle-invasive Bladder Cancer. Eur Urol 2016;69(6):1046-1052 View Article PubMed/NCBI
  186. Kleef R, Moss R, Szasz AM, Bohdjalian A, Bojar H, Bakacs T. Complete Clinical Remission of Stage IV Triple-Negative Breast Cancer Lung Metastasis Administering Low-Dose Immune Checkpoint Blockade in Combination With Hyperthermia and Interleukin-2. Integr Cancer Ther 2018;17(4):1297-1303 View Article PubMed/NCBI
  187. Kleef R, Nagy R, Baierl A, Bacher V, Bojar H, McKee DL, et al. Low-dose ipilimumab plus nivolumab combined with IL-2 and hyperthermia in cancer patients with advanced disease: exploratory findings of a case series of 131 stage IV cancers - a retrospective study of a single institution. Cancer Immunol Immunother 2021;70(5):1393-1403 View Article PubMed/NCBI
  188. Issels RD, Lindner LH, Verweij J, Wessalowski R, Reichardt P, Wust P, et al. Effect of Neoadjuvant Chemotherapy Plus Regional Hyperthermia on Long-term Outcomes Among Patients With Localized High-Risk Soft Tissue Sarcoma: The EORTC 62961-ESHO 95 Randomized Clinical Trial. JAMA Oncol 2018;4(4):483-492 View Article PubMed/NCBI
  189. Quénet F, Elias D, Roca L, Goéré D, Ghouti L, Pocard M, et al. Cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy versus cytoreductive surgery alone for colorectal peritoneal metastases (PRODIGE 7): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2021;22(2):256-266 View Article PubMed/NCBI
  190. van Driel WJ, Koole SN, Sikorska K, Schagen van Leeuwen JH, Schreuder HWR, Hermans RHM, et al. Hyperthermic Intraperitoneal Chemotherapy in Ovarian Cancer. N Engl J Med 2018;378(3):230-240 View Article PubMed/NCBI
  191. Thiesen B, Jordan A. Clinical applications of magnetic nanoparticles for hyperthermia. Int J Hyperthermia 2008;24(6):467-474 View Article PubMed/NCBI
  192. Johannsen M, Gneveckow U, Eckelt L, Feussner A, Waldöfner N, Scholz R, et al. Clinical hyperthermia of prostate cancer using magnetic nanoparticles: presentation of a new interstitial technique. Int J Hyperthermia 2005;21(7):637-647 View Article PubMed/NCBI
  193. Colson P, La Scola B, Levasseur A, Caetano-Anollés G, Raoult D. Mimivirus: leading the way in the discovery of giant viruses of amoebae. Nat Rev Microbiol 2017;15(4):243-254 View Article PubMed/NCBI
  194. Ryu WS. Molecular Virology of Human Pathogenic Viruses. Boston: Academic Press; 2017, 31-45 View Article
  195. DeLong JP, Al-Sammak MA, Al-Ameeli ZT, Dunigan DD, Edwards KF, Fuhrmann JJ, et al. Towards an integrative view of virus phenotypes. Nat Rev Microbiol 2022;20(2):83-94 View Article PubMed/NCBI
  196. Uchiyama J, Rashel M, Maeda Y, Takemura I, Sugihara S, Akechi K, et al. Isolation and characterization of a novel Enterococcus faecalis bacteriophage phiEF24C as a therapeutic candidate. FEMS Microbiol Lett 2008;278(2):200-206 View Article PubMed/NCBI
  197. Šivec K, Podgornik A. Determination of bacteriophage growth parameters under cultivating conditions. Appl Microbiol Biotechnol 2020;104(20):8949-8960 View Article PubMed/NCBI
  198. Chen HY, Di Mascio M, Perelson AS, Ho DD, Zhang L. Determination of virus burst size in vivo using a single-cycle SIV in rhesus macaques. Proc Natl Acad Sci U S A 2007;104(48):19079-19084 View Article PubMed/NCBI
  199. Bird SW, Kirkegaard K. Escape of non-enveloped virus from intact cells. Virology 2015;479-480:444-449 View Article PubMed/NCBI
  200. Liu Z, Qiao J, Niu Z, Wang Q. Natural supramolecular building blocks: from virus coat proteins to viral nanoparticles. Chem Soc Rev 2012;41(18):6178-6194 View Article PubMed/NCBI
  201. Buzón P, Maity S, Christodoulis P, Wiertsema MJ, Dunkelbarger S, Kim C, et al. Virus self-assembly proceeds through contact-rich energy minima. Sci Adv 2021;7(45):eabg0811 View Article PubMed/NCBI
  202. Sun S, Rao VB, Rossmann MG. Genome packaging in viruses. Curr Opin Struct Biol 2010;20(1):114-120 View Article PubMed/NCBI
  203. Buzón P, Maity S, Roos WH. Physical virology: From virus self-assembly to particle mechanics. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2020;12(4):e1613 View Article PubMed/NCBI
  204. Kiss B, Mudra D, Török G, Mártonfalvi Z, Csík G, Herényi L, et al. Single-particle virology. Biophys Rev 2020;12(5):1141-1154 View Article PubMed/NCBI
  205. Bayer ME, Blumberg BS, Werner B. Particles associated with Australia antigen in the sera of patients with leukaemia, Down’s Syndrome and hepatitis. Nature 1968;218(5146):1057-1059 View Article PubMed/NCBI
  206. Chung YH, Cai H, Steinmetz NF. Viral nanoparticles for drug delivery, imaging, immunotherapy, and theranostic applications. Adv Drug Deliv Rev 2020;156:214-235 View Article PubMed/NCBI
  207. Wu Y, Li J, Shin H-J. Self-assembled Viral Nanoparticles as Targeted Anticancer Vehicles. Biotechnol Bioprocess Eng 2021;26(1):25-38 View Article PubMed/NCBI
  208. Plummer EM, Manchester M. Viral nanoparticles and virus-like particles: platforms for contemporary vaccine design. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2011;3(2):174-196 View Article PubMed/NCBI
  209. Steinmetz NF. Viral nanoparticles as platforms for next-generation therapeutics and imaging devices. Nanomedicine 2010;6(5):634-641 View Article PubMed/NCBI
  210. Watanabe M, Nishikawaji Y, Kawakami H, Kosai KI. Adenovirus Biology, Recombinant Adenovirus, and Adenovirus Usage in Gene Therapy. Viruses 2021;13(12):2502 View Article PubMed/NCBI
  211. Shaw AR, Suzuki M. Immunology of Adenoviral Vectors in Cancer Therapy. Mol Ther Methods Clin Dev 2019;15:418-429 View Article PubMed/NCBI
  212. Wold WS, Toth K. Adenovirus vectors for gene therapy, vaccination and cancer gene therapy. Curr Gene Ther 2013;13(6):421-433 View Article PubMed/NCBI
  213. Venkataraman S, Apka P, Shoeb E, Badar U, Hefferon K. Plant Virus Nanoparticles for Anti-cancer Therapy. Front Bioeng Biotechnol 2021;9:642794 View Article PubMed/NCBI
  214. Kwon SJ, Park MR, Kim KW, Plante CA, Hemenway CL, Kim KH. cis-Acting sequences required for coat protein binding and in vitro assembly of Potato virus X. Virology 2005;334(1):83-97 View Article PubMed/NCBI
  215. Tanaka T, Huang J, Hirai S, Kuroki M, Kuroki M, Watanabe N, et al. Carcinoembryonic antigen-targeted selective gene therapy for gastric cancer through FZ33 fiber-modified adenovirus vectors. Clin Cancer Res 2006;12(12):3803-3813 View Article PubMed/NCBI
  216. Brunel FM, Lewis JD, Destito G, Steinmetz NF, Manchester M, Stuhlmann H, et al. Hydrazone ligation strategy to assemble multifunctional viral nanoparticles for cell imaging and tumor targeting. Nano Lett 2010;10(3):1093-1097 View Article PubMed/NCBI
  217. Chatterji A, Ochoa W, Shamieh L, Salakian SP, Wong SM, Clinton G, et al. Chemical conjugation of heterologous proteins on the surface of Cowpea mosaic virus. Bioconjug Chem 2004;15(4):807-813 View Article PubMed/NCBI
  218. Leong HS, Steinmetz NF, Ablack A, Destito G, Zijlstra A, Stuhlmann H, et al. Intravital imaging of embryonic and tumor neovasculature using viral nanoparticles. Nat Protoc 2010;5(8):1406-1417 View Article PubMed/NCBI
  219. Kim HJ, Kim HJ. Yeast as an expression system for producing virus-like particles: what factors do we need to consider?. Lett Appl Microbiol 2017;64(2):111-123 View Article PubMed/NCBI
  220. Lünsdorf H, Gurramkonda C, Adnan A, Khanna N, Rinas U. Virus-like particle production with yeast: ultrastructural and immunocytochemical insights into Pichia pastoris producing high levels of the hepatitis B surface antigen. Microb Cell Fact 2011;10:48 View Article PubMed/NCBI
  221. Sakai C, Hosokawa K, Watanabe T, Suzuki Y, Nakano T, Ueda K, et al. Human hepatitis B virus-derived virus-like particle as a drug and DNA delivery carrier. Biochem Biophys Res Commun 2021;581:103-109 View Article PubMed/NCBI
  222. Firdaus MER, Mustopa AZ, Ekawati N, Chairunnisa S, Arifah RK, Hertati A, et al. Optimization, characterization, comparison of self-assembly VLP of capsid protein L1 in yeast and reverse vaccinology design against human papillomavirus type 52. J Genet Eng Biotechnol 2023;21(1):68 View Article PubMed/NCBI
  223. Srivastava V, Nand KN, Ahmad A, Kumar R. Yeast-Based Virus-like Particles as an Emerging Platform for Vaccine Development and Delivery. Vaccines (Basel) 2023;11(2):479 View Article PubMed/NCBI
  224. Tornesello AL, Tagliamonte M, Buonaguro FM, Tornesello ML, Buonaguro L. Virus-like Particles as Preventive and Therapeutic Cancer Vaccines. Vaccines (Basel) 2022;10(2):227 View Article PubMed/NCBI
  225. Sominskaya I, Skrastina D, Dislers A, Vasiljev D, Mihailova M, Ose V, et al. Construction and immunological evaluation of multivalent hepatitis B virus (HBV) core virus-like particles carrying HBV and HCV epitopes. Clin Vaccine Immunol 2010;17(6):1027-1033 View Article PubMed/NCBI
  226. Kheirvari M, Liu H, Tumban E. Virus-like Particle Vaccines and Platforms for Vaccine Development. Viruses 2023;15(5):1109 View Article PubMed/NCBI
  227. Cheng L, Wang Y, Du J. Human Papillomavirus Vaccines: An Updated Review. Vaccines (Basel) 2020;8(3):391 View Article PubMed/NCBI
  228. Palladini A, Thrane S, Janitzek CM, Pihl J, Clemmensen SB, de Jongh WA, et al. Virus-like particle display of HER2 induces potent anti-cancer responses. Oncoimmunology 2018;7(3):e1408749 View Article PubMed/NCBI
  229. Zhang S, Yong LK, Li D, Cubas R, Chen C, Yao Q. Mesothelin virus-like particle immunization controls pancreatic cancer growth through CD8+ T cell induction and reduction in the frequency of CD4+ foxp3+ ICOS- regulatory T cells. PLoS One 2013;8(7):e68303 View Article PubMed/NCBI
  230. Speiser DE, Schwarz K, Baumgaertner P, Manolova V, Devevre E, Sterry W, et al. Memory and effector CD8 T-cell responses after nanoparticle vaccination of melanoma patients. J Immunother 2010;33(8):848-858 View Article PubMed/NCBI
  231. Kerstetter-Fogle A, Shukla S, Wang C, Beiss V, Harris PLR, Sloan AE, et al. Plant Virus-Like Particle In Situ Vaccine for Intracranial Glioma Immunotherapy. Cancers (Basel) 2019;11(4):515 View Article PubMed/NCBI
  232. Xie N, Shen G, Gao W, Huang Z, Huang C, Fu L. Neoantigens: promising targets for cancer therapy. Signal Transduct Target Ther 2023;8(1):9 View Article PubMed/NCBI
  233. Finbloom JA, Aanei IL, Bernard JM, Klass SH, Elledge SK, Han K, et al. Evaluation of Three Morphologically Distinct Virus-Like Particles as Nanocarriers for Convection-Enhanced Drug Delivery to Glioblastoma. Nanomaterials (Basel) 2018;8(12):1007 View Article PubMed/NCBI
  234. Le DH, Lee KL, Shukla S, Commandeur U, Steinmetz NF. Potato virus X, a filamentous plant viral nanoparticle for doxorubicin delivery in cancer therapy. Nanoscale 2017;9(6):2348-2357 View Article PubMed/NCBI
  235. Kernan DL, Wen AM, Pitek AS, Steinmetz NF. Featured Article: Delivery of chemotherapeutic vcMMAE using tobacco mosaic virus nanoparticles. Exp Biol Med (Maywood) 2017;242(14):1405-1411 View Article PubMed/NCBI
  236. Shukla S, Roe AJ, Liu R, Veliz FA, Commandeur U, Wald DN, et al. Affinity of plant viral nanoparticle potato virus X (PVX) towards malignant B cells enables cancer drug delivery. Biomater Sci 2020;8(14):3935-3943 View Article PubMed/NCBI
  237. Hu H, Steinmetz NF. Cisplatin Prodrug-Loaded Nanoparticles Based on Physalis Mottle Virus for Cancer Therapy. Mol Pharm 2020;17(12):4629-4636 View Article PubMed/NCBI
  238. Hoopes PJ, Mazur CM, Osterberg B, Song A, Gladstone DJ, Steinmetz NF, et al. Effect of intra-tumoral magnetic nanoparticle hyperthermia and viral nanoparticle immunogenicity on primary and metastatic cancer. Proc SPIE Int Soc Opt Eng 2017;10066:100660G View Article PubMed/NCBI
  239. Liu JF, Jang B, Issadore D, Tsourkas A. Use of magnetic fields and nanoparticles to trigger drug release and improve tumor targeting. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2019;11(6):e1571 View Article PubMed/NCBI
  240. Fatima H, Kim K-S. Magnetic nanoparticles for bioseparation. Korean J Chem Eng 2017;34(3):589-599 View Article
  241. Shukla S, Steinmetz NF. Virus-based nanomaterials as positron emission tomography and magnetic resonance contrast agents: from technology development to translational medicine. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2015;7(5):708-721 View Article PubMed/NCBI
  242. Liu M, Zhao Y, Shi Z, Zink JI, Yu Q. Virus-like Magnetic Mesoporous Silica Particles as a Universal Vaccination Platform against Pathogenic Infections. ACS Nano 2023;17(7):6899-6911 View Article PubMed/NCBI
  243. Shao P, Wang B, Wang Y, Li J, Zhang Y. The Application of Thermosensitive Nanocarriers in Controlled Drug Delivery. J Nanomater 2011;2011(1):389640 View Article
  244. Thong QX, Biabanikhankahdani R, Ho KL, Alitheen NB, Tan WS. Thermally-responsive Virus-like Particle for Targeted Delivery of Cancer Drug. Sci Rep 2019;9(1):3945 View Article PubMed/NCBI
  245. Weller M, Wen PY, Chang SM, Dirven L, Lim M, Monje M, et al. Glioma. Nat Rev Dis Primers 2024;10(1):33 View Article PubMed/NCBI
  246. Wu D, Chen Q, Chen X, Han F, Chen Z, Wang Y. The blood-brain barrier: structure, regulation, and drug delivery. Signal Transduct Target Ther 2023;8(1):217 View Article PubMed/NCBI
  247. Ghosh A, Majie A, Karmakar V, Chatterjee K, Chakraborty S, Pandey M, et al. In-depth Mechanism, Challenges, and Opportunities of Delivering Therapeutics in Brain Using Intranasal Route. AAPS PharmSciTech 2024;25(5):96 View Article PubMed/NCBI
  248. Baek SH, Hwang EH, Hur GH, Kim G, An YJ, Park JH, et al. Intranasal administration enhances size-dependent pulmonary phagocytic uptake of poly(lactic-co-glycolic acid) nanoparticles. EJNMMI Radiopharm Chem 2024;9(1):12 View Article PubMed/NCBI
  249. Cho HJ, Choi MK, Lin H, Kim JS, Chung SJ, Shim CK, et al. Expression and functional activity of P-glycoprotein in passaged primary human nasal epithelial cell monolayers cultured by the air-liquid interface method for nasal drug transport study. J Pharm Pharmacol 2011;63(3):385-391 View Article PubMed/NCBI
  250. Gizurarson S. The effect of cilia and the mucociliary clearance on successful drug delivery. Biol Pharm Bull 2015;38(4):497-506 View Article PubMed/NCBI
  251. Safarov R, Fedotova O, Uvarova A, Gordienko M, Menshutina N. Review of Intranasal Active Pharmaceutical Ingredient Delivery Systems. Pharmaceuticals (Basel) 2024;17(9):1180 View Article PubMed/NCBI
  252. Fernandes J. Oncogenes: The Passport for Viral Oncolysis Through PKR Inhibition. Biomark Cancer 2016;8:101-110 View Article PubMed/NCBI
  253. Lin D, Shen Y, Liang T. Oncolytic virotherapy: basic principles, recent advances and future directions. Signal Transduct Target Ther 2023;8(1):156 View Article PubMed/NCBI
  254. Foreman PM, Friedman GK, Cassady KA, Markert JM. Oncolytic Virotherapy for the Treatment of Malignant Glioma. Neurotherapeutics 2017;14(2):333-344 View Article PubMed/NCBI
  255. Sun K, Shi X, Li L, Nie X, Xu L, Jia F, et al. Oncolytic Viral Therapy for Glioma by Recombinant Sindbis Virus. Cancers (Basel) 2023;15(19):4738 View Article PubMed/NCBI
  256. Ozduman K, Wollmann G, Piepmeier JM, van den Pol AN. Systemic vesicular stomatitis virus selectively destroys multifocal glioma and metastatic carcinoma in brain. J Neurosci 2008;28(8):1882-1893 View Article PubMed/NCBI
  257. Nguyen A, Ho L, Wan Y. Chemotherapy and Oncolytic Virotherapy: Advanced Tactics in the War against Cancer. Front Oncol 2014;4:145 View Article PubMed/NCBI
  258. Moreno R. Mesenchymal stem cells and oncolytic viruses: joining forces against cancer. J Immunother Cancer 2021;9(2):e001684 View Article PubMed/NCBI
  259. Danielyan L, Schäfer R, von Ameln-Mayerhofer A, Buadze M, Geisler J, Klopfer T, et al. Intranasal delivery of cells to the brain. Eur J Cell Biol 2009;88(6):315-324 View Article PubMed/NCBI
  260. Spencer D, Yu D, Morshed RA, Li G, Pituch KC, Gao DX, et al. Pharmacologic modulation of nasal epithelium augments neural stem cell targeting of glioblastoma. Theranostics 2019;9(7):2071-2083 View Article PubMed/NCBI
  261. Hashizume R, Ozawa T, Gryaznov SM, Bollen AW, Lamborn KR, Frey WH, et al. New therapeutic approach for brain tumors: Intranasal delivery of telomerase inhibitor GRN163. Neuro Oncol 2008;10(2):112-120 View Article PubMed/NCBI
  262. Banskota S, Raguram A, Suh S, Du SW, Davis JR, Choi EH, et al. Engineered virus-like particles for efficient in vivo delivery of therapeutic proteins. Cell 2022;185(2):250-265.e16 View Article PubMed/NCBI
  263. Chao CN, Yang YH, Wu MS, Chou MC, Fang CY, Lin MC, et al. Gene therapy for human glioblastoma using neurotropic JC virus-like particles as a gene delivery vector. Sci Rep 2018;8(1):2213 View Article PubMed/NCBI
  264. Bernstock JD, Blitz SE, Hoffman SE, Gerstl JVE, Chiocca EA, Friedman GK. Recent oncolytic virotherapy clinical trials outline a roadmap for the treatment of high-grade glioma. Neurooncol Adv 2023;5(1):vdad081 View Article PubMed/NCBI
  265. Calderón-Peláez MA, Maradei Anaya SJ, Bedoya-Rodríguez IJ, González-Ipuz KG, Vera-Palacios D, Buitrago IV, et al. Zika Virus: A Neurotropic Warrior against High-Grade Gliomas-Unveiling Its Potential for Oncolytic Virotherapy. Viruses 2024;16(4):561 View Article PubMed/NCBI
  266. Wang C, Fernández de Ávila BE, Mundaca-Uribe R, Lopez-Ramirez MA, Ramírez-Herrera DE, Shukla S, et al. Active Delivery of VLPs Promotes Anti-Tumor Activity in a Mouse Ovarian Tumor Model. Small 2020;16(20):e1907150 View Article PubMed/NCBI
  267. Komane MD, Kayoka-Kabongo PN, Rutkowska DA. The Use of Plant Viral Nanoparticles in Cancer Biotherapy-A Review. Viruses 2025;17(2):218 View Article PubMed/NCBI
  268. Parsamian P, Liu Y, Xie C, Chen Z, Kang P, Wijesundara YH, et al. Enhanced Nanobubble Formation: Gold Nanoparticle Conjugation to Qβ Virus-like Particles. ACS Nano 2023;17(8):7797-7805 View Article PubMed/NCBI
  269. Javidpour L, Lošdorfer Božič A, Podgornik R, Naji A. Role of metallic core for the stability of virus-like particles in strongly coupled electrostatics. Sci Rep 2019;9(1):3884 View Article PubMed/NCBI
  270. Benjamin CE, Chen Z, Kang P, Wilson BA, Li N, Nielsen SO, et al. Site-Selective Nucleation and Size Control of Gold Nanoparticle Photothermal Antennae on the Pore Structures of a Virus. J Am Chem Soc 2018;140(49):17226-17233 View Article PubMed/NCBI
  271. Nowak-Jary J, Machnicka B. Pharmacokinetics of magnetic iron oxide nanoparticles for medical applications. J Nanobiotechnology 2022;20(1):305 View Article PubMed/NCBI
  272. Enomoto T, Kawano M, Fukuda H, Sawada W, Inoue T, Haw KC, et al. Viral protein-coating of magnetic nanoparticles using simian virus 40 VP1. J Biotechnol 2013;167(1):8-15 View Article PubMed/NCBI
  273. Kim KR, Lee AS, Heo HR, Park SY, Kim CS. Bioinspired synthesis of virus-like particle-templated thin silica-layered nanocages with enhanced biocompatibility and cellular uptake as drug delivery carriers. Colloids Surf B Biointerfaces 2025;247:114418 View Article PubMed/NCBI
  274. Gupta R, Arora K, Roy SS, Joseph A, Rastogi R, Arora NM, et al. Platforms, advances, and technical challenges in virus-like particles-based vaccines. Front Immunol 2023;14:1123805 View Article PubMed/NCBI
  275. Ruzzi F, Semprini MS, Scalambra L, Palladini A, Angelicola S, Cappello C, et al. Virus-like Particle (VLP) Vaccines for Cancer Immunotherapy. Int J Mol Sci 2023;24(16):12963 View Article PubMed/NCBI
  276. Luo H, Ma Y, Su Z, Gu Y, Zhang S, Gerstweiler L. Investigating the stability of chimeric murine polyomavirus VP1 Capsomeres via molecular dynamics simulations and experimental analysis. Int J Biol Macromol 2025;286:138372 View Article PubMed/NCBI
  277. Zhang L, Tang R, Bai S, Connors NK, Lua LH, Chuan YP, et al. Energetic changes caused by antigenic module insertion in a virus-like particle revealed by experiment and molecular dynamics simulations. PLoS One 2014;9(9):e107313 View Article PubMed/NCBI
  278. Huang X, Bronstein LM, Retrum J, Dufort C, Tsvetkova I, Aniagyei S, et al. Self-Assembled Virus-like Particles with Magnetic Cores. Nano Lett 2007;7(8):2407-16 View Article PubMed/NCBI
  279. Li J, Wang J, Yao Q, Li T, Yan Y, Li Z, et al. Why synthetic virus-like nanoparticles can achieve higher cellular uptake efficiency?. Nanoscale 2020;12(27):14911-14918 View Article PubMed/NCBI
  280. Mohammapdour R, Ghandehari H. Mechanisms of immune response to inorganic nanoparticles and their degradation products. Adv Drug Deliv Rev 2022;180:114022 View Article PubMed/NCBI

About this Article

Cite this article
Bernardo RR, de Oliveira LAS, Paz GS, Fernandes J. The Combination of Cutting-edge Strategies in Nano-delivery Systems to Overcome Drawbacks for Malignant Tumor Treatment. J Explor Res Pharmacol. 2025;10(3):e00020. doi: 10.14218/JERP.2025.00020.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
March 31, 2025 May 9, 2025 July 31, 2025 August 28, 2025
DOI http://dx.doi.org/10.14218/JERP.2025.00020
  • Journal of Exploratory Research in Pharmacology
  • pISSN 2993-5121
  • eISSN 2572-5505
Back to Top

The Combination of Cutting-edge Strategies in Nano-delivery Systems to Overcome Drawbacks for Malignant Tumor Treatment

Robson Roney Bernardo, Luiz Augusto Sousa de Oliveira, Grazielle Silva Paz, Janaina Fernandes
  • Reset Zoom
  • Download TIFF