Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
Int J Mol Sci ; 25(4)2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38396904

ABSTRACT

Accurately characterizing DNA double-stranded breaks (DSBs) and understanding the DNA damage response (DDR) is crucial for assessing cellular genotoxicity, maintaining genomic integrity, and advancing gene editing technologies. Immunofluorescence-based techniques have proven to be invaluable for quantifying and visualizing DSB repair, providing valuable insights into cellular repair processes. However, the selection of appropriate markers for analysis can be challenging due to the intricate nature of DSB repair mechanisms, often leading to ambiguous interpretations. This comprehensively summarizes the significance of immunofluorescence-based techniques, with their capacity for spatiotemporal visualization, in elucidating complex DDR processes. By evaluating the strengths and limitations of different markers, we identify where they are most relevant chronologically from DSB detection to repair, better contextualizing what each assay represents at a molecular level. This is valuable for identifying biases associated with each assay and facilitates accurate data interpretation. This review aims to improve the precision of DSB quantification, deepen the understanding of DDR processes, assay biases, and pathway choices, and provide practical guidance on marker selection. Each assay offers a unique perspective of the underlying processes, underscoring the need to select markers that are best suited to specific research objectives.


Subject(s)
DNA Breaks, Double-Stranded , DNA Damage , DNA/metabolism , DNA Repair , DNA End-Joining Repair
2.
J Biomed Sci ; 31(1): 1, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38163894

ABSTRACT

BACKGROUND: As of 2020, hepatocellular carcinoma (HCC), a form of liver cancer, stood as the third most prominent contributor to global cancer-related mortality. Combining immune checkpoint inhibitors (ICI) with other therapies has shown promising results for treating unresectable HCC, offering new opportunities. Recombinant adeno-associated viral type 2 (AAV2) virotherapy has been approved for clinical use but it efficacy is stifled through systemic administration. On the other hand, iron oxide nanoparticles (ION) can be cleared via the liver and enhance macrophage polarization, promoting infiltration of CD8+ T cells and creating a more favorable tumor microenvironment for immunotherapy. METHODS: To enhance the efficacy of virotherapy and promote macrophage polarization towards the M1-type in the liver, ION-AAV2 were prepared through the coupling of ION-carboxyl and AAV2-amine using 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC)/N-hydroxysulfosuccinimide (Sulfo-NHS). Efficacy after systemic delivery of ION-AAV2 in an orthotopic HCC model was evaluated. RESULTS: After 28 days, the tumor weight in mice treated with ION-AAV2 was significantly reduced by 0.56-fold compared to the control group. The ION-AAV2 treatment led to an approximate 1.80-fold increase in the level of tumor associated M1-type macrophages, while the number of M2-type macrophages was reduced by 0.88-fold. Moreover, a proinflammatory response increased the population of tumor-infiltrating CD8+ T cells in the ION-AAV2 group. This transformation converted cold tumors into hot tumors. CONCLUSIONS: Our findings suggest that the conjugation of ION with AAV2 could be utilized in virotherapy while simultaneously exploiting macrophage-modulating cancer immunotherapies to effectively suppress HCC growth.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Liver Neoplasms/drug therapy , Carcinoma, Hepatocellular/drug therapy , CD8-Positive T-Lymphocytes , Tumor Microenvironment , Dependovirus , Cell Line, Tumor
3.
Pharmacol Ther ; 250: 108521, 2023 10.
Article in English | MEDLINE | ID: mdl-37657673

ABSTRACT

In 2015, oncolytic virotherapy was approved for clinical use, and in 2017, recombinant adeno-associated virus (AAV) delivery was also approved. However, systemic administration remains challenging due to the limited number of viruses that successfully reach the target site. Although the US Food and Drug Administration (FDA) permits the use of higher doses of AAV to achieve greater rates of transduction, most AAV still accumulates in the liver, potentially leading to toxicity there and elsewhere. Targeting the tumor microenvironment is a promising strategy for cancer treatment due to the critical role of the tumor microenvironment in controlling tumor progression and influencing the response to therapies. Newly discovered evidence indicates that administration routes focusing on the tumor microenvironment can promote delivery specificity and transduction efficacy within the tumor. Here, we review approaches that involve modifying viral surface features, modulating the immune system, and targeting the physicochemical characteristics in tumor microenvironment to regulate therapeutic delivery. Targeting tumor acidosis presents advantages that can be leveraged to enhance virotherapy outcomes and to develop new therapeutic approaches that can be integrated with standard treatments.


Subject(s)
Neoplasms , Oncolytic Virotherapy , Humans , Tumor Microenvironment , Neoplasms/therapy , Neoplasms/pathology , Dependovirus
4.
Antioxidants (Basel) ; 12(8)2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37627612

ABSTRACT

Nanoparticle-based therapeutics are being clinically translated for treating cancer. Even when thought to be biocompatible, nanoparticles are being increasingly identified as altering cell regulation and homeostasis. Antioxidant pathways are important for maintaining cell redox homeostasis and play important roles by maintaining ROS levels within tolerable ranges. Here, we sought to understand how a model of a relatively inert nanoparticle without any therapeutic agent itself could antagonize a cancer cell lines' antioxidant mechanism. A label-free protein expression approach was used to assess the glutathione-thioredoxin antioxidative pathway in a prostate cancer cell line (PC-3) after exposure to gold nanoparticles conjugated with a targeting moiety (transferrin). The impact of the nanoparticles was also corroborated through morphological analysis with TEM and classification of pro-apoptotic cells by way of the sub-G0/G1 population via the cell cycle and annexin V apoptosis assay. After a two-hour exposure to nanoparticles, major proteins associated with the glutathione-thioredoxin antioxidant pathway were downregulated. However, this response was acute, and in terms of protein expression, cells quickly recovered within 24 h once nanoparticle exposure ceased. The impact on PRDX-family proteins appears as the most influential factor in how these nanoparticles induced an oxidative stress response in the PC-3 cells. An apparent adaptive response was observed if exposure to nanoparticles continued. Acute exposure was observed to have a detrimental effect on cell viability compared to continuously exposed cells. Nanoparticle effects on cell regulation likely provide a compounding therapeutic advantage under some circumstances, in addition to the action of any cytotoxic agents; however, any therapeutic advantage offered by nanoparticles themselves with regard to vulnerabilities specific to the glutathione-thioredoxin antioxidative pathway is highly temporal.

5.
Phys Eng Sci Med ; 46(2): 529-560, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37160539

ABSTRACT

Particle therapies are becoming increasingly available clinically due to their beneficial energy deposition profile, sparing healthy tissues. This may be further promoted with ultra-high dose rates, termed FLASH. This review comprehensively summarises current knowledge based on studies relevant to proton- and carbon-FLASH therapy. As electron-FLASH literature presents important radiobiological findings that form the basis of proton and carbon-based FLASH studies, a summary of key electron-FLASH papers is also included. Preclinical data suggest three key mechanisms by which proton and carbon-FLASH are able to reduce normal tissue toxicities compared to conventional dose rates, with equipotent, or enhanced, tumour kill efficacy. However, a degree of caution is needed in clinically translating these findings as: most studies use transmission and do not conform the Bragg peak to tumour volume; mechanistic understanding is still in its infancy; stringent verification of dosimetry is rarely provided; biological assays are prone to limitations which need greater acknowledgement.


Subject(s)
Biological Assay , Protons , Carbon/therapeutic use , Electrons , Health Status
6.
Methods Mol Biol ; 2644: 15-33, 2023.
Article in English | MEDLINE | ID: mdl-37142913

ABSTRACT

The MTT assay is extensively used, most often to infer a measure of cytotoxicity of treatments to cells. As with any assay though, there are a number of limitations. The method described here is designed with consideration of how the MTT assay fundamentally works to account for, or at least identify, confounding factors in measurements. It also provides a decision-making framework to best interpret and complement the MTT assay to apply it as either a measure of metabolic activity or cell viability.


Subject(s)
Research Design , Tetrazolium Salts/metabolism , Cell Survival
7.
ACS Appl Bio Mater ; 2022 Aug 26.
Article in English | MEDLINE | ID: mdl-36027561

ABSTRACT

Nanoparticle (NP) internalization by cells is complex, highly heterogeneous, and fundamentally important for nanomedicine. We report powerful probabilistic statistics from single-cell data on quantitative NP uptake of PEG-coated transferrin receptor-targeted gold NPs for cancer-derived and fibroblast cells according to their cell size, receptor expression, and receptor density. The smaller cancer cells had a greater receptor density and more efficient uptake of targeted NPs. However, simply due to fibroblasts being larger with more receptors, they exhibited greater NP uptake. While highly heterogeneous, targeted NP uptake strongly correlated with receptor expression. When uptake was normalized to cell size, no correlation existed. Consequently, skewed population distributions in cell sizes explain the distribution in NP uptake. Furthermore, exposure to the transferrin receptor-targeted NPs alters the fibroblast size and receptor expression, suggesting that the receptor-targeted NPs may interfere with the metabolic flux and nutrient exchange, which could assist in explaining the altered regulation of cells exposed to nanoparticles.

9.
Int J Mol Sci ; 23(9)2022 Apr 24.
Article in English | MEDLINE | ID: mdl-35563097

ABSTRACT

Aluminium (Al) compounds are used as adjuvants in human and veterinary prophylactic vaccines due to their improved tolerability compared to other adjuvants. These Al-based adjuvants form microparticles (MPs) of heterogeneous sizes ranging from ~0.5 to 10 µm and generally induce type 2 (Th2)-biased immune responses. However, recent literature indicates that moving from micron dimension particles toward the nanoscale can modify the adjuvanticity of Al towards type 1 (Th1) responses, which can potentially be exploited for the development of vaccines for which Th1 immunity is crucial. Specifically, in the context of cancer treatments, Al nanoparticles (Al-NPs) can induce a more balanced (Th1/Th2), robust, and durable immune response associated with an increased number of cytotoxic T cells compared to Al-MPs, which are more favourable for stimulating an oncolytic response. In this review, we compare the adjuvant properties of Al-NPs to those of Al-MPs in the context of infectious disease vaccines and cancer immunotherapy and provide perspectives for future research.


Subject(s)
Nanoparticles , Vaccines , Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic , Aluminum , Humans
10.
Int J Mol Sci ; 23(9)2022 Apr 29.
Article in English | MEDLINE | ID: mdl-35563326

ABSTRACT

Radiotherapy is a highly affordable treatment and provides many excellent outcomes [...].


Subject(s)
Nanoparticles , Radiation-Sensitizing Agents , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/therapeutic use
11.
J Control Release ; 346: 98-109, 2022 06.
Article in English | MEDLINE | ID: mdl-35447296

ABSTRACT

Preclinical, clinical and epidemiologic studies have established the potent anticancer and radiosensitisation effects of HMG-CoA reductase inhibitors (statins). However, the low bioavailability of oral statin formulations is a key barrier to achieving effective doses within tumour. To address this issue and ascertain the radiosensitisation potential of simvastatin, we developed a parenteral high density lipoprotein nanoparticle (HDL NP) formulation of this commonly used statin. A scalable method for the preparation of the simvastatin-HDL NPs was developed using a 3D printed microfluidic mixer. This enables the production of litre scale amounts of particles with minimal batch to batch variation. Simvastatin-HDL NPs enhanced the radiobiological response in 2D/3D head and neck squamous cell carcinoma (HNSCC) in vitro models. The simvastatin-HDL NPs radiosensitisation was comparable to that of 10 and 5 times higher doses of free drug in 2D and 3D cultures, respectively, which could be partially explained by more efficient cellular uptake of the statin in the nanoformulation as well as by the inherent biological activity of the HDL NPs on the cholesterol pathway. The radiosensitising potency of the simvastatin-HDL nanoformulation was validated in an immunocompetent MOC-1 HNSCC tumour bearing mouse model. This data supports the rationale of repurposing statins through reformulation within HDL NPs. Statins are safe and readily available molecules including as generic, and their use as radiosensitisers could lead to much needed effective and affordable approaches to improve treatment of solid tumours.


Subject(s)
Head and Neck Neoplasms , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Animals , Cholesterol, HDL , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lipoproteins, HDL , Mice , Simvastatin/pharmacology , Simvastatin/therapeutic use , Squamous Cell Carcinoma of Head and Neck/drug therapy
12.
Int J Mol Sci ; 22(23)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34884632

ABSTRACT

The MTT assay for cellular metabolic activity is almost ubiquitous to studies of cell toxicity; however, it is commonly applied and interpreted erroneously. We investigated the applicability and limitations of the MTT assay in representing treatment toxicity, cell viability, and metabolic activity. We evaluated the effect of potential confounding variables on the MTT assay measurements on a prostate cancer cell line (PC-3) including cell seeding number, MTT concentration, MTT incubation time, serum starvation, cell culture media composition, released intracellular contents (cell lysate and secretome), and extrusion of formazan to the extracellular space. We also assessed the confounding effect of polyethylene glycol (PEG)-coated gold nanoparticles (Au-NPs) as a tested treatment in PC-3 cells on the assay measurements. We additionally evaluated the applicability of microscopic image cytometry as a tool for measuring intracellular MTT reduction at the single-cell level. Our findings show that the assay measurements are a result of a complicated process dependant on many of the above-mentioned factors, and therefore, optimization of the assay and rational interpretation of the data is necessary to prevent misleading conclusions on variables such as cell viability, treatment toxicity, and/or cell metabolism. We conclude, with recommendations on how to apply the assay and a perspective on where the utility of the assay is a powerful tool, but likewise where it has limitations.


Subject(s)
Culture Media/pharmacology , Gold/chemistry , Metal Nanoparticles/administration & dosage , Prostatic Neoplasms/pathology , Secretome , Single-Cell Analysis/methods , Cell Count , Cell Survival , Humans , Male , Metal Nanoparticles/chemistry , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured
13.
Drug Discov Today ; 26(11): 2508-2514, 2021 11.
Article in English | MEDLINE | ID: mdl-34325010

ABSTRACT

Targeted-therapy failure in treating nonsmall cell lung cancer (NSCLC) frequently occurs because of the emergence of drug resistance and genetic mutations. The same mutations also result in aerobic glycolysis, which further antagonizes outcomes by localized increases in lactate, an immune suppressor. Recent evidence indicates that enzymatic lowering of lactate can promote an oncolytic immune microenvironment within the tumour. Here, we review factors relating to lactate expression in NSCLC and the utility of lactate oxidase (LOX) for governing therapeutic delivery, its role in lactate oxidation and turnover, and relationships between lactate depletion and immune cell populations. The lactate-rich characteristic of NSCLC provides an exploitable property to potentially improve NSCLC outcomes and design new therapeutic strategies to integrate with conventional therapies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lactic Acid/metabolism , Lung Neoplasms/metabolism , Tumor Microenvironment , Anaplastic Lymphoma Kinase/genetics , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Citric Acid Cycle , Drug Resistance, Neoplasm , Genes, erbB-1/genetics , Glucose/metabolism , Humans , Immune Checkpoint Inhibitors/therapeutic use , L-Lactate Dehydrogenase/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Metabolic Networks and Pathways , Mixed Function Oxygenases/therapeutic use , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics
14.
Med Phys ; 48(6): 3120-3133, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33818799

ABSTRACT

PURPOSE: Intrinsic radioresistance and increased proliferation rates in head and neck cancers (HNCs) are associated with negative radiotherapy (RT) treatment responses. The use of gold nanoparticles (AuNPs) as radiosensitizers could enable total radiation dose reduction and lowered radiation toxicity. AuNP radiosensitization may overcome hypoxia-induced radioresistance and treatment-induced accelerated repopulation of cancer cells in HNCs, improving radiotherapy outcomes. METHODS: Tumor control was determined by considering individual cancer cell responses in probabilistic computational simulations using HYP-RT software for clinical radiotherapy doses and fractionation schedules along with three different nanoparticle administration schedules. Antagonistic tumor hypoxia and rapid tumor regrowth due to accelerated repopulation of cancers cells were taken into consideration. RESULTS: Simulations indicate that tumors that are conventionally uncontrollable can be controlled with AuNP radiosensitization. In simulations where the absence of AuNPs required radiotherapy doses above standard clinical prescriptions, reoccurring AuNP administration allowed for radiation dose reductions below standard clinical dose prescriptions. For example, considering a 2 Gy per fraction radiotherapy schedule, tumor control was achieved with 57.2 ± 5.1 Gy (P = <0.0001) for weekly AuNP administration and 53.0 ± 4.0 Gy (P = <0.0001) for biweekly AuNP administration compared to 69.9 ± 5.8 Gy with no radiosensitization. CONCLUSIONS: AuNPs decreased the predicted RT total doses required to achieve tumor control via total stem cell elimination, offering an optimistic prediction and method for which hypoxia-induced and rapidly growing radioresistant tumors are treated more effectively. Outcomes are also shown to be sensitive to the RT schedule with data for hyperfractionated RT indicating the greatest benefits from radiosensitization.


Subject(s)
Head and Neck Neoplasms , Metal Nanoparticles , Dose Fractionation, Radiation , Gold , Head and Neck Neoplasms/radiotherapy , Humans , Hypoxia
15.
Article in English | MEDLINE | ID: mdl-32686321

ABSTRACT

Metal-based nanoparticles applied to potentiating the effects of radiotherapy have drawn significant attention from the research community and are now available clinically. By improving our mechanistic understanding, nanoparticles are likely to evolve to provide very significant improvements in radiotherapy outcomes with only incremental increase in cost. This review critically assesses the inconsistent observations surrounding physical, physicochemical, chemical and biological mechanisms of radiosensitization. In doing so, a number of needs are identified for continuing research and are highlighted. The large degree of variability from one nanoparticle to another emphasizes that it is a mistake to generalize nanoparticle radiosensitizer mechanisms. Nanoparticle formulations should be considered in an analogous way as pharmacological agents and as a broad class of therapeutic agents, needing to be considered with a high degree of individuality with respect to their interactions and ultimate impact on radiobiological response. In the same way that no universal anti-cancer drug exists, it is unlikely that a single nanoparticle formulation will lead to the best therapeutic outcomes for all cancers. The high degree of complexity and variability in mechanistic action provides notable opportunities for nanoparticle formulations to be optimized for specific indications. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.


Subject(s)
Metal Nanoparticles , Nanoparticles , Neoplasms , Radiation-Sensitizing Agents , Drug Discovery , Humans , Neoplasms/drug therapy , Neoplasms/radiotherapy
16.
Nanomedicine (Lond) ; 15(29): 2837-2850, 2020 12.
Article in English | MEDLINE | ID: mdl-33252312

ABSTRACT

Background: The use of gold nanoparticles (AuNPs) as radiosensitizers may offer a new approach in the treatment of head and neck cancers; minimizing treatment-associated toxicities and improving patient outcomes. AuNPs promote localized dose deposition; permitting improved local control and/or dose reduction. Aim: This work aimed to address the theoretical optimization of radiation doses, fractionation and nanoparticle injection schedules to maximize therapeutic benefits. Materials & methods: Probabilistic nanoparticle sensitization factors were incorporated into the individual cell-based HYP-RT computer model of tumor growth and radiotherapy. Results: Total dose outcomes across all radiation therapy treatment regimens were found to be significantly reduced with the presence of AuNPs, with bi-weekly injections showing the most decrease. Conclusion: Outcomes suggest the need for regular AuNP administration to permit effective radiosensitization.


Subject(s)
Head and Neck Neoplasms , Metal Nanoparticles , Radiation-Sensitizing Agents , Computer Simulation , Gold , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Humans
17.
Nanomedicine ; 29: 102243, 2020 10.
Article in English | MEDLINE | ID: mdl-32623018

ABSTRACT

Many tumors develop resistance to most of the apoptosis-based cancer therapies. In this sense targeting non-apoptotic forms of cell death including necroptosis, autophagy and ferroptosis may have therapeutic benefits in apoptosis-defective cancer cells. Nanomaterials have shown great advantages in cancer treatment owing to their unique characteristics. Besides, the capability of nanomaterials to induce different forms of cell death has gained widespread attention in cancer treatment. Reports in this field reflect the therapeutic potential of necroptotic cell death induced by nanomaterials in cancer. Also, autophagic cell death induced by nanomaterials alone and as a part of chemo-, radio- and photothermal therapy holds great promise as anticancer therapeutic option. Besides, ferroptosis induction by iron-based nanomaterials in drug delivery, immunotherapy, hyperthermia and imaging systems shows promising results in malignancies. Hence, this review is devoted to the latest efforts and the challenges in this field of research and its clinical merits.


Subject(s)
Cell Death/drug effects , Nanostructures/therapeutic use , Necroptosis/drug effects , Neoplasms/drug therapy , Apoptosis/genetics , Autophagy/drug effects , Autophagy/genetics , Cell Death/genetics , Ferroptosis/drug effects , Ferroptosis/genetics , Humans , Necroptosis/genetics , Neoplasms/genetics , Neoplasms/pathology
18.
Int J Mol Sci ; 21(12)2020 Jun 22.
Article in English | MEDLINE | ID: mdl-32580352

ABSTRACT

Gold nanoparticle (GNP) enhanced proton therapy is a promising treatment concept offering increased therapeutic effect. It has been demonstrated in experiments which provided indications that reactive species play a major role. Simulations of the radiolysis yield from GNPs within a cell model were performed using the Geant4 toolkit. The effect of GNP cluster size, distribution and number, cell and nuclear membrane absorption and intercellular yields were evaluated. It was found that clusters distributed near the nucleus increased the nucleus yield by 91% while reducing the cytoplasm yield by 7% relative to a disperse distribution. Smaller cluster sizes increased the yield, 200 nm clusters had nucleus and cytoplasm yields 117% and 35% greater than 500 nm clusters. Nuclear membrane absorption reduced the cytoplasm and nucleus yields by 8% and 35% respectively to a permeable membrane. Intercellular enhancement was negligible. Smaller GNP clusters delivered near sub-cellular targets maximise radiosensitisation. Nuclear membrane absorption reduces the nucleus yield, but can damage the membrane providing another potential pathway for biological effect. The minimal effect on adjacent cells demonstrates that GNPs provide a targeted enhancement for proton therapy, only effecting cells with GNPs internalised. The provided quantitative data will aid further experiments and clinical trials.


Subject(s)
Cells/radiation effects , Gold/chemistry , Metal Nanoparticles/chemistry , Models, Biological , Proton Therapy , Pulse Radiolysis , Radiation-Sensitizing Agents/chemistry , Monte Carlo Method
19.
J Control Release ; 324: 482-492, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32497570

ABSTRACT

Lung cancer is the primary cause of cancer-related death worldwide. 85%-90% of cases are non-small cell lung cancer (NSCLC) which characteristically exhibits altered epidermal growth factor receptor (EGFR) signaling is a major driver pathway. Unfortunately, therapeutic outcomes in treating NSCLC are compromised by the emergence of drug resistance in response to EGFR-tyrosine kinase inhibitor (TKI) targeted therapy due to the acquired resistance mutation EGFR T790M or activation of alternative pathways. There is current need for a new generation of TKIs to be developed to treat EGFR-TKI-resistant NSCLC. To overcome the above problems and improve clinical efficacy, nanotechnology with targeting abilities and sustained release has been proposed for EGFR-TKI-resistant NSCLC treatment and has already achieved success in in vitro or in vivo models. In this review, we summarize and illustrate representative nano-formulations targeting EGFR-TKI-resistant NSCLC. The described advances may pave the way to better understanding and design of nanocarriers and multifunctional nanosystems for efficient treatment for drug resistant NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm , ErbB Receptors/genetics , Humans , Lung Neoplasms/drug therapy , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
20.
Int J Mol Sci ; 21(8)2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32326054

ABSTRACT

Radiotherapy is a highly multidisciplinary field with respect to its foundations of research and development, and in its clinical utility [...].


Subject(s)
Nanoparticles , Radiation-Sensitizing Agents/administration & dosage , Radiation-Sensitizing Agents/chemistry , Animals , Humans , Radiotherapy/methods , Radiotherapy/standards
SELECTION OF CITATIONS
SEARCH DETAIL
...