Abstract
The article provides an insightful overview of the pivotal role of nanotechnology in revolutionizing cancer diagnosis and treatment. It discusses the critical importance of nanoparticles in enhancing the accuracy of cancer detection through improved imaging contrast agents and the synthesis of various nanomaterials designed for oncology applications. The review broadly classifies nanoparticles used in therapeutics, including metallic, magnetic, polymeric, and many other types, with an emphasis on their functions in drug delivery systems for targeted cancer therapy. It details targeting mechanisms, including passive and intentional targeting, to maximize treatment efficacy while minimizing side effects. Furthermore, the article addresses the clinical applications of nanomaterials in cancer treatment, highlights prospects, and addresses the challenges of integrating nanotechnology into cancer treatment.
Keywords: nanomaterial synthesis, targeted drug delivery, nanotechnology in cancer, therapeutics
Introduction
Highlights
Nanoparticles for cancer diagnosis
Synthesis of nanomaterials.
Nanoparticles in imaging enhancement.
Drug delivery via nanotechnology.
Mechanisms of targeting cancer cells.
Real-world clinical applications.
Despite notable progress in the field of medical research and Technology, cancer continues to be an illness that has a restricted range of therapeutic options. The occurrence of metastasis and cancer recurrence significantly contribute to both disability and mortality. However, the precise mechanisms behind these processes have yet to be fully understood1,2.
Existing methods for diagnosing cancer involve the use of imaging techniques, laboratory testing, and the examination of tissues and cells for their physical characteristics. This approach is generally regarded as very dependable for most cancer diagnoses3. Pathological features, including immunohistochemistry (IHC) analysis, histological changes, and mutational and molecular genetics studies, are also utilized in the diagnosis of cancer4. Standard cancer treatment typically involves surgical excision, chemotherapy, radiation therapy, and biological therapy. Surgery is a highly efficient method for eliminating malignant solid tumors, particularly when performed during the initial stages of cancer progression. Combined therapy encompasses a range of treatments, including surgical intervention, chemotherapy, and radiotherapy. Chemotherapy has gained popularity over the years due to its ease and convenience in treating patients with cancer5.
Chemotherapy and radiation treatment alone can only cure 5% of cancer patients; however, surgery can cure around 45% of cancer patients3. The remaining patients do not get well from this illness. Of all the cancer therapies available, although chemotherapy is a commonly used cancer treatment, it nevertheless has some serious drawbacks. The inability to diagnose cancer early, the dispersion of non-targeted drugs, etc., are the limits of cancer therapy6. Since the non-targeted medication concentration spreads throughout the body, excluding the tumor site, it is less able to monitor the drug’s effects on the body and treat the tumor site. One of the most concerning aspects of employing conventional cancer therapy is that the drug’s toxicity level impacts both healthy and tumor cells. Heart failure is one of the major consequences that the poisoning may induce. The scientific community is working to develop novel therapies employing cutting-edge nanotechnology to address these issues. Dendrimers, polymeric, magnetic, liposomal, and nucleic acid-based nanoparticles are among the most widely employed types of nanocarriers7,8. By reducing the drug’s toxicity level and, therefore, raising its effectiveness and enabling it to reach a steady state therapeutic level over a longer period, nanocarriers may enhance the therapeutic index of presently existing medications. The area of nanotechnology is always evolving and is quickly emerging as the most vital component in the fight against cancer. This review article aims to elucidate the pivotal role of nanotechnology in advancing cancer diagnosis and treatment, explore the functional diversity of nanoparticles in oncologic applications, and assess future directions and challenges in nanomedicine for cancer care.
Importance of nanoparticles in the diagnosis of cancer
Nanoparticles, including magnetic, metallic, metal oxide, polymeric, graphene, quantum dots, liposomes, fullerene, dendrimers, and carbon nanotubes, are used in the diagnosis of breast, colon, and cervical cancer….. etc9,10. They also play a crucial role in imaging capabilities. Nanoparticles persist in the bloodstream for extended periods before reaching their intended cellular targets, navigating biological barriers like cell wall membranes. Cancer-specific antigens can be connected to nanoparticles to enhance the affinity and identification of cancer cells. Current research shows the potential of sensors and nanoparticles to improve tumor detection sensitivity and cancer diagnosis11. Metal nanoparticles are more sensitive in identifying cancerous cells due to their ability to be linked with cancer-specific antibodies. Nanocomposites incorporating multiple types of nanoparticles, including cobalt, silver, platinum, copper, and gold, exhibit superior potential for scanning and detecting cancer. These nanomaterials have distinct chemical, physical, and magnetic characteristics, enabling resolving imaging scanners with enhanced contrast, increased sensitivity, and regulated distribution within living organisms. These characteristics can result in clinical benefits such as early identification, immediate disease evaluation, and customized medical treatment12.
Nanomaterials synthesis
Nanomaterials can be synthesized using either conventional methods or environmentally friendly technology. The synthetic techniques encompass thermal decomposition, chemical vapor deposition, solvothermal preparation, hydrothermal treatment preparation, combustion, pulsed ablation using lasers, templating, microwave synthesis, traditional sol-gel synthesis, and gas phase method. Every technique possesses its own merits and drawbacks13.
Function of nanoparticles in contrasting agents and image enhancement in cancer
Research primarily focuses on improving cancer imaging techniques, with nanoparticles playing a crucial role in diagnosing and treating cancer, particularly as photosensitizers, and their value in personalized therapies based on nanotechnology in medicine14. MRI is a crucial application that utilizes magnetic nanoparticles. Iron oxide nanoparticles (IONPs), are widely used in liver and lymph node imaging and cell tracking14. Nanotheranostics, a promising strategy for cancer treatment and imaging, utilizes intricate nanoparticles made of polymeric substances, bismuth, proteins, gold, and calcium phosphate. These nanoparticles have been shown to improve screening, imaging, and early identification of patients with colorectal cancer (CRC). Colonoscopy is the primary method for diagnosing CRC, but alternative techniques like transrectal ultrasonography, MRI, and computed tomographic colonography (CTC) have also been employed for CRC detection15. MRI has made significant progress in diagnosing CRC, but it’s not highly efficient for detecting small polyps. Alternative methods like Fluorodeoxyglucose-positron emission tomography and (FDG-PET) have been used, but they have shown inaccurate results due to radiation exposure15. Metallic nanoparticles and cancer-specific antibodies have improved the detection of breast and colon cancer in urine or blood tests. Proteins and biomarkers present on cell membranes or extracellular environments enable precise diagnosis across imaging modalities like computed tomography (CT) and MRI. Photoacoustic imaging offers high resolution and strong contrast in optics, while Raman imaging is an efficient method for identifying cancer cells. These techniques enhance precision in tumor excision and can be validated through histological examination15.
Chemotherapy, despite its high toxicity and detrimental adverse effects, is a primary treatment approach employed for treating different forms of cancer. Multiple investigations have been undertaken to identify precise transporters for this treatment16. Magnetic microcapsules and nanoparticles have shown improved therapeutic effects in chemotherapy for liver and brain cancers. Prodrugs can enhance drug distribution, biodegradable polymers, and macromolecular matrix approaches. Magnetic nanoparticles have minimal toxicity and have numerous biomedical uses, especially in cancer diagnosis and treatment16.
Currently, extracellular vehicles (EVs) are widely recognized as nanocarriers that can help to some extent in resolving the problems related to chemotherapy. According to reports, EVs can effectively traverse the natural membrane due to their smaller dimensions and have the opportunity to stay in the blood circulation for a long time, allowing them to target cells more effectively. EVs exhibit reduced toxicity and immunogenicity compared to alternative medication delivery strategies. Research has demonstrated that drug delivery via electric vehicles is successful in treating cancer, as evidenced by positive outcomes in both laboratory and live tumor and cancer models. EVs offer numerous benefits, including enhanced loading and target efficacy, improved circulation duration, and reduced occurrence of side effects. The utilization of diverse nanoparticles in the diagnosis of cancer is documented in Table 1.
Table 1.
Utilizing nanoparticles to diagnose cancer17
| Type of nanoparticles | Properties | Cancer diagnosis example |
|---|---|---|
| Lipid-based nanoparticles | Superior biocompatibility and little toxicity as compared to inorganic nanoparticles | The process of attaching anti-HER2 antibodies to phospholipid-coated quantum dots (QDs) demonstrated the capability to specifically target tumors that are positive for the HER2 protein |
| Carbon-based nanoparticles | The physio-chemical features of this substance are exceptional, characterized by its ability to penetrate the cell membrane easily, its large surface area, and its excellent ability to load drugs | Both in-vivo and in-vitro investigations demonstrate that nanodroplets serve as a very efficient contrast agent for both photoacoustic and ultrasonic imaging |
| Polymeric nanoparticles | Encased inside a polymer shell | Targeted tumor delivery is achieved by the use of block copolymer-coated nanoparticles conjugated using pigment molecules and RGD peptides |
| Ceramic nanoparticles | Excellent biocompatibility | Gold nanoparticle-based photothermal treatment is being considered for clinical research to effectively destroy recurrent head and neck cancers and for cancer imaging purposes |
| Metallic nanoparticles | Magnetic nanoparticles have a vital role in the identification and prevention of metastatic breast cancer | Utilizing gold nanoparticles for Raman imaging |
RGD peptides are short amino acid sequences that contain the amino acids arginine (R), glycine (G), and aspartic acid (D); The name “RGD” comes from the initials of these three amino acids.
Frequently used nanoparticles in the therapeutic of cancer
Nanotherapeutics is the term used to describe the use of nanoparticles in the field of medical diagnosis18. In recent times, nanotechnology has advanced to enhance clinical therapeutics, owing to its heightened sensitivity and capacity for early identification of cancer. Various types of nanomaterials, including quantum dots, polymeric nanoparticles, dendrimers, and carbon nanotubes, are utilized in cancer therapeutics. To improve the ability of nanoparticles to detect cancer, they can be combined with antibodies, aptamers, peptides, carbohydrates, and other additional little compounds that selectively target molecules and enable them to reach the desired location19. Research has been carried out employing gold nanoparticles for cancer diagnosis. Au-doped nanoparticles exhibited remarkably high luminous intensity20. In addition, fluorescence has been employed to detect the cancer biomarker carcinoembryonic antigen (CEA) using color visualization. An identifiable red color change was used to establish the limit of detection (LOD), which was found to be 10.00 ng/ml, while an LOD of 0.10 ng/ml was attained by differentiating fluorescence intensity. An assessment was conducted on the feasibility of this approach using actual clinical specimens21. According to reports, using an immunoassay technique provides better control over the placement of antibodies and has the potential to accurately analyze the liver cancer biomarker Hsp90α22. In addition, miR-377-3p and miR-381-3p were employed as diagnosis biomarkers for CRC22.
Because of their quick detection times, nanoparticles made of gold, silica, silver, iron, and magnetic oxide are used in the identification of cancer and economic nature. Furthermore, When compared to radiotherapy and chemical-based treatments, they have fewer side effects. Electrochemical biosensors are characterized by their simplicity, affordability, and effectiveness, making them a highly valuable approach to cancer therapeutics23. Functionalization of nanoparticles can enhance their ability to detect cancer. As an illustration, in the identification of breast adenocarcinoma cells, cancer-specific antibodies were linked with polyethylene glycol (PEG). The antibody-PEG complex was affixed to the surface of the nanoparticles by a group that contains sulfur located at the far end of the PEG linker19.
Metallic nanoparticles
Metallic nanoparticles, ranging from 1 to 100 nm in size, are classified into four categories: metallic nanowires, metallic nanoplatelets, metallic nanoparticles, and metallic nanostructures. Due to their intense energy, they clump together, promoting coalescence through thermodynamic processes. To stabilize metallic nanoparticles in liquids, two methods are commonly employed: static stabilization and steric stabilization. Static stabilization involves attaching negatively charged ions to the surface of the nanoparticles, forming an electrical double-layer. This electrostatic repulsion between particles helps prevent their aggregation. On the other hand, steric stabilization entails coating the nanoparticles with a surfactant, polymer, or ligand. This protective layer creates a physical barrier that inhibits the close approach of nanoparticles, thus preventing their agglomeration24.
Platinum nanoparticles (PtNPs)
PtNPs have significant medical applications due to their antioxidant properties, which suppress tumor growth. They also enhance tumor targeting through the use of ligands that bind to functionalized metal PtNPs. PtNPs also promote superior drug release and delivery effectiveness. However, current research shows harmful consequences due to the nanoparticles’ size, which tend to concentrate in vital organs and cells25. Table 2 exhibits the utilization of platinum-based nanoparticles in cancer therapies.
Table 2.
Use of platinum-based nanoparticles in many cancer therapy methods
| Cancer | Nanoparticle type | Application | Results | Ref. |
|---|---|---|---|---|
| Colon cancer cells | Pt/MgO nanoparticles | Induced apoptosis in colon cancer cells | Colon cancer cells have a decrease in the expression of Bcl2. Elevated expression of Bax and p53 in colon cancer cells under laboratory conditions | 26 |
| Colon cancer cells | Gold-decorated palladium and platinum nanoparticles | Enhanced the efficacy of simulated proton therapy for anti-cancer treatment | Apoptosis-induced cell death in cancer cells (in vitro) | 27 |
| Glioblastoma and melanoma cells | Ag-Pt nanoparticles | Managing the cancer cell types glioblastoma and melanoma | There were no harmful effects seen in healthy cells when tested in a laboratory setting (in vitro) | 28 |
| Neuroblastoma cancer | PtNPs | Stimulation of cancer cell apoptosis | The PtNPs caused cancer cells to undergo apoptosis, resulting in their death. Additionally, they generated oxidative DNA damage in vitro | 29 |
Ag, silver; Bax, is a pro-apoptotic protein and a member of the Bcl2 family; Bcl2, B-cell lymphoma 2 proteins; MgO, Magnesium oxide; p53, is a tumor suppressor protein that acts as a central regulator of cellular responses to various stress signals; Pt, platinum; PtNPs, platinum nanoparticles.
Gold nanoparticles
Because of their numerous distinctive qualities, gold nanoparticles (AuNPs) are useful for a range of medical purposes. AuNPs are diminutive molecules that exhibit numerous distinctive features suitable for imaging techniques. Gold nanoparticles in cancer imaging techniques provide extended circulation durations in the bloodstream, resulting in improved tumor targeting and higher-quality diagnosis. In addition, AuNPs have a broad range of uses, including nucleic acid delivery, medication administration, radiotherapy, and photothermal ablation26. AuNPs can be produced in diverse dimensions and configurations, exhibiting exceptional adaptability26. Furthermore, AuNPs have exhibited low cytotoxicity and enhanced biocompatibility, rendering them very attractive contenders for clinical applications (Table 3).
Table 3.
Use of gold nanoparticles in tumor imaging and identification
| Nanoparticles | Nanoparticle type | Imaging | Application | Ref. |
|---|---|---|---|---|
| Gold nanoparticles | Spheres | X-ray imaging | High payload delivery | 30 |
| Rods, shells, labeled spheres | Fluorescence imaging | |||
| Stars, spheres | Imaging using surface-enhanced Raman spectroscopy | Produces a powerful electromagnetic signal | ||
| Clusters, spheres, rods, shells | Photoacoustic imaging | Demands high levels of absorption within the near-infrared (NIR) range | ||
| Primarily spheres | Optical imaging | The light utilized falls within the near-infrared (NIR) spectrum |
The Food and Drug Administration (FDA) has approved many therapeutic items based on AuNPs, which are currently accessible in the market. Additionally, several other formulations of AuNPs are currently undergoing experimental phases. Table 4 shows how gold nanoparticles are used in various cancer identification and diagnosis methods.
Table 4.
Effects of gold nanoparticles on various cell types
| Type of cell | Application | Results | Ref. |
|---|---|---|---|
| Finding a biomarker for cancer carcinoembryonic antigen (CEA) | Fluorescence of resonance energy transfer (FITC) via Fluorescein isothiocyanate (FRET) | More selectively, fluorescence FITC employing the FRET approach may identify cancer cells (in vitro, in vivo) | 20 |
| MicroRNA biology | Detection of miRNA-155 | Under ideal experimental circumstances, it is capable of detecting cancer (in vitro, in vivo) | 10 |
| Oral squamous cell carcinoma | Gold nanorod assay in conjunction with nano-ELISA | The sensitivity of cancer analyses was increased using enzyme-linked immunosorbent assay (ELISA) (in vitro, in vivo) | 27 |
| Prostate-specific antigen | Nanomaterial based on immunosensor | Superior specificity, sensibility, and durable-term stability immunosensor for in-vitro cancer bioassay analysis | 28 |
Magnetic nanoparticles
Magnetic nanoparticles are characterized by their small dimensions (10–50 nm), strong affinity for binding, and possession of various advantageous attributes, including the existence of holes, protons, electrons, and both positive and negative charges. Moreover, they are harmless and compatible with living organisms. They are commonly employed as a labeling bioconjugate for the detection of cancer biomarkers (Table 5). Furthermore, nanoparticles have a low detection limit and shorter test duration due to their interaction in a liquid solution rather than on an electrode’s surface. They have significant saturation magnetization, allowing particles to pass through tissues and organs easily. They exhibit stability even in aqueous solutions with a neutral pH of 7. Nanoparticles can be administered intravenously for cancer identification and can influence cells and transport medications to specific areas. They are ideal for MRI and can be easily manufactured and customized to meet individual needs. They are also safe and free from harmful substances, making them suitable for use in medical and environmental contexts. They can be easily manufactured and customized to meet individual requirements, making them an excellent choice for various applications.
Table 5.
Applying magnetic nanoparticles for cancer screening and detection
| Type of nanoparticle | Cancer cells | Applications | Ref. |
|---|---|---|---|
| Superparamagnetic iron oxide nanoparticles | Pancreatic cancer cells | MRI-based pancreatic cancer diagnosis and the possibility of early detection with focused techniques | 31 |
| Surface-modified magnetic nanoparticles | Colon cancer cells | For in-vitro colon cancer cell therapeutics | 31 |
| Magnetic nanoparticles | Liver cancer cells | Improved in-vitro identification of liver malignant cells | 21 |
| Magnetic nanoparticles | Brain cancer cells | In vivo, cancer identification and therapy using magnetic nanoparticles as contrast materials | 32 |
| Magnetic gold nanoparticles | Breast cancer checks | Breast cancer detection with ELISA, particularly for those with HER2 breast cancer | 33 |
ELISA, enzyme-linked immunosorbent assay.
Nanoparticle synthesis involves various techniques, allowing for precise characteristics like enhanced magnetic potency or enhanced drug transportation. Control over dimensions, configuration, and constituents allows for selective reactions with chemicals or cells. Functionalized magnetic nanoparticles, using magnetic fields or near-infrared light, have been used to treat cancer by enhancing their ability to interact with specific chemicals or cells29.
Ferrous oxide nanoparticles are beneficial for in-vivo research due to their high degradability and cytotoxic action. They are commonly used in cancer treatment using magnetic fields to produce oxygen radicals and can be manipulated from a distance using an external electromagnetic field. These nanoparticles can be locally hazardous due to reactive nitrogen and oxygen species but are beneficial for treating tumors. Iron oxide nanoparticles, loaded with anti-cancer medications, have advantages over traditional treatments due to their ability to be regulated from a distance, as demonstrated in a breast cancer mouse model30. During the progression of breast cancer, there is an observed overexpression of miRNA-155, which is recognized as a biomarker for this type of cancer. Mapping miRNA-155 is challenging because of the absence of highly sensitive approaches. A novel methodology has been devised to address this issue, enabling the fast production of magnetic nanoprobes capable of quantifying miRNA-15534.
Polymeric-based nanoparticles (PNPs)
PNPs are solid substances with dimensions ranging from 1 to 1000 nm, exhibiting strong mechanical, electrical, thermal, and optical properties. They can be combined with drug delivery methods, fluorophores, and active pharmaceutical ingredients for improved efficacy35–37. Nanoparticles are used in drug delivery to protect medication molecules. Biopolymers, including PNPs, are used for pharmaceutical administration, including chemotherapeutic medicines, antiviral compounds, essential nutrients, antioxidants, and plasmid DNA. PNPs are produced from biodegradable and non-biodegradable synthetic polymers. Non-biodegradable polymers like poly-acrylates are used for drug administration through the skin, while biodegradable polymers like poly(lactic-co-glycolic acid (PLGA) are used for transdermal drug delivery35. PNPs have been employed to enhance optical and magnetic resonance imaging techniques to diagnose brain cancer38. Dendrimers are a type of three-dimensional core-shell polymer that possesses the capacity to cross the blood-brain barrier, hence enhancing their targeting capabilities. Dendrimers, namely poly-amidoamine, are extensively utilized for drug delivery purposes. In particular, dendrimers that are linked with tamoxifen serve as effective drug carriers38. A surfactant is frequently used during the manufacturing of polymeric-based nanoparticles to help reduce the solution’s surface tension. Subsequently, the surfactant is combined with the monomer, which serves as the fundamental unit of the polymer. Subsequently, the monomer undergoes polymerization, resulting in the formation of nanoparticles.
Polymeric nanoparticles exhibit certain drawbacks, including limited stability and an aggregation propensity. Their qualities might provide challenges in certain applications, such as drug delivery39. In addition, the synthesis of nanoparticles based on polymers might be challenging due to the requirement for precise control of reaction conditions. Polymeric nanoparticles are susceptible to oxidation, which can restrict their application in certain contexts. Polymeric-based nanoparticles pose challenges for scaling up in industrial applications due to the need for meticulous monitoring of reaction conditions. Protease-activated near-infrared fluorescent polymeric nanoparticles, doxorubicin-loaded, have been utilized for cancer detection and treatment39.
Metal oxide nanoparticles
Metal oxide nanoparticles are particles of metal oxides that are extremely small in size. Metal oxide nanoparticles, such as nickel oxide (NiO), zinc oxide (ZnO), manganese dioxide (MnO2), iron(III) oxide(Fe2O3), titanium dioxide (TiO2), and cobalt oxide (Co3O4), are composite materials composed of many metal oxides13,40. These nanoparticles have been recently employed in electro-analysis to identify and measure biomolecules. Utilizing metal oxides for biomolecule detection provides benefits like enhanced biocompatibility for enzymes, hence enhancing detection precision. Metal oxide nanoparticles can modify their structure, hence influencing the electrical conductivity and chemical properties of nanoparticles41. Moreover, oxides of transition metals can decompose diverse colors when exposed to sunlight or UV light. The use of ecologically friendly and sustainable synthesized biological components, such as metal oxide processes, is becoming more and more prominent. The green approach has numerous advantages, including the utilization of inexpensive constituents obtained from diverse plant sources, which may be readily expanded to produce Nanostructures of transitional metal oxides42. The nanoparticles of metal oxide are widely used and have a variety of applications in therapeutic and therapeutic contexts. For instance, zinc oxide (ZnO) nanoparticles were employed as a remedy for cardiovascular illness associated with diabetes. This was done by injecting streptozotocin into rats with diabetes. The findings demonstrated that administering a small dosage of ZnO nanoparticles effectively shielded heart cells from harm by decreasing serum concentrations of cholesterol33.
Quantum dots (QDs)
QDs are minuscule crystals that possess the capability to convey electrons. When exposed to UV light, QDs can generate light of several colors with exceptionally high energy. These QDs have numerous applications, such as in the advancement of solar cells, fluorescent biological markers, and nanocomposites. High-resolution cellular imaging has led to advancements in cancer diagnosis. The fluorescence characteristics of quantum dots undergo alterations when exposed to various substances. Furthermore, QDs possess an inert region on their outer layer, which allows for the convenient attachment of particular antibodies. QDs have been utilized in many bio-applications for imaging purposes. QDs have been used in drug administration and cancer therapy, particularly in treating lung cancer. They effectively eradicate bacteria-induced infections and suppress lung cancer cells’ P-glycoprotein DNA and protein activity by stimulating miR-34b and miR-185 production. These miR-34b and miR-185 targets for lung cancer treatment. Combining QDs with Camellia sinensis leaf extract has been shown to impede the lung tumor cell cycle and reduce cancer cell viability43. Another research investigation found that the application of uncapped cadmium telluride quantum dots (520Q cytotoxic, 580Q cytotoxic, and 730Q non-cytotoxic) caused oxidative stress in lung malignancies and human bronchial epithelial cells. Nevertheless, it was shown that 730Q had a discernible impact only when the exposure time was extended but not when it was shortened44. In addition to their numerous advantages, QDs include heavy metals like cadmium, which is a recognized hazardous substance and carcinogen. This presents potential risks for their use in therapeutic applications.
Graphene
Graphene, commonly known as individual graphite atomic layers, has a planar structure composed of uniform hexagonal rings. It possesses exceptional thinness, transparency, and lightness and exhibits excellent thermal and electrical conductivity, rendering it a highly desirable substance for the identification and detection of cancer. Graphene possesses several advantageous characteristics, such as its ability to exhibit a bipolar transistor impact and generate significant quantum fluctuations45. These traits render it a promising contender for efficient cancer imaging. Graphene possesses a significant specific surface area, making it advantageous for the incorporation of anti-cancer medicines as a result of the existence of π-π stacking and hydrophobic interactions45. Graphene oxide, an oxidized version of graphene, has applications in cellular visualization, drug delivery, and cancer treatment. It improves the properties of complex materials, and a study showed it can identify cancer cells through isotope separation of graphene Au nanocrystals46.
Fullerene
Fullerene is a carbon allotrope with a lattice-like structure used in chemical applications to encapsulate drug molecules for effective drug delivery. Its C60 fullerene has been used in oncology for cancer diagnosis and detection and in biosensors for detecting glucose concentrations in blood serum. Fullerene’s interconnected carbon atoms allow for efficient drug delivery and have been successfully used in biosensors47. He can be captive by subjecting C60 to heating in the presence of helium vapor and applying pressure, as demonstrated by another study48. A recent study has found that fullerenes and metal nanostructures possess reducing capabilities that enable them to defend people from internal and external threats generated by reactive oxygen molecules. Recent research has discovered that fullerenes and metallic nanomaterials can specifically eliminate unhealthy cells in tissues, hence preventing the development of prolonged inflammatory illnesses. Fullerenes and metallic nanomaterials possess significant prospects in the management of diseases accompanying aging48.
Carbon nanotubes
Carbon nanotubes are cylindrical carbon atoms with exceptional thermal and electrical conductivity, resilience, and lightness. They are useful in rechargeable lithium batteries, electronic nanodevices, hydrogen storage, composite materials, biosensors, and touch screens due to their 1 nanometer diameter and significant length. Single-wall nanotubes have better properties than multi-wall nanotubes like silver or copper49. Furthermore, carbon nanotube biosensors have been employed for the detection of organophosphorous chemicals; as demonstrated in previous works, Carbon nanotubes and gold nanoparticles were placed onto a gold cable. An enzyme called acetylcholine-esterase was used in the biosensor. They were mounted on carboxylic-functionalized single-walled carbon nanotubes. These nanotubes were then attached to the cardiac electrode. Nafion was added to the electrode to prevent enzyme release and convert it into a sensor electrode. This nanosensor operates based on the mechanism of suppressing the acetylcholinesterase enzyme49. A research experiment was performed in which Carbon nanotubes with carboxyl functionality were introduced to a human T-cell cell line. This led to the activation of caspase-2 gene expression within the living cells. The findings demonstrated a marginal decline in the cellular survival of cancer cells when subjected to carboxyl-functionalized carbon nanotubes. The molecular study revealed an upregulation of Cas2 mRNA in the cancer cells that were subjected to treatment with Carbon nanotubes with carboxyl functionality50. Nanotubes of carbon have been utilized in the domain of cancer therapeutics and imaging due to their biological compatibility, thermodynamical characteristics, and flexible functionality51.
Liposomes
Liposomes are lipid-based nanoparticles consisting of a sealed lipid bilayer sphere containing an inner chamber for water solutions. The bilayer consists of two phospholipid sheets, a hydrophilic head area, and a hydrophobic tail, with hydrophobic tails exhibiting mutual attraction and membrane heads orienting towards water52. This structure consists of a bilayer of phospholipid molecules, which serves as An impermeable barrier that restricts the release of the internal solution to the exterior. Subsequently, the solution can be conveyed to the desired location by liposomes. They are innovative in the field of Pharmaceutical medication delivery methods and drug transfer mechanisms (Fig. 1). Liposome nanomolecules have shown a substantial influence on chemotherapy by enhancing selectivity, reducing cytotoxicity, and improving the solubility of hydrophobic medicines (Fig. 1). Microfluidics can be utilized to create small liposomes measuring around 50 nm in diameter, whereas giant liposomes measuring 75 nm in diameter demonstrated comparable drug retentivity in both in vitro and animal experiments. However, the extent of leakage was influenced by the size of the liposomes, with smaller liposomes exhibiting superior distribution throughout the tissues compared to larger liposomes52. Liposomes play a crucial role in chemotherapy administration, as they enhance transfection effectiveness and reduce chemo-resistant proteins. Studies have used liposomes to transport “Glucose regulated protein 78” and concentrate 1, 2-dioleoyloxy-3-trimethylammoniumpropane liposomes in cancer stem cells and mammary tumors, demonstrating effective transportation in both types of cancer52. Furthermore, magnetic liposomes were employed in the field of cancer therapy. Specifically, liposomal IR-780, an exceptionally stable nano-therapeutic drug, has been employed to cure brain tumors.
Figure 1.

Applications and structure of liposomes (reproduced with permission from Noor et al. 53).
Dendrimers
Dendrimers are precisely engineered synthetic macromolecules characterized by an accurately specified three-dimensional arrangement and a large number of functional groups. Dendrimers, which are characterized by their compact size and dendritic architecture surrounding a core made of linear polymers, have found applications in the field of nanomedicine. Dendrimers serve as effective vehicles for the transportation of medications and genetics. Additionally, they may serve as antibacterial agents that are antibacterial and antitumor (Fig. 2). Dendrimers consist of three different layers: the center molecular, the middle layer, and the exterior layer. Furthermore, they possess viscosity, dissociation, and cellular properties. Dendrimers, due to their multipurpose structure, are valuable for creating advanced nanodevices in imaging and diagnosis. They have numerous applications in the medical field, including drug delivery, encapsulation, conjugation, nanocarriers, anti-cancer agents, gene delivery, photodynamic treatment, medical theranostics, and biosensors. Their contributions to materials sciences are substantial54. Furthermore, Dendrimers are used in the transportation of anti-cancer drugs due to their durability, solubility in water, and reduced immunogenicity. They can induce hypervascularization, increase tumor permeability, and impair lymphatic drain, facilitating passive targeting. Encapsulation or conjugation of dendrimers into therapeutic molecules may lead to safer and more effective medicines45. Dendrimers are created through divergent synthesis, a process where monomers are gradually incorporated into a central nucleus, forming a complex, branching architecture. Modifying these monomers allows for changes in the dendrimer’s dimensions, configuration, and chemical responsiveness. This intricate process requires careful management of reaction parameters. Dendrimers can be customized by changing monomer choice, reaction parameters, and monomer amount. They have unique properties, such as exceptional stability, durability, and solubility, making them useful for targeted drug delivery.
Figure 2.

Dendrimers’ composition and use in drug conjugation and encapsulation (reproduced with permission from Noor et al.53).
Nanostructure lipid carriers (NLCs)
Nanocarriers based on lipids have demonstrated encouraging outcomes in the field of cancer therapeutics and treatment. Polymeric nanoparticles, formed by lipid-based chemicals or in conjunction with vectors like liposomes, ethosomes, and transfersomes, can potentially address challenges associated with drug absorption resistance in biological membranes. The synergistic impact of lipid-based nanocarriers is recognized for enhancing the effectiveness and precision of polymeric nanoparticles55. A study was performed to investigate the efficacy of a medication that combines nanostructured lipid carriers loaded with quercetin and coated with conatumumab, which is sensitive to reactive oxygen species and is being used to treat colon cancer19.
Alternative nanoparticles and their utilization in diverse cancer cell types
The utilization of diverse nanoparticles has resulted in the proliferation of nanotechnologies across multiple domains, including pharmaceuticals, energy production, chemicals, drug administration, and enhancement of operational effectiveness. Various types of nanoparticles are currently employed for cancer cell detection and monotherapy. The functionalities of these nanoparticles can be determined by their size, kind, and structure56. Nanoparticles, including amino acids and semiconductor nanoparticles, are unique and deviate from ordinary particles. Nanorobots, miniature machines made of nanoscale molecules, can interface with the cellular barrier, providing direct access to cellular locations. They can enhance treatment effectiveness through advanced biological therapies and less invasive procedures. Currently, nanorobots are designed to identify 12 different types of cancer cells, demonstrating their potential in cancer treatment and screening. The effect of nanoparticles on different varieties of cancer cells, including cancer of the colon, neuroblastoma, breast cancer, glioblastoma and melanoma, oral squamous cell cancer, cervical cancer, prostate cancer, lung cancer, liver cancer, pancreatic cancer57, bladder cancer, brain cancer, cancer stem cells, and colorectal cancer58, has been studied. Table 6 provides a summary of the utilization of various nanoparticles in the field of cancer therapy.
Table 6.
Using several kinds of nanoparticles in cancer treatment
| Nanoparticle | Cancer type | Application | Ref. |
|---|---|---|---|
| Fumed silica nanoparticles | Detecting cancer pathways | For more accurate cancer detection, multi-site phosphorylated peptides may be effectively bound by nanoparticles (in vitro, in vivo) | 59 |
| miRNA | CRC | It may serve as a circulating biomarker for the early detection of CRC (in vitro). | 60 |
| Raman-active nanoprobe | Circulating cancer stem cells | Enhanced imaging via the use of the Raman imaging technique for the identification of cancerous cells (in laboratory settings and inside living organisms) | 61 |
CRC, colorectal cancer.
Creating nanomaterials to deliver drugs
Traditional administration methods like oral or intravenous injections have drawbacks, but controlled release systems offer numerous benefits, such as enhanced medicine efficacy, reduced side effects, and improved patient adherence to therapy, attracting medical professionals and patients, thereby creating significant market opportunities62. In recent years, various controlled release methods, such as micelles, nanoparticles, hydrogels, and electrospun barriers, were extensively studied to meet specific therapeutic application needs63. Poly (d,l-lactide-co-glycolide) mono/bicomponent electrospun polymer membranes have received approval from the FDA and have been employed as implant materials for an extensive duration. By changing the proportion of lactic acid and glycolic acid in the copolymer, these membranes’ mechanical characteristics and rate of decomposition can be customized63. To change the fibrous membrane’s swelling characteristics and regulate its rupture pattern, five distinct polymers, all of which have obtained FDA authorization for use in implantation, were included through the fiber layer as a secondary component. The polymers mentioned were poly(ethylene glycol) and poly(ethylene glycol)-b-poly(d,l-lactide)63. Hydrophilic polymer carriers often exhibit a rapid elimination profile, while hydrophobic polymers tend to liberate their loaded pharmaceuticals at a gradual rate. Extensive research has been conducted on multicomponent electrospun fibers that comprise both hydrophilic and hydrophobic polymers for the purpose of drug delivery64. In order to achieve prolonged therapeutic levels of anti-cancer compounds, a combination of rapid and gradual drug release mechanisms was employed using a hybrid material made of poly-caprolactone and gelatin. This material was utilized to encapsulate both free curcumin (CUR) and mesoporous silicon nanoparticles loaded with CUR64.
Drug-targeted delivery system using nanotechnology in cancer therapy
Nanotechnology exhibits significant progress in the field of cancer therapy23. Nanotechnology enables the targeted delivery of therapeutic molecules to specific areas while minimizing harm to healthy cells65. Nanomedicine enhances the stability, solubility, medication half-life, and bioavailability of numerous chemotherapy medicines. Nanomedicine effectively decreased the maximum drug concentration, enhanced drug accumulation at specific locations, and raised the area under the curve65. The small size and distinctive nanoparticle coating of these particles enable the efficient transport of hydrophobic anti-cancer medications to specific areas in the body while minimizing their recognition and elimination by the immune system.
Nanoparticles (NPs) can enhance medication accumulation in cancer cells by increasing their Permeability and retention, a phenomenon referred to as improved retention and accessibility (EPR). In conclusion, The utilization of NPs in conjunction with anti-cancer drugs can augment the effectiveness of treatment by mitigating adverse effects via target-ligand interactions66.
Different forms of targeted drug delivery systems nanoparticles are utilized for breast cancer research (Fig. 3). NPs can be classified into different types, including liposomal, mesoporous silica, polymer, carbon, metal, and protein-based NPs66.
Figure 3.

Classification of nanoparticle formulations used in breast cancer research for targeted drug delivery systems (reproduced with permission from Yedi et al.67).
The efficacy of NPs in drug delivery is contingent upon various factors, including their shape, size, content, and other structural characteristics. NPs possess a complex structure consisting of several components, which are designed to achieve therapeutic and/or therapeutic objectives68.
Nurse practitioners use many ways to exert an effect on cancer cells:
Pharmaceuticals that are shielded against hepatic inactivation, enzymatic decomposition, and fast removal69.
The functionalization of ligands over-expressed in tumor cells, which are identifiable by receptors (targets), has improved the internalization of cancer cells68.
It exerts an influence on the progression of cancer by regulating the tumor microenvironment (TME)70. Polymers have the ability to detect and react to external stimuli such as light, temperature, ultrasound, and electrochemical triggers, as well as the specific conditions within a tumor, such as pH, enzyme activity, and redox characteristics. This enables them to release drugs in order to overcome these obstacles70.
Avoided the mechanisms of multiple drug resistance (MDR) efflux transporters68.
Diminished the occurrence and severity of adverse reactions71.
The contrast moieties were encapsulated in a carrier, enabling direct viewing of the carrier in vivo 72. Table 7 showcases a variety of nanoparticles in different forms that have been clinically studied for treating various cancer types73.
Table 7.
Some of the nanomedicines, the subject of clinical studies in recent years, as compiled from clinicaltrials.gov
| Nanodrug | Conventional drug | Type of cancer | Clinical Trials.gov Identifier |
|---|---|---|---|
| ONPATTRO | Patisiran | Transthyretin amyloidosis | NCT03862807 |
| AGuIX | Polysiloxane gadolinium-chelates-based nanoparticles | Brain metastases | NCT02820454 |
| VYXEOS | Cytarabine, daunorubicin | Acute myeloid leukemia | NCT04920500 |
| Paclitaxel Nab | Mifepristone | Recurrent breast cancer, Male breast cancer | NCT01493310 |
| Advanced or metastatic | Azacitidine (Vidaza) Breast cancer | NCT00748553 | |
| Bevacizumab, Gemcitabine hydrochloride | Breast cancer | NCT00623233 |
Targeting mechanisms
Accurate localization is a key feature of small carriers of tumor cells. Employed in medication transportation since it increases the effectiveness of therapy. At the same time, it safeguards regular cells from harmful effects. Several research studies have been conducted to investigate the design of targeting medications based on nanoparticles. To effectively tackle the difficulties associated with cancer cell targeting and the design of nanocarrier systems, having a thorough comprehension of the biology of malignancies and how they interact between cancerous cells and tiny particles is crucial. The targeting techniques can be categorized into two main groups: both inactive and active targeting (Fig. 4).
Figure 4.

Inactive and active delivery of nanoparticles (NPs) to cancer cells. The targeting of nanoparticles improves the effectiveness of therapy and decreases the toxicity that affects the entire system. The primary method for passively targeting nanoparticles is via the increased Permeability as well as retention phenomenon. This effect takes advantage of the heightened arterial permeability and reduced lymphatic outflow in cancer cells, allowing NPs to target cancer cells selectively without active intervention. Active targeting is accomplished through the interaction between receptors and ligands. The cancer cells possess many types of receptors, including transferrin receptors, folate receptors, glycoprotein receptors (such as lectin), and receptors for the epidermal growth stimulant (reproduced with permission from Yao et al.74).
Passive targeting
Passive targeting pertains to the power of nanoparticles ranging from 10 to 150 nm to selectively move from the circulatory system into tumor tissue. Nanoparticles have the ability to concentrate on tumor tissue precisely because of the improper formation of binding connections among endothelial cells in newly formed blood arteries within tumors3. The process by which nanoparticles passively enter the microenvironment of the tumor is known as the increased permeability and retentivity phenomenon. This phenomenon was first observed around thirty years earlier during the investigation of macromolecule transportation within tumor tissues3.
QDs are known for their light-resistant properties, adjustable emission, and high energy conversion efficiency, making them ideal for applications like tumor tissue visualization due to their passive accumulation and improved permeability and retention impact. Hong et al. 75. utilized a novel near-infrared II (NIR-II) fluorophore, known as six-armed PEG-Ag2S QDs, to visualize subcutaneous xenograft 4T1 mouse tumors. The researchers continuously tracked the distribution of the NIR-II signal in mice over a prolonged time period of time (up to 24 h after injection). The fluorescence emitted by 6PEG-Ag2S QDs in tumors remained high but dropped in other parts of the body and skin within 30 minutes. Until 24 h post-infection, the pharmacokinetics of 6PEG-Ag2S QDs in living organisms showed a remarkable build-up in tumors, with a concentration above 10% ID/gram. This accumulation is attributed to augmented Permeability and retention. The use of near-infrared II quantum dots provides accurate visualization of internal organs, enhanced contrast for tumor imaging, and quicker tumor identification.
Researchers have utilized AuNPs for in-vivo tumor imaging by means of passive targeting. In their study, Lai et al. 76. demonstrated that gold nanoparticles coated with mercaptoundecanoic acid were able to accurately detect and monitor primary glioma cells in the areas where they were injected into the brains of mice. Moreover, these particles accurately identified the microvasculature associated with tumors. Chitosan nanoparticles have been utilized in certain instances for in-vivo imaging by exploiting the enhanced permeability and retention effect. Nam et al. 77. described a targeted nanoparticle for tumors that may be used as a versatile imaging scan for optical/MR (MR: magnetic resonance) dual imaging. This nanoparticle is built on Glycol chitosan self-assembles. By employing conjugation and alteration of chemicals conjugation and alteration of techniques, researchers successfully created chitosan nanoparticles that are both stable and tagged with Cy5.5. Researchers have developed a new method to target tumors in living organisms using glycol chitosan nanocrystals. The nanoparticles, encapsulated by Gd(III), have a spherical morphology and are around 350 nm in diameter. They have been found to be efficient in uptake and dispersion within the cytoplasm. After intravenous treatment in animals with tumors, the nanoparticles accumulate abundantly in the tumor, as confirmed by non-invasive fluorescence imaging and magnetic resonance imaging. EPR effect could significantly influence their potential for cancer identification and control.
The dimensions and morphology of nanoparticles significantly impact their EPR impact, making it crucial to consider these characteristics when designing nanoparticle probes for tumor formation. Nanoparticles smaller than ten nm can be cleared by the kidneys, reducing their accumulation in malignant tissue. Anisotropic molecules increase circulation duration due to the reduced likelihood of permeating liver endothelial gaps. A study using silica-coated QDs with different thicknesses investigated the impact of nanoparticle dimensions on particle accumulation in tumor tissue. 12-nm QDs entered tumor tissue without obstruction, while 60-nm QDs leaked out of blood vessels but remained within 10-micrometer vessels78.
Upon coming into touch with a biological fluid, nanoparticles will acquire a layer of biological components on their surface, known as a “corona”79. The presence of serum proteins on the nanoparticle’s surface (opsonization) significantly alters the way nanoparticles are transported, taken up, and eliminated in the body. Applying PEG as a coating on the surface of a nanoparticle decreases the occurrence of unspecific binding of serum proteins and limits the creation of a protein corona, consequently expanding the duration of nanoparticle circulation. PEGylation of different nanoparticles, like AuNPs and QDs, leads to an extended duration of presence in the bloodstream, as well as gradual build-up in the liver and spleen.
Nanotechnology-based imaging is anticipated to enhance the precision and selectivity of cancer detection while simultaneously decreasing toxicity. In a recent study, Garrigue et al. 80. demonstrated the utilization of nanoparticles and the EPR effect to create a novel nanosystem for positron emission tomography (PET) imaging. The method utilizes a self-assembling amphiphilic dendrimer that maintains different PET reporting units at its ends. The dendrimer exhibited the ability to autonomously create small, homogeneous nano micelles that are selectively concentrated in tumors, so enabling efficient PET imaging. Thanks to the dendrimeric multivalence and tumor targeting inactive facilitated by EPR, the nanosystem demonstrated enhanced imaging sensitivity and heightened particularity. The ratios of PET signals rose by around 14 times compared with the clinical gold standard, 2-fluorodeoxyglucose. Furthermore, the dendrimer exhibited exceptional safety characteristics and favorable pharmacokinetics for PET imaging. The authors assert that their study makes a valuable contribution to the advancement of dendrimer nanosystems for efficient and auspicious cancer imaging.
Targeting with intention
Researchers have explored using cell receptor membrane recognition to target tumor tissues and nanoparticle accumulation for tumor imaging. These techniques increase the amount of nanoparticles transported to tumor tissue, enhancing sensitivity in in-vivo tumor detection. Active tumor targeting is more effective than EPR effect-based passive targeting for early cancer detection using high-contrast imaging81.
Levenson and Nie’s study demonstrates the use of antibody-conjugated QDs to target prostate-specific membrane antigen (PSMA) for tumor targeting. They analyzed in-vivo imaging outcomes of three QD surface modifications: carboxylic acid (COOH), polyethylene glycol (PEG), and a combination of PEG and prostate-specific membrane antigen-antibody. The results showed that active targeting using a tumor-specific ligand is more efficient and faster than passive targeting in terms of incorporation, retention, and tumor penetration81.
A study has shown that regular administration of peptides can effectively target malignant areas in living organisms. The RGD peptide, which binds to the αvβ3 protein, plays a role in cancer angiogenesis and metastasis. A system using gold nanoparticles was developed to target specific cells precisely. The technique involves combining a tumor-homing penetration peptide called iRGD with a legumain-responsive aggregable gold nanoparticle. The compounds, administered intravenously, are specifically attached to the tumor’s blood vessels and distributed throughout the surrounding tissue. iRGD targets tumors through a three-step process: binding to αv integrins on the tumor endothelium, creating a binding motif for neuropilin-1, and controlling cell penetration. This addition enhances the sensitivity of tumor imaging agents and antitumor medicine efficacy82.
A study has developed a platform using DNA to create near-infrared sensitive nanoparticles for cancer treatment. The platform consists of complementary DNA strands, 50 × 10 nm gold nanorods, and a PEG layer. The DNA strands, composed of cytosine-guanine (CG) base pairs, provide binding sites for the chemotherapy medication Dox. The drug loading can be adjusted by changing the amount of CG base pairs. The capture strand transports the medication, while the targeting strand targets cells. Gold nanorods are efficient transducers of near-infrared light, making them useful in cancer thermotherapy and denaturing double-stranded DNA when exposed to NIR radiation. This process releases loaded medications at the desired location for chemotherapy83.
Clinical use of nanomaterials in the treatment of cancer
Nanomaterials have shown promising results in therapeutic scenarios, particularly in the delivery of anti-cancer medications. One example is CRLX101, a medication that combines camptothecin with nanoparticles. It was tested in clinical trials for advanced rectal patients with cancer, showing well-tolerance and a complete pathologic response84. A phase I-IIa clinical trial investigated the effectiveness of CRLX101 in combination with bevacizumab in metastatic renal cell patients with cancer. A phase 1/2a trial tested the effectiveness of CRLX101, a nanopharmaceutical combining a cyclodextrin-containing polymer and camptothecin, on patients with advanced solid tumor malignancies85.
Types of nanoparticles used for each organ cancer
Breast cancer
Polymeric nanoparticles: Used for targeted drug delivery, often carrying chemotherapeutic agents like doxorubicin. The surface of these nanoparticles can be modified with antibodies targeting receptors over-expressed in breast cancer cells, such as HER2.
Liposomes: Liposomal formulations (e.g. liposomal doxorubicin) are used to reduce toxicity and improve the efficacy of chemotherapy.
Lung cancer
Carbon nanotubes: Engineered for targeted delivery of chemotherapeutic agents directly into lung cancer cells, potentially through inhalation therapies that target the lung specifically.
Gold nanoparticles: Utilized in photothermal therapy, where they are targeted to lung cancer cells and then heated with infrared light to kill cancer cells without harming surrounding healthy tissue.
Prostate cancer
Gold nanoparticles: Can be functionalized with prostate-specific membrane antigen (PSMA) ligands to target and image prostate cancer cells specifically.
Magnetic nanoparticles: Used in hyperthermia treatment and as MRI contrast agents, targeted to prostate cancer cells with specific antibodies.
Brain cancer (glioblastoma)
Magnetic nanoparticles: Functionalized to cross the blood-brain barrier (BBB) and target glioblastoma cells, used in conjunction with MRI for diagnosis or hyperthermia for treatment.
Polymeric nanoparticles: Designed to bypass the BBB and deliver chemotherapeutic drugs directly to brain tumors, often modified with targeting moieties that recognize brain tumor cells.
Pancreatic cancer
Polymeric-based nanoparticles: These nanoparticles can be designed to release their drug load in response to the specific microenvironment of pancreatic tumors, such as low pH or high levels of certain enzymes.
Metallic nanoparticles: Especially gold nanoparticles have been explored for the photothermal treatment of pancreatic cancer, where they are targeted to the tumor and then irradiated to produce heat and kill cancer cells.
Future prospects and current challenges
Magnetic nanoparticles, with their nanoscale dimensions, heightened reactivity, and enhanced surface area, can potentially cause harmful effects on human cells or tissues. They can enter the human body through ingestion or inhalation and migrate to different organs and tissues. The environmental impact of nanoparticles is recognized, and efforts are being made to develop harmless materials. Nanoparticles, like dendrimers, can disrupt enzymatic assays or generate excessive charges, potentially affecting cell membrane integrity or cell death. Despite their benefits in cancer therapeutics and imaging, concerns remain about their potential toxicity and adverse effects86. Nanoparticles can accumulate in organs like the liver and kidneys, potentially causing health issues. However, their exact impact on the human body is not yet fully understood. Experts are concerned about the toxicity and potential hazards they pose to the environment and human health, particularly metallic nanoparticles, which can interact with other pollutants and enhance their hazardous effects. The exact impact of these accumulations on the human body is not yet fully understood. Nanomedicine has unique adverse impacts compared to conventional medications due to its varying toxicity and ability to traverse multiple body barriers, including the blood-brain barrier. Nanoparticles, which are extremely small, can also pass through cell membranes, intracellular organelles, and even the nucleus, affecting various organs and potentially causing various effects on the brain87. Nanomedicine balances toxicity and efficacy in therapeutic treatments by combining biological, chemical, and physical characteristics. It evaluates nano-compound biodistribution in pre-clinical and clinical investigations, considering the unique characteristics and limitations of each Technology. It helps assess immediate build-up in cells, organs, and tissues, ensuring a harmonious balance between toxicity and efficacy88.
Green nanotechnology is a promising alternative to harsh methods and toxic chemicals used in nanoparticle synthesis. It involves integrating microbes and plants, resulting in highly optimized, low-toxicity, and environmentally friendly nanoparticles. These nanoparticles are safe for humans and the ecosystem. Intelligent Ag nanoparticles have unique scattering characteristics, promoting resistance against bacteria and aggregation. Their enhanced dispersion features offer potential for smart nanotechnologies in various domains. This approach eliminates the need for foreign stabilizing or coating agents89. Nanoparticles can cross physiological barriers, like blood-brain, and interact with cell membranes, organelles, and nuclei, influencing drug administration in nanomedicine. However, toxicity is largely influenced by the composition of the nanoparticle formula90.
The biodistribution of nanomedicine is crucial in pre-clinical and clinical investigations. Nanoparticles have been found to accumulate on the skin, demonstrating resistance against bacteria and promoting aggregation. This is due to their surface ligands resembling the bacterial membrane, leading to a notable antibacterial impact. The enhanced dispersion features of this invention offer excellent prospects for the utilization of smart nanotechnologies across diverse domains. Research has shown that each Technology has unique characteristics and limitations in real-time substance accumulation91. Nanoparticles of heavy minerals, such as tin, mercury, and lead, are recognized for their stability and resistance to degradation, resulting in significant environmental toxicity. Research has evaluated the overall toxicity, ability to cause genetic damage, and potential to cause cancer in rats exposed to both tannic acid and iron (III) nanoparticles, as well as in both normal and malignant human cells92. The future of nano-based materials for cancer therapeutics lies in the development of biocompatible nanomaterials with reduced toxicity and improved biodistribution.
Conclusion
The development of a more potent cancer therapy is the primary driving force behind the establishment of cancer nanotechnology. Many restrictions exist with the most widely used cancer treatment procedures, such as radiation therapy and chemotherapy. The restrictions have been partially removed with the advent of nanotechnology. Research on nanoparticles, liposomes, dendrimers, and nanoshells is still ongoing in the field of nanotechnology. An expanding toolkit for treating cancer is cancer nanotechnology. Nanotechnology offers many important advantages in cancer diagnosis compared to traditional methods, including:
Early detection: Nanotechnologies allow early detection of cancer at the level of cells or macromolecules, allowing early intervention and thus increasing the chances of recovery.
Accuracy and specificity: Nanotechnology can improve the accuracy and specificity of diagnosis, as nanomaterials can be used to specifically target cancer cells without affecting surrounding healthy tissue.
High sensitivity: Nanotechnologies are more sensitive in detecting subtle changes in the body associated with cancer, which enables it to be detected at early stages and increases the effectiveness of treatment.
Nano-X-ray imaging: Nano-X-ray imaging allows the viewing of tiny structures inside the body with greater accuracy than traditional imaging, which helps in better diagnosing and staging cancer.
Precise guidance and delivery of treatment: Nanotechnology can be used to precisely direct and deliver treatment to areas affected by cancer without affecting healthy tissue, reducing side effects.
Future directions for nanotechnology innovation in cancer therapy include the creation of self-organizing and reactive systems. In addition to continuing to be of assistance, medical science will continue to be greatly benefited by nanotechnology in the years to come. This will be a very exciting period for nanomedicine.
Ethical approval
Ethics approval was not required for this review article.
Consent
Informed consent was not required for this review article.
Source of funding
Not applicable.
Author contribution
O.O.L.: conceptualization, data curation, writing—original draft preparation, writing—reviewing and editing. I.B.A.: writing—reviewing and editing, visualization, supervision. H.H.: writing—reviewing and editing, visualization, supervision. W.H.: writing—reviewing and editing, visualization, supervision. S.Z.: writing—reviewing and editing, visualization, supervision. A.B.A.: writing—reviewing and editing, visualization, supervision. M.B.: writing—reviewing and editing, visualization, supervision. A.A.A.: data curation, writing—original draft preparation, writing—reviewing and editing.
Conflicts of interest disclosure
The authors declare no conflicts of interest.
Research registration unique identifying number (UIN)
Not applicable.
Guarantor
Ali Alnazza Alhamad.
Data availability statement
Not applicable.
Provenance and peer review
Not applicable.
Footnotes
Sponsorships or competing interests that may be relevant to content are disclosed at the end of this article.
Published online 17 June 2024
Contributor Information
Olfa Ouled Ltaief, Email: olfa.ol@live.fr.
Ilham Ben Amor, Email: ilhambenamor97@gmail.com.
Hadia Hemmami, Email: hemmami.h@gmail.com.
Wiem Hamza, Email: hamzawiem@gmail.com.
Soumeia Zeghoud, Email: zsoumeia@gmail.com.
Asma Ben Amor, Email: 3licencegp@gmail.com.
Mourad Benzina, Email: mourad_benzina@yahoo.fr.
Ali Alnazza Alhamad, Email: che.alialnazaa@gmail.com.
References
- 1.López-Soto A, Gonzalez S, Smyth MJ, et al. Control of metastasis by NK cells. Cancer Cell 2017;32:135–154. [DOI] [PubMed] [Google Scholar]
- 2.Gallaher JA, Enriquez-Navas PM, Luddy KA, et al. Spatial heterogeneity and evolutionary dynamics modulate time to recurrence in continuous and adaptive cancer therapies. Cancer Res 2018;78:2127–2139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Zhang Y, Li M, Gao X, et al. Nanotechnology in cancer diagnosis: progress, challenges and opportunities. J Hematol Oncol 2019;12:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Cryer AM, Thorley AJ. Nanotechnology in the diagnosis and treatment of lung cancer. Pharmacol Ther 2019;198:189–205. [DOI] [PubMed] [Google Scholar]
- 5.Jardim GA, da Cruz EH, Valença WO, et al. Synthesis of selenium-quinone hybrid compounds with potential antitumor activity via Rh-catalyzed CH bond activation and click reactions. Molecules 2017;23:83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Chatterjee P, Kumar S. Current developments in nanotechnology for cancer treatment. Mater Today: Proc 2022;48:1754–1758. [Google Scholar]
- 7.Mohapatra S, Ranjan S, Dasgupta N, et al. Nanocarriers for drug delivery: nanoscience and nanotechnology in drug delivery. Elsevier; 2018. [Google Scholar]
- 8.Sun L, Liu H, Ye Y, et al. Smart nanoparticles for cancer therapy. Signal Transduct Target Ther 2023;8:418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zhou SK, Greenspan H, Davatzikos C, et al. A review of deep learning in medical imaging: Imaging traits, technology trends, case studies with progress highlights, and future promises. Proc IEEE 2021;109:820–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Weiss CR, Hafezi-Nejad N. Interventional radiology: past, present, and future. Radiol Soc North Am 2023;308:e230809; ed. [DOI] [PubMed] [Google Scholar]
- 11.Schiffman JD, Fisher PG, Gibbs P. Early detection of cancer: past, present, and future. Am Soc Clin Oncol Educ Book 2015;35:57–65. [DOI] [PubMed] [Google Scholar]
- 12.Runowicz CD, Leach CR, Henry NL, et al. American cancer society/American society of clinical oncology breast cancer survivorship care guideline. CA Cancer J Clin 2016;66:43–73. [DOI] [PubMed] [Google Scholar]
- 13.Alhamad AA, Zeghoud S, Amor IB, et al. AA short review of nanomaterials: synthesis methods, properties, and applications. Algerian J Chem Eng AJCE 2023;1:01–07. [Google Scholar]
- 14.Hsu JC, Nieves LM, Betzer O, et al. Nanoparticle contrast agents for X‐ray imaging applications. Wiley Interdiscipl Rev Nanomed Nanobiotechnol 2020;12:e1642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Tamandl D, Mang T, Ba-Ssalamah A. Imaging of colorectal cancer–the clue to individualized treatment. Innovative Surg Sci 2018;3:3–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Senapati S, Mahanta AK, Kumar S, et al. Controlled drug delivery vehicles for cancer treatment and their performance. Signal Transduct Target Ther 2018;3:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Baranwal J, Barse B, Di Petrillo A, et al. Nanoparticles in cancer diagnosis and treatment. Materials 2023;16:5354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Wang X, Li J, Zhang W, et al. Evaluating diabetic ketoacidosis via a MOF sensor for fluorescence imaging of phosphate and pH. Chem Commun 2022;58:3023–3026. [DOI] [PubMed] [Google Scholar]
- 19.Pavitra E, Dariya B, Srivani G, et al. Engineered nanoparticles for imaging and drug delivery in colorectal cancer. Semin Cancer Biol 2021;69:293–306. [DOI] [PubMed] [Google Scholar]
- 20.Teodor ED, Gatea F, Ficai A, et al. Functionalized magnetic nanostructures for anticancer therapy. Curr Drug Targets 2018;19:239–247. [DOI] [PubMed] [Google Scholar]
- 21.Zhai R, Gong X, Xie J, et al. Ultrasensitive analysis of heat shock protein 90α with antibodies orderly arrayed on a novel type of immunoprobe based on magnetic COFs. Talanta 2019;191:553–560. [DOI] [PubMed] [Google Scholar]
- 22.Huyan T, Li H, Peng H, et al. Extracellular vesicles–advanced nanocarriers in cancer therapy: progress and achievements. Int J Nanomedicine 2020;15:6485–6502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Jaishree V, Gupta P. Nanotechnology: a revolution in cancer diagnosis. Indian J Clin Biochem 2012;27:214–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Schröfel A, Kratošová G, Šafařík I, et al. Applications of biosynthesized metallic nanoparticles–a review. Acta Biomater 2014;10:4023–4042. [DOI] [PubMed] [Google Scholar]
- 25.Wang J, Cao F, He S, et al. FRET on lateral flow test strip to enhance sensitivity for detecting cancer biomarker. Talanta 2018;176:444–449. [DOI] [PubMed] [Google Scholar]
- 26.Aygun A, Gülbagca F, Ozer LY, et al. Biogenic platinum nanoparticles using black cumin seed and their potential usage as antimicrobial and anticancer agent. J Pharm Biomed Anal 2020;179:112961. [DOI] [PubMed] [Google Scholar]
- 27.Singh P, Pandit S, Mokkapati V, et al. Gold nanoparticles in diagnostics and therapeutics for human cancer. Int J Mol Sci 2018;19:1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Mieszawska AJ, Mulder WJ, Fayad ZA, et al. Multifunctional gold nanoparticles for diagnosis and therapy of disease. Mol Pharm 2013;10:831–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Yang Y, Yan Q, Liu Q, et al. An ultrasensitive sandwich-type electrochemical immunosensor based on the signal amplification strategy of echinoidea-shaped Au@ Ag-Cu2O nanoparticles for prostate specific antigen detection. Biosens Bioelectron 2018;99:450–457. [DOI] [PubMed] [Google Scholar]
- 30.Chakraborty D, Viveka TS, Arvind K, et al. A facile gold nanoparticle–based ELISA system for detection of osteopontin in saliva: towards oral cancer diagnostics. Clin Chim Acta 2018;477:166–172. [DOI] [PubMed] [Google Scholar]
- 31.Pan S, Jeevanandam J, Acquah C, et al. Drug delivery systems for cardiovascular ailments. Drug Delivery Devices and Therapeutic Systems. Elsevier; 2021:567–599. [Google Scholar]
- 32.Immanuel S, Aparna T, Sivasubramanian R. Graphene–metal oxide nanocomposite modified electrochemical sensors. Graphene-based electrochemical sensors for biomolecules. Elsevier; 2019:113–138. [Google Scholar]
- 33.Sardoiwala MN, Kaundal B, Choudhury SR. Development of engineered nanoparticles expediting diagnostic and therapeutic applications across blood–brain barrier. Handbook Nanomater Industr Appls 2018;696–709. [Google Scholar]
- 34.Gebre SH, Sendeku MG. New frontiers in the biosynthesis of metal oxide nanoparticles and their environmental applications: an overview. SN Appl Sci 2019;1:1–28. [Google Scholar]
- 35.Orel VE, Tselepi M, Mitrelias T, et al. Nanomagnetic modulation of tumor redox state. Nanomed: Nanotechnol Biol Med 2018;14:1249–1256. [DOI] [PubMed] [Google Scholar]
- 36.Khaled MTO, Zaater A, Amor IB, et al. Drug delivery methods based on nanotechnology for the treatment of eye diseases. Ann Med Surg, 85:6029–60402023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zeghoud S, Amor IB, Alhamad AA, et al. Osteoporosis therapy using nanoparticles: a review. Ann Med Surg 2024;86:284–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zhou J, Meli VS, Chen EY-T, et al. Magnetic resonance imaging of tumor-associated-macrophages (TAMs) with a nanoparticle contrast agent. RSC Adv 2022;12:7742–7756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Vilímová I, Chourpa I, David S, et al. Two-step formulation of magnetic nanoprobes for microRNA capture. RSC Adv 2022;12:7179–7188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Amor IB, Hemmami H, Laouini SE, et al. Biosynthesis MgO and ZnO nanoparticles using chitosan extracted from Pimelia Payraudi Latreille for antibacterial applications,. World J Microbiol Biotechnol 2023;39:19. [DOI] [PubMed] [Google Scholar]
- 41.Sankaranarayanan S, Hariram M, Vivekanandhan S, et al. Biosynthesized transition metal oxide nanostructures for photocatalytic degradation of organic dyes. Green Functionalized Nanomaterials for Environmental Applications. Elsevier; 2022:417–460. [Google Scholar]
- 42.Shamsipur M, Emami M, Farzin L, et al. A sandwich-type electrochemical immunosensor based on in situ silver deposition for determination of serum level of HER2 in breast cancer patients. Biosens Bioelectron 2018;103:54–61. [DOI] [PubMed] [Google Scholar]
- 43.Ren L, Wang L, Rehberg M, et al. Applications and immunological effects of quantum dots on respiratory system. Front Immunol 2022;12:795232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Xu Y-M, Tan HW, Zheng W, et al. Cadmium telluride quantum dot-exposed human bronchial epithelial cells: a further study of the cellular response by proteomics,. Toxicol Res 2019;8:994–1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Abbasi E, Akbarzadeh A, Kouhi M, et al. Graphene: synthesis, bio-applications, and properties. Artific Cells, Nanomed Biotechnol 2016;44:150–156. [DOI] [PubMed] [Google Scholar]
- 46.Lee XJ, Lim HN, Rahman MBA, et al. Functionalization of graphene for nanodelivery of drugs. Synthesis, Technology and Applications of Carbon Nanomaterials. Elsevier; 2019:157–176. [Google Scholar]
- 47.Sabry F. Fullerene: Building nano-sized machines which can be inserted into the human body to detect and repair diseased cells for cancer and AIDS. One Billion Knowledgeable; 2022. vol. 7. [Google Scholar]
- 48.Mao C-C, Cai X. Nanomaterials and aging. Curr Stem Cell Res Ther 2021;16:57–65. [DOI] [PubMed] [Google Scholar]
- 49.Amreddy N, Ahmed RA, Munshi A, et al. Tumor-targeted dendrimer nanoparticles for combinatorial delivery of siRNA and chemotherapy for cancer treatment. Drug Deliv Syst 2020;2059:167–189. [DOI] [PubMed] [Google Scholar]
- 50.Lotfipanah S, Zeinali M, Yaghmaei P. Induction of caspase-2 gene expression in carboxyl-functionalized carbon nanotube-treated human T-cell leukemia (Jurkat) cell line. Drug Chem Toxicol 2021;44:394–399. [DOI] [PubMed] [Google Scholar]
- 51.Shaki H, Raissi H, Mollania F, et al. Modeling the interaction between anti-cancer drug penicillamine and pristine and functionalized carbon nanotubes for medical applications: Density functional theory investigation and a molecular dynamics simulation. J Biomol Struct Dyn 2020;38:1322–1334. [DOI] [PubMed] [Google Scholar]
- 52.Dong Y-D, Tchung E, Nowell C, et al. Microfluidic preparation of drug-loaded PEGylated liposomes, and the impact of liposome size on tumour retention and penetration. J Liposome Res 2019;29:1–9. [DOI] [PubMed] [Google Scholar]
- 53.Alrushaid N, Khan FA, Al-Suhaimi EA, et al. Nanotechnology in cancer diagnosis and treatment. Pharmaceutics 2023;15:1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zhang M, June S, Long TE. Principles of step-growth polymerization (polycondensation and polyaddition). Polymer Science: A Comprehensive Reference. Elsevier; 2012:7–47; 10 Volume Set. [Google Scholar]
- 55.Cho H-Y, Hossain MK, Lee J-H, et al. Selective isolation and noninvasive analysis of circulating cancer stem cells through Raman imaging. Biosens Bioelectron 2018;102:372–382. [DOI] [PubMed] [Google Scholar]
- 56.Odiba A, Ottah V, Ottah C, et al. Therapeutic nanomedicine surmounts the limitations of pharmacotherapy. Open Med 2017;12:271–287. [Google Scholar]
- 57.Arms L, Smith DW, Flynn J, et al. Advantages and limitations of current techniques for analyzing the biodistribution of nanoparticles. Front Pharmacol 2018;9:802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Hlaing CB, Chariyakornkul A, Pilapong C, et al. Assessment of systemic toxicity, genotoxicity, and early phase hepatocarcinogenicity of iron (III)-tannic acid nanoparticles in rats. Nanomaterials 2022;12:1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Liu Y, Peng N, Yao Y, et al. Breaking the nanoparticle’s dispersible limit via rotatable surface ligands,. Nat Commun 2022;13:3581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kim IY, Kwak M, Kim J, et al. Comparative study on nanotoxicity in human primary and cancer cells. Nanomaterials 2022;12:993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Salem SS, Fouda A. Green synthesis of metallic nanoparticles and their prospective biotechnological applications: an overview. Biol Trace Elem Res 2021;199:344–370. [DOI] [PubMed] [Google Scholar]
- 62.Arami H, Teeman E, Troksa A, et al. Tomographic magnetic particle imaging of cancer targeted nanoparticles. Nanoscale 2017;9:18723–18730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Oddone N, Lecot N, Fernández M, et al. In vitro and in vivo uptake studies of PAMAM G4. 5 dendrimers in breast cancer. J Nanobiotechnology 2016;14:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Yildiz T, Gu R, Zauscher S, et al. Doxorubicin-loaded protease-activated near-infrared fluorescent polymeric nanoparticles for imaging and therapy of cancer. Int J Nanomedicine 2018;13:6961–6986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Afzal M, Ameeduzzafar KS, Alharbi KS, et al. Nanomedicine in treatment of breast cancer–A challenge to conventional therapy. Semin Cancer Biol 2021;69:279–292. [DOI] [PubMed] [Google Scholar]
- 66.Liyanage PY, Hettiarachchi SD, Zhou Y, et al. Nanoparticle-mediated targeted drug delivery for breast cancer treatment. Biochim Biophys Acta (BBA)-Rev Cancer 2019;1871:419–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Herdiana Y, Wathoni N, Shamsuddin S, et al. Chitosan-based nanoparticles of targeted drug delivery system in breast cancer treatment. Polymers 2021;13:1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Mirza Z, Karim S. Nanoparticles-based drug delivery and gene therapy for breast cancer: recent advancements and future challenges. Semin Cancer Biol 2021;69:226–237. [DOI] [PubMed] [Google Scholar]
- 69.Sun L, Wu Q, Peng F, et al. Strategies of polymeric nanoparticles for enhanced internalization in cancer therapy. Colloids Surfaces B: Biointerfaces 2015;135:56–72. [DOI] [PubMed] [Google Scholar]
- 70.Truffi M, Mazzucchelli S, Bonizzi A, et al. Nano-strategies to target breast cancer-associated fibroblasts: rearranging the tumor microenvironment to achieve antitumor efficacy. Int J Mol Sci 2019;20:1263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Allahverdiyev AM, Parlar E, Dinparvar S, et al. Current aspects in treatment of breast cancer based of nanodrug delivery systems and future prospects. Artific Cells Nanomed Biotechnol 2018;46:755–762. [DOI] [PubMed] [Google Scholar]
- 72.Chow JC. Application of nanomaterials in biomedical imaging and cancer therapy. Nanomaterials 2022;12:726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Mundekkad D, Cho WC. Nanoparticles in clinical translation for cancer therapy. Int J Mol Sci 2022;23:1685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Yao Y, Zhou Y, Liu L, et al. Nanoparticle-based drug delivery in cancer therapy and its role in overcoming drug resistance. Front Mol Biosci 2020;7:193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Hong G, Robinson JT, Zhang Y, et al. In vivo fluorescence imaging with Ag2S quantum dots in the second near‐infrared region. Angewandte Chem Int Ed 2012;51:9818–9821. [DOI] [PubMed] [Google Scholar]
- 76.Lai S-F, Ko B-H, Chien C-C, et al. Gold nanoparticles as multimodality imaging agents for brain gliomas. J Nanobiotechnology 2015;13:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Nam T, Park S, Lee S-Y, et al. Tumor targeting chitosan nanoparticles for dual-modality optical/MR cancer imaging. Bioconjug Chem 2010;21:578–582. [DOI] [PubMed] [Google Scholar]
- 78.Stylianopoulos T, Economides E-A, Baish JW, et al. Towards optimal design of cancer nanomedicines: Multi-stage nanoparticles for the treatment of solid tumors. Ann Biomed Eng 2015;43:2291–2300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Lundqvist M, Stigler J, Cedervall T, et al. The evolution of the protein corona around nanoparticles: a test study. ACS Nano 2011;5:7503–7509. [DOI] [PubMed] [Google Scholar]
- 80.Garrigue P, Tang J, Ding L, et al. Self-assembling supramolecular dendrimer nanosystem for PET imaging of tumors. Proc Natl Acad Sci 2018;115:11454–11459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Bertrand N, Wu J, Xu X, et al. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev 2014;66:2–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Yang Y, Chen Q, Li S, et al. iRGD-mediated and enzyme-induced precise targeting and retention of gold nanoparticles for the enhanced imaging and treatment of breast cancer. J Biomed Nanotechnol 2018;14:1396–1408. [DOI] [PubMed] [Google Scholar]
- 83.He Z, Wang Q, Zhang N, et al. Gold nanorods/tetrahedral DNA composites for chemo-photothermal therapy. Regen Biomater 2022;9:rbac032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ekinci M, Santos-Oliveira R, Ilem-Ozdemir D. Biodistribution of 99mTc-PLA/PVA/Atezolizumab nanoparticles for non-small cell lung cancer diagnosis. Eur J Pharm Biopharm 2022;176:21–31. [DOI] [PubMed] [Google Scholar]
- 85.Ansari MT, Ramlan TA, Jamaluddin NN, et al. Lipid-based nanocarriers for cancer and tumor treatment. Curr Pharm Des 2020;26:4272–4276. [DOI] [PubMed] [Google Scholar]
- 86.Chattha GM, Arshad S, Kamal Y, et al. Nanorobots: an innovative approach for DNA-based cancer treatment. J Drug Deliv Sci Technol 2023;80:104173. [Google Scholar]
- 87.Hwang R, Chang CH, Zhu Y, et al. Biotransformation and potential adverse effects of rare earth oxide nanoparticles. Nanotechnol Character Tools Environ Health Safety 2019;47–63. [Google Scholar]
- 88.Liu Y, Zhang H, Cui H, et al. Combined and targeted drugs delivery system for colorectal cancer treatment: Conatumumab decorated, reactive oxygen species sensitive irinotecan prodrug and quercetin co-loaded nanostructured lipid carriers,. Drug Deliv 2022;29:342–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Lv S, Wu Y, Cai K, et al. High drug loading and sub-quantitative loading efficiency of polymeric micelles driven by donor–receptor coordination interactions. J Am Chem Soc 2018;140:1235–1238. [DOI] [PubMed] [Google Scholar]
- 90.Culver HR, Clegg JR, Peppas NA. Analyte-responsive hydrogels: intelligent materials for biosensing and drug delivery. Acc Chem Res 2017;50:170–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wu J, Zhang Z, Zhou W, et al. Mechanism of a long-term controlled drug release system based on simple blended electrospun fibers. J Control Release 2020;320:337–346. [DOI] [PubMed] [Google Scholar]
- 92.Sanoff HK, Moon DH, Moore DT, et al. Phase I/II trial of nano-camptothecin CRLX101 with capecitabine and radiotherapy as neoadjuvant treatment for locally advanced rectal cancer,. Nanomed: Nanotechnol Biol Med 2019;18:189–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Not applicable.
