Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 331
Filter
1.
J Pharmacol Toxicol Methods ; 127: 107519, 2024.
Article in English | MEDLINE | ID: mdl-38797368

ABSTRACT

Thymoquinone (TH) has been one of the major phytochemical used in the treatment of cancers since long time, especially in the management of glioblastoma multiforme (GBM). The formulation of lipo-polymeric nanoshells (LPNs) and their nasal delivery are fascinating approaches for overcoming the drawbacks of low solubility and poor bioavailability of TH. Hence targeting LPNs to the brain requires a validated bioanalytical method for the assessment of TH concentration in Cerebrospinal fluid (CSF) and brain tissue homogenates (BTH). Therefore, the current work focuses on the development and validation of high-performance liquid chromatography (HPLC) method in CSF by employing nasal simulated fluid (NSF) as one of the major components of the mobile phase. The developed method was checked for linearity in the range of 0.05 to 1.6 µg/mL, having an r2 value of 0.999 with mean % recovery >95% and % RSD values below <2.0%. The developed method gave a clear separation of TH at 6.021 ± 0.17 min with an internal standard at 4.102 ± 0.09 min and a CSF spike at 2.170 ± 0.12 min. The developed method assisted in determining the in-vitro and in-vivo drug release study of LPNs, pharmacokinetic profiling, qualitative in-vivo brain uptake study, in-vitro cellular uptake, and generating stability data of formulated LPNs proposed for intranasal administration in rats.


Subject(s)
Administration, Intranasal , Benzoquinones , Brain , Nanoshells , Animals , Benzoquinones/pharmacokinetics , Benzoquinones/administration & dosage , Benzoquinones/cerebrospinal fluid , Benzoquinones/chemistry , Rats , Chromatography, High Pressure Liquid/methods , Brain/metabolism , Male , Nanoshells/chemistry , Rats, Wistar , Biological Availability
2.
ACS Appl Mater Interfaces ; 16(20): 25836-25842, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38728653

ABSTRACT

We demonstrate the use of DNA origami to create virus-trapping nanoshells that efficiently neutralize hepatitis B virus (HBV) in cell culture. By modification of the shells with a synthetic monoclonal antibody that binds to the HBV envelope, the effective neutralization potency per antibody is increased by approximately 100 times compared to using free antibodies. The improvements in neutralizing the virus are attributed to two factors: first, the shells act as a physical barrier that blocks the virus from interacting with host cells; second, the multivalent binding of the antibodies inside the shells lead to stronger attachment to the trapped virus, a phenomenon known as avidity. Pre-incubation of shells with HBV and simultaneous addition of both components separately to cells lead to comparable levels of neutralization, indicating rapid trapping of the virions by the shells. Our study highlights the potential of the DNA shell system to rationally create antivirals using components that, when used individually, show little to no antiviral effectiveness.


Subject(s)
DNA , Hepatitis B virus , Nanoshells , Hepatitis B virus/drug effects , Humans , Nanoshells/chemistry , DNA/chemistry , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Neutralization Tests , Antiviral Agents/chemistry , Antiviral Agents/pharmacology
3.
J Mater Chem B ; 11(41): 10003-10018, 2023 10 25.
Article in English | MEDLINE | ID: mdl-37843459

ABSTRACT

Radiotherapy (RT) is dominantly used in breast cancer therapy but is facing fierce side effects because of the limited difference between tumor and normal tissues in response to ionizing radiation. Herein, we construct a core-shell nanoparticle of UiO-66-NH2@AuNS. Then the solid gold shell was etched into hollow AuNS (HAuNS) and further modified with biotin-PEG-SH (PEG-bio) to obtain HAuNS@PEG-bio. HAuNS@PEG-bio demonstrates effective near infrared II (NIR-II) region photothermal therapy (PTT) performance, and the increase of temperature at the tumor site promotes the blood circulation to alleviate the hypoxia in the tumor microenvironment (TME). Meanwhile, HAuNS exhibits strong X-ray absorption and deposition ability due to the high atomic coefficient of elemental Au (Z = 79) and hollowed-out structure. Through the dual radiosensitization of the high atomic coefficient of Au and the hypoxia alleviation from PTT of HAuNS, the breast cancer cells could undergo immunogenic cell death (ICD) to activate the immune response. At the in vivo level, HAuNS@PEG-bio performs NIR-II photothermal, radiosensitization, and ICD therapies through cellular targeting, guided by infrared heat and CT imaging. This work highlights that the constructed biotin-decorated hollow gold nanoshell has a promising potential as a diagnostic and treatment integration reagents for the breast cancer.


Subject(s)
Breast Neoplasms , Nanoshells , Humans , Female , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/therapy , Nanoshells/chemistry , Biotin , Gold/pharmacology , Gold/chemistry , Hypoxia , Tumor Microenvironment
4.
J Nanobiotechnology ; 21(1): 138, 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37106405

ABSTRACT

Since the successful clinical trial of AuroShell for photothermal therapy, there is currently intense interest in developing gold-based core-shell structures with near-infrared (NIR) absorption ranging from NIR-I (650-900 nm) to NIR-II (900-1700 nm). Here, we propose a seed-mediated successive growth approach to produce gold nanoshells on the surface of the nanoscale metal-organic framework (NMOF) of UiO-66-NH2 (UiO = the University of Oslo) in one pot. The key to this strategy is to modulate the proportion of the formaldehyde (reductant) and its regulator / oxidative product of formic acid to harness the particle nucleation and growth rate within the same system. The gold nanoshells propagate through a well-oriented and controllable diffusion growth pattern (points → facets → octahedron), which has not been identified. Most strikingly, the gold nanoshells prepared hereby exhibit an exceedingly broad and strong absorption in NIR-II with a peak beyond 1300 nm and outstanding photothermal conversion efficiency of 74.0%. Owing to such superior performance, these gold nanoshells show promising outcomes in photoacoustic (PA), computed tomography (CT), and photothermal imaging-guided photothermal therapy (PTT) for breast cancer, as demonstrated both in vitro and in vivo.


Subject(s)
Nanoshells , Nanoshells/chemistry , Photothermal Therapy , Gold/chemistry , Multimodal Imaging , Phototherapy
5.
Nat Commun ; 13(1): 5249, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36068273

ABSTRACT

In vivo surface-enhanced Raman scattering (SERS) imaging allows non-invasive visualization of tumors for intraoperative guidance and clinical diagnostics. However, the in vivo utility of SERS is greatly hampered by the strong optical scattering and autofluorescence background of biological tissues and the lack of highly active plasmonic nanostructures. Herein, we report a class of porous nanostructures comprising a cubic AuAg alloy nanoshell and numerous nanopores. Such porous nanostructures exhibit excellent near-infrared II plasmonic properties tunable in a broad spectral range by varying the pore features while maintaining a small dimension. We demonstrate their exceptional near-infrared II SERS performance varying with the porous properties. Additionally, near-infrared II SERS probes created with porous cubic AuAg nanoshells are demonstrated with remarkable capability for in vivo visualization of sub-millimeter microtumors in a living mouse model. Our near-infrared II SERS probes hold great potentials for precise demarcation of tumor margins and identification of microscopic tumors.


Subject(s)
Nanopores , Nanoshells , Neoplasms , Animals , Gold/chemistry , Mice , Nanoshells/chemistry , Neoplasms/diagnostic imaging , Porosity , Spectrum Analysis, Raman/methods
6.
Proc Natl Acad Sci U S A ; 119(29): e2123527119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858309

ABSTRACT

A promising clinical trial utilizing gold-silica core-shell nanostructures coated with polyethylene glycol (PEG) has been reported for near-infrared (NIR) photothermal therapy (PTT) of prostate cancer. The next critical step for PTT is the visualization of therapeutically relevant nanoshell (NS) concentrations at the tumor site. Here we report the synthesis of PEGylated Gd2O3-mesoporous silica/gold core/shell NSs (Gd2O3-MS NSs) with NIR photothermal properties that also supply sufficient MRI contrast to be visualized at therapeutic doses (≥108 NSs per milliliter). The nanoparticles have r1 relaxivities more than three times larger than those of conventional T1 contrast agents, requiring less concentration of Gd3+ to observe an equivalent signal enhancement in T1-weighted MR images. Furthermore, Gd2O3-MS NS nanoparticles have r2 relaxivities comparable to those of existing T2 contrast agents, observed in agarose phantoms. This highly unusual combination of simultaneous T1 and T2 contrast allows for MRI enhancement through different approaches. As a rudimentary example, we demonstrate T1/T2 ratio MR images with sixfold contrast signal enhancement relative to its T1 MRI and induced temperature increases of 20 to 55 °C under clinical illumination conditions. These nanoparticles facilitate MRI-guided PTT while providing real-time temperature feedback through thermal MRI mapping.


Subject(s)
Contrast Media , Gadolinium , Gold , Magnetic Resonance Imaging , Nanoshells , Photothermal Therapy , Contrast Media/chemical synthesis , Gadolinium/chemistry , Gold/chemistry , Magnetic Resonance Imaging/methods , Nanoshells/chemistry , Photothermal Therapy/methods , Polyethylene Glycols/chemistry , Silicon Dioxide/chemistry
7.
Phys Chem Chem Phys ; 24(9): 5700-5709, 2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35187554

ABSTRACT

Plasmonic nanoshells have attracted significant interest due to their resonant optical properties providing excellent spectral tunability, promising for various biophotonic applications. In this work we discuss our experimental and theoretical results related to the synthesis and optical characterization of surface-modified gold nanoshells. The nanoshell growth mechanism is monitored by IR spectroscopy, and the effects of modification of the gold nanoshell surface by PEG-SH ((11-mercaptoundecyl)tetra(ethylene glycol)) molecules are studied using TEM and optical methods. A red shift of localized surface plasmon resonance is observed upon formation of a layer of PEG-SH molecules on the completed gold nanoshells. Uncompleted gold shells show tendency to detach from the spherical silica cores, and the underlying destabilizing mechanism is discussed. The experimentally measured optical extinction properties are in good agreement with the results of numerical simulations, which additionally shed light on the localized plasmon modes contributing to the extinction, as well as on the effects of nanoshell surface nonuniformity on the resonant plasmonic properties and local field enhancements.


Subject(s)
Nanoshells , Gold/chemistry , Nanoshells/chemistry , Silicon Dioxide/chemistry , Surface Plasmon Resonance
8.
J Nanobiotechnology ; 19(1): 297, 2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34593005

ABSTRACT

BACKGROUND: Photothermal therapy (PTT) is a highly effective treatment for solid tumors and can induce long-term immune memory worked like an in situ vaccine. Nevertheless, PTT inevitably encounters photothermal resistance of tumor cells, which hinders therapeutic effect or even leads to tumor recurrence. Naïve CD8+ T cells are mainly metabolized by oxidative phosphorylation (OXPHOS), followed by aerobic glycolysis after activation. And the differentiate of effector CD8+ T cell (CD8+ Teff) into central memory CD8+ T cell (CD8+ TCM) depends on fatty acid oxidation (FAO) to meet their metabolic requirements, which is regulated by adenosine monophosphate activated protein kinase (AMPK). In addition, the tumor microenvironment (TME) is severely immunosuppressive, conferring additional protection against the host immune response mediated by PTT. METHODS: Metformin (Met) down-regulates NADH/NADPH, promotes the FAO of CD8+ T cells by activating AMPK, increases the number of CD8+ TCM, which boosts the long-term immune memory of tumor-bearing mice treated with PTT. Here, a kind of PLGA microspheres co-encapsulated hollow gold nanoshells and Met (HAuNS-Met@MS) was constructed to inhibit the tumor progress. 2-Deoxyglucose (2DG), a glycolysis inhibitor for cancer starving therapy, can cause energy loss of tumor cells, reduce the heat stress response of tumor cell, and reverse its photothermal resistance. Moreover, 2DG prevents N-glycosylation of proteins that cause endoplasmic reticulum stress (ERS), further synergistically enhance PTT-induced tumor immunogenic cell death (ICD), and improve the effect of immunotherapy. So 2DG was also introduced and optimized here to solve the metabolic competition among tumor cells and immune cells in the TME. RESULTS: We utilized mild PTT effect of HAuNS to propose an in situ vaccine strategy based on the tumor itself. By targeting the metabolism of TME with different administration strategy of 2DG and perdurable action of Met, the thermotolerance of tumor cells was reversed, more CD8+ TCMs were produced and more effective anti-tumor was presented in this study. CONCLUSION: The Step-by-Step starving-photothermal therapy could not only reverse the tumor thermotolerance, but also enhance the ICD and produce more CD8+ TCM during the treatment.


Subject(s)
Immunologic Memory , Neoplasms , Photothermal Therapy , Thermotolerance , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Gold/chemistry , Male , Mice , Mice, Inbred C57BL , Nanoshells/chemistry , Neoplasms/immunology , Neoplasms/metabolism
9.
Theranostics ; 11(17): 8270-8282, 2021.
Article in English | MEDLINE | ID: mdl-34373741

ABSTRACT

Rationale: Glucose oxidase (GOx) has gained tremendous research interest recently as a glucose-consuming enzyme for tumor starvation therapy, while its in vivo applications are strictly limited by rapid deactivation, as well as side effects of non-specific catalysis. Methods: To address these issues, here we report a protective nano-shell to encapsule GOx for localized melanoma therapy delivered by dissolving microneedles (MNs). Inspired by cell membrane that separates and protects cell organelles and components from outside environment while selectively ingesting nutrition sources, we designed polydopamine (PDA)-structured nano-shell to allow free transportation of glucose for catalytic reaction, while impede the penetration of GOx, proteinase, and other GOx-deactivating macromolecules across the shell membrane. Results: GOx was well protected in core layer with persistent catalytic activity for at least 6 d under various biological matrixes (e.g., PBS, serum, and cell lysate) and surviving different harsh conditions (e.g., acid/base treatments, and proteinase-induced degradation). Such long-acting nano-catalyst can be easily integrated into MNs as topical delivery carrier for effective glucose consumption in melanoma tissue, achieving significant tumor growth inhibition via starvation therapy with minimized side effects as compared to systemic administration. Conclusion: This work provides an elegant platform for in vivo delivery of GOx, and our cell-mimicking nano-system can also be applied for other enzyme-based therapeutics.


Subject(s)
Drug Delivery Systems/methods , Glucose Oxidase/pharmacology , Melanoma/drug therapy , Nanoshells/administration & dosage , Administration, Cutaneous , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Glucose Oxidase/metabolism , Indoles/chemistry , Mice , Mice, Inbred C57BL , Nanoshells/chemistry , Polymers/chemistry , Skin Neoplasms/drug therapy , Starvation
10.
Int J Biol Macromol ; 189: 792-801, 2021 Oct 31.
Article in English | MEDLINE | ID: mdl-34455003

ABSTRACT

Tailoring nanomaterials with tunable properties is of great importance to develop multifunctional candidates in the biomedical field. In the present study, we aimed to develop a promising nano-hybrid system composed of chitosan (CS) and mesoporous silica nanoparticles with a silver nanoshell coat (CS-AgMSNs). The physicochemical properties of CS-AgMSNs films were characterized using various techniques. Further, the mechanical properties of CS-AgMSNs were evaluated and compared with those of undoped CS film. Moreover, the antimicrobial activities of CS-AgMSNs (with different concentrations) were assessed against E-coli, S. aureus, C. albicans, and A. niger. Our results demonstrated that increasing the concentrations of doped AgMSNs (10 to 40 mg) in CS films lowered their transparency and blocked light transmission effectively. The measured elastic modulus of CS-AgMSNs films (20 and 30 mg) showed a decrease in the stiffness of CS films. Also, the elongation at break for CS-AgMSNs (40 mg) indicated a better flexibility. CS-AgMSNs films (10-40 mg) showed an enhanced antimicrobial activity in a concentration-dependent manner compared to undoped CS films. Collectively, the results suggest that our nano-hybrid CS-AgMSNs matrix has unique and promising properties, and holds potential for use in the biomedical field, food packaging, and textile industry.


Subject(s)
Anti-Infective Agents/pharmacology , Chemical Phenomena , Chitosan/pharmacology , Nanoshells/chemistry , Silver/chemistry , Bacteria/drug effects , Elastic Modulus , Microbial Sensitivity Tests , Nanoshells/ultrastructure , Optical Phenomena , Porosity , Spectrometry, X-Ray Emission , Spectroscopy, Fourier Transform Infrared , Tensile Strength , X-Ray Diffraction , Yeasts/drug effects
11.
J Nanobiotechnology ; 19(1): 157, 2021 May 26.
Article in English | MEDLINE | ID: mdl-34039370

ABSTRACT

BACKGROUND: Smart nanoscale drug delivery systems that target acidic tumor microenvironments (TME) could offer controlled release of drugs and modulate the hypoxic TME to enhance cancer therapy. The majority of previously reported MnO2 nanostructures are nanoparticles, nanosheets, or nanocomposites incorporated with other types of nanoparticles, which may not offer the most effective method for drug loading or for the controlled release of therapeutic payloads. Previous studies have designed MnO2 nanoshells that achieve tumor-specific and enhanced combination therapy for localized advanced cancer. However, the therapeutic effect of MnO2 nanoshells on metastatic cancer is still uncertain. RESULT: Here, intelligent "theranostic" platforms were synthesized based on hollow mesoporous MnO2 (H-MnO2) nanoshells that were loaded with chemotherapy agents docetaxel and cisplatin (TP) to form H-MnO2-PEG/TP nanoshells, which were designed to alleviate tumor hypoxia, attenuate angiogenesis, trigger the dissolution of Mn2+, and synergize the efficacy of first-class anticancer chemotherapy. The obtained H-MnO2-PEG/TP nanoshells decomposed in the acidic TME, releasing the loaded drugs (TP) and simultaneously attenuated tumor hypoxia and hypoxia-inducible factor-1α (HIF-1α) expression by inducing endogenous tumor hydrogen peroxide (H2O2) decomposition. In vitro experiments showed that compared with the control group, the proliferation, colony formation and migration ability of CAL27 and SCC7 cells were significantly reduced in H-MnO2-PEG/TP group, while cell apoptosis was enhanced, and the expression of hypoxia-inducible factor-1α(HIF-1α) was down-regulated. In vivo experiments showed that tumor to normal organ uptake ratio (T/N ratio) of mice in H-MnO2-PEG/TP group was significantly higher than that in TP group alone (without the nanoparticle), and tumor growth was partially delayed. In the H-MnO2-PEG/TP treatment group, HE staining showed that most of the tumor cells were severely damaged, and TUNEL assay showed cell apoptosis was up-regulated. He staining of renal and liver sections showed no obvious fibrosis, necrosis or hypertrophy, indicating good biosafety. Fluorescence staining showed that HIF-1α expression was decreased, suggesting that the accumulation of MnO2 in the tumor caused the decomposition of H2O2 into O2 and alleviated the hypoxia of the tumor. CONCLUSION: In conclusion, a remarkable in vivo and in vitro synergistic therapeutic effect is achieved through the combination of TP chemotherapy, which simultaneously triggered a series of antiangiogenic and oxidative antitumor reactions.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Drug Therapy/methods , Hypoxia/drug therapy , Manganese Compounds/chemistry , Mouth Neoplasms/drug therapy , Nanoshells/chemistry , Squamous Cell Carcinoma of Head and Neck/drug therapy , Tumor Microenvironment/drug effects , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Delivery Systems , Head and Neck Neoplasms/drug therapy , Humans , Hydrogen Peroxide/metabolism , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Oxides/chemistry , Theranostic Nanomedicine/methods , Tumor Hypoxia/drug effects
12.
Sci Rep ; 11(1): 9404, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33931720

ABSTRACT

Pancreatic cancer is one of the most lethal malignancies with limited therapeutic options and dismal prognosis. Gemcitabine is the front-line drug against pancreatic cancer however with limited improvement of therapeutic outcomes. In this study we envisaged the integration of GEM with gold nanoshells which constitute an interesting class of nanomaterials with excellent photothermal conversion properties. Nanoshells were coated with thiol-capped poly(ethylene glycol) methacrylate polymers of different molecular weight via Au-S attachment. It was found that the molecular weight of the polymers affects the in vitro performance of the formulations; more importantly we demonstrate that the EC50 of nanoshell loaded GEM can be suppressed but fully restored and even improved upon laser irradiation. Our proposed nanoformulations outperformed the cytotoxicity of the parent drug and showed confined synergism under the tested in vitro conditions.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Deoxycytidine/analogs & derivatives , Nanoshells/chemistry , Pancreatic Neoplasms/drug therapy , Photochemotherapy , Cell Line, Tumor , Deoxycytidine/administration & dosage , Drug Screening Assays, Antitumor , Gold , Humans , Polymers , Gemcitabine
13.
J Nanobiotechnology ; 19(1): 77, 2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33741008

ABSTRACT

BACKGROUND: Although many treatments for breast cancer are available, poor tumour targeting limits the effectiveness of most approaches. Consequently, it is difficult to achieve satisfactory results with monotherapies. The lack of accurate diagnostic and monitoring methods also limit the benefits of cancer treatment. The aim of this study was to design a nanocarrier comprising porous gold nanoshells (PGNSs) co-decorated with methoxy polyethylene glycol (mPEG) and trastuzumab (Herceptin®, HER), a therapeutic monoclonal antibody that binds specifically to human epidermal receptor-2 (HER2)-overexpressing breast cancer cells. Furthermore, a derivative of the microtubule-targeting drug maytansine (DM1) was incorporated in the PGNSs. METHODS: Prepared PGNSs were coated with mPEG, DM1 and HER via electrostatic interactions and Au-S bonds to yield DM1-mPEG/HER-PGNSs. SK-BR-3 (high HER2 expression) and MCF-7 (low HER2) breast cancer cells were treated with DM1-mPEG/HER-PGNSs, and cytotoxicity was evaluated in terms of cell viability and apoptosis. The selective uptake of the coated PGNSs by cancer cells and subsequent intracellular accumulation were studied in vitro and in vivo using inductively coupled plasma mass spectrometry and fluorescence imaging. The multimodal imaging feasibility and synergistic chemo-photothermal therapeutic efficacy of the DM1-mPEG/HER-PGNSs were investigated in breast cancer tumour-bearing mice. The molecular mechanisms associated with the anti-tumour therapeutic use of the nanoparticles were also elucidated. RESULT: The prepared DM1-mPEG/HER-PGNSs had a size of 78.6 nm and displayed excellent colloidal stability, photothermal conversion ability and redox-sensitive drug release. These DM1-mPEG/HER-PGNSs were taken up selectively by cancer cells in vitro and accumulated at tumour sites in vivo. Moreover, the DM1-mPEG/HER-PGNSs enhanced the performance of multimodal computed tomography (CT), photoacoustic (PA) and photothermal (PT) imaging and enabled chemo-thermal combination therapy. The therapeutic mechanism involved the induction of tumour cell apoptosis via the activation of tubulin, caspase-3 and the heat shock protein 70 pathway. M2 macrophage suppression and anti-metastatic functions were also observed. CONCLUSION: The prepared DM1-mPEG/HER-PGNSs enabled nanodart-like tumour targeting, visibility by CT, PA and PT imaging in vivo and powerful tumour inhibition mediated by chemo-thermal combination therapy in vivo. In summary, these unique gold nanocarriers appear to have good potential as theranostic nanoagents that can serve both as a probe for enhanced multimodal imaging and as a novel targeted anti-tumour drug delivery system to achieve precision nanomedicine for cancers.


Subject(s)
Breast Neoplasms/diagnostic imaging , Gold/chemistry , Multimodal Imaging/methods , Nanoshells/chemistry , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/therapy , Cell Line, Tumor , Drug Liberation , Drug Tapering , Drug Therapy , Female , Humans , MCF-7 Cells , Macrophages , Maytansine/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Oxidation-Reduction , Photothermal Therapy , Polyethylene Glycols/chemistry , Porosity , Receptor, ErbB-2/metabolism
14.
Nanomedicine ; 33: 102349, 2021 04.
Article in English | MEDLINE | ID: mdl-33359414

ABSTRACT

Drug resistance and inefficient localization of chemotherapeutic agent limit the current treatment strategy in locally advanced melanoma (MEL), accounting to the 10-year survival rate from 24% to 68%. In this study we constructed anti-PD-L1 conjugated and doxorubicin loaded hollow gold nanoshell (T-HGNS-DOX) for targeted and localized chemo-photothermal therapy of MEL by the conjugation of LA-PEG-anti-PD-L1 antibody and short PEG chain on the surface of HGNS-DOX. Near infrared (NIR) as well as pH dependent drug release profile was observed. Significant uptake of DOX following NIR due to high PD-L1 receptors resulted in pronounced anticancer effect of T-HGNS-DOX. Following intratumoral administration, maximum nanoparticles retention with the significant reduction in tumor growth was observed as a result of elevated apoptosis marker (cleaved caspase-3, cleaved PARP) as well as downregulation of proliferative (Ki-67) and angiogenesis marker (CD31). Cumulatively, our system avoids the systemic toxicities of the nanosystem thereby providing maximum chemotherapeutic retention in tumor.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Doxorubicin/chemistry , Gold/chemistry , Melanoma/drug therapy , Melanoma/radiotherapy , Nanocapsules/chemistry , Nanoshells/chemistry , Animals , Antibodies, Monoclonal, Humanized/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Membrane Permeability , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Drug Compounding , Drug Liberation , Humans , Hydrogen-Ion Concentration , Male , Mice, Inbred C57BL , Molecular Targeted Therapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Phototherapy , Surface Properties
15.
J Biomater Appl ; 35(3): 430-445, 2020 09.
Article in English | MEDLINE | ID: mdl-32515640

ABSTRACT

A multifunctional targeted nanoplatform combining photothermal therapy and chemotherapy has emerged as a promising strategy for comprehensive therapies of breast cancer. In this study, we constructed human epidermal growth factor receptor 2 (Her2)-targeted gold nanoshelled poly(lactic-co-glycolic acid) hybrid nanocapsules encapsulating perfluorooctyl bromide, superparamagnetic iron oxide nanoparticles, and doxorubicin (Her2-GPDH nanocapsules) as theranostic agent for bimodal ultrasound/magnetic resonance imaging and synergistic photothermal-chemotherapy of Her2-postive breast cancer cells. Her2-GPDH nanocomposites possessed well-defined spherical morphology, and the average diameter was about 296 nm with good dispersion. Targeting assays demonstrated that Her2-GPDH nanocapsules exhibited higher targeting binding to Her2-positive SKBR3 cells than Her2-negative MDA-MB-231cells. The encapsulation efficiency and the loading content of doxorubicin in Her2-GPDH nanocapsules were 39 ± 1.45% and 3.8 ± 0.52%, respectively, and the agent exhibited pH-responsive and near-infrared light-triggered stepwise release behavior of doxorubicin. In vitro, the agent had potential to serve as feasible candidate for ultrasound imaging and T2-weighted magnetic resonance imaging with a relatively high relaxivity. Cell experiments confirmed that the agent had significant photothermal cytotoxicity on SKBR3 cells, and the combined photothermal-chemotherapy could significantly enhance the anti-tumor effect. In summary, the present Her2-GPDH nanocapsules, a novel multifunctional nanoplatform, will offer a new way for early bimodal molecular-level diagnosis and synergistic treatment of Her2-positve breast cancer.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Doxorubicin/chemistry , Gold/chemistry , Nanocapsules/chemistry , Nanoshells/chemistry , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Combined Modality Therapy , Contrast Media/chemistry , Doxorubicin/pharmacology , Drug Liberation , Female , Fluorocarbons/chemistry , Humans , Hydrocarbons, Brominated/chemistry , Magnetic Iron Oxide Nanoparticles/chemistry , Magnetic Resonance Imaging , Photochemotherapy , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Receptor, ErbB-2/metabolism , Ultrasonography
16.
Molecules ; 25(11)2020 Jun 05.
Article in English | MEDLINE | ID: mdl-32516956

ABSTRACT

Different types of gold nanoparticles have been synthesized that show great potential in medical applications such as medical imaging, bio-analytical sensing and photothermal cancer therapy. However, their stability, polydispersity and biocompatibility are major issues of concern. For example, the synthesis of gold nanorods, obtained through the elongated micelle process, produce them with a high positive surface charge that is cytotoxic, while gold nanoshells are unstable and break down in a few weeks due to the Ostwald ripening process. In this work, we report the self-assembly of the capsid protein (CP) of cowpea chlorotic mottle virus (CCMV) around spherical gold nanoparticles, gold nanorods and gold nanoshells to form virus-like particles (VLPs). All gold nanoparticles were synthesized or treated to give them a negative surface charge, so they can interact with the positive N-terminus of the CP leading to the formation of the VLPs. To induce the protein self-assembly around the negative gold nanoparticles, we use different pH and ionic strength conditions determined from a CP phase diagram. The encapsidation with the viral CP will provide the nanoparticles better biocompatibility, stability, monodispersity and a new biological substrate on which can be introduced ligands toward specific cells, broadening the possibilities for medical applications.


Subject(s)
Bromovirus/metabolism , Capsid Proteins/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Nanoshells/chemistry , Virion/metabolism , Ligands
17.
ACS Appl Mater Interfaces ; 12(28): 31172-31181, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32532159

ABSTRACT

The development of stimuli-responsive multifunctional nanocarriers for therapeutic drug delivery is extremely desirable for highly specific treatment of disease. Herein, thiol-polyethylene glycol-folate acid-modified hollow mesoporous bismuth nanoshells (HM-Bi@PEG-FA NSs) were developed as the new dual-stimuli-responsive single-"elemental" photothermal nanocarriers for synergistic chemo-photothermal therapy of tumor. The designed hollow-mesoporous-type nanocarriers present excellent photothermal conversion capacity (∼34.72%) and good biocompatibility. Meanwhile, acidic pH and near-infrared (NIR) laser dual-stimulated doxorubicin (DOX) release is successfully achieved. More importantly, the DOX-loaded HM-Bi@PEG-FA NSs hold an efficient in vitro/in vivo antitumor effect through the synergistic chemo-photothermal therapy. Therefore, our findings provide the possibility of designing a dual-stimuli-responsive hollow mesoporous Bi-based photothermal nanocarrier for synergistically enhanced antitumor therapy.


Subject(s)
Drug Delivery Systems/methods , Nanoshells/chemistry , Cell Line, Tumor , Doxorubicin/chemistry , Humans , Nanoparticles/chemistry , Phototherapy/methods
18.
Biomed Res Int ; 2020: 5869235, 2020.
Article in English | MEDLINE | ID: mdl-32352001

ABSTRACT

The purpose of this study was to investigate the effect of photothermal treatment (PTT) with gold nanoshell (ANS) using a macrophage-mediated delivery system in a head and neck squamous cell carcinoma (HNSCC) cell line. To achieve this, ANS-loaded rat macrophages (ANS-MAs) were prepared via the coculture method with ANS. The human HNSCC (FaDu cell) and macrophage (rat macrophage; NR8383 cell) hybrid spheroid models were generated by the centrifugation method to determine the possibility of using ANS-MAs as a cancer therapy. These ANS-MAs were set into the tumor and macrophage hybrid spheroid model to measure PTT efficacy. Kinetic analysis of the spheroid growth pattern revealed that this PTT process caused a decreasing pattern in the volume of the hybrid model containing ANS-MAs (p < 0.001). Comparison with empty macrophages showed harmony between ANS and laser irradiation for the generation of PTT. An annexin V/dead cell marker assay indicated that the PTT-treated hybrid model induced increasing apoptosis and dead cells. Further studies on the toxicity of ANS-MAs are needed to reveal whether it can be considered biocompatible. In summary, the ANS was prepared with a macrophage as the delivery method and protective carrier. The ANS was successfully localized to the macrophages, and their photoabsorption property was stationary. This strategy showed significant growth inhibition of the tumor and macrophage spheroid model under NIR laser irradiation. In vivo toxicology results suggest that ANS-MA is a promising candidate for a biocompatible strategy to overcome the limitations of fabricated nanomaterials. This ANS-MA delivery and PTT strategy may potentially lead to improvements in the quality of life of patients with HNSCC by providing a biocompatible, minimally invasive modality for cancer treatment.


Subject(s)
Gold , Hyperthermia, Induced , Nanoshells , Squamous Cell Carcinoma of Head and Neck , Animals , Cell Line, Tumor , Gold/chemistry , Gold/pharmacology , Humans , Macrophages/metabolism , Macrophages/pathology , Nanoshells/chemistry , Nanoshells/therapeutic use , Rats , Rats, Sprague-Dawley , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/therapy
19.
Soft Matter ; 16(17): 4173-4181, 2020 May 07.
Article in English | MEDLINE | ID: mdl-32286601

ABSTRACT

Among the lipid nanoparticles, lipid polymer hybrid nanoparticles (HNPs) composed of an oily core and a polymeric shell display interesting features as efficient drug carriers due to the high loading capability of the oil phase and the stability and surface functionalization of the polymer shell. Herein, we formulated lipid-core/polymer-shell hybrid nanoparticles (HNPs) using a simple nanoprecipitation method involving Vitamin E Acetate (VEA) as the oily core and a tailor-made amphiphilic polymer as a wrapping shell. The fluorescence labeling of the oil, using a newly developed green fluorogenic BODIPY tracker, and of the polymer using a covalent attachment of a red emitting rhodamine was done to assess the formation, the composition and the stability of these new hybrid nanoparticles using dual color electrophoresis gel analysis. This technique, combined to conventional DLS and electronic microscopy analysis, allowed us to quickly determine that 20 wt% of the polymer was an optimal ratio for obtaining stable HNPs by nanoprecipiation. Finally, we showed that using different polymeric shells, various HNPs can be obtained and finely discriminated using a combined approach of electrophoresis and two-color labeling.


Subject(s)
Drug Carriers/chemistry , Fluorescent Dyes/chemistry , Lipids/chemistry , Nanoshells/chemistry , Polymers/chemistry , Acetates/chemistry , Boron Compounds/chemistry , Rhodamines/chemistry , Spectrometry, Fluorescence , Vitamin E/chemistry
20.
Nanoscale ; 12(11): 6429-6437, 2020 Mar 21.
Article in English | MEDLINE | ID: mdl-32141450

ABSTRACT

The high reactivity of silver nanoparticles leads to their broad applications in the anti-bacterial field; however, the safety of silver nanoparticles has attracted increasing public attention. After exposure to silver nanoparticles in vivo, the liver serves as their potential deposition site; however the potential biological effects of such nanoparticles on hepatocytes at low dosages are not well understood. Here, we study the interaction between gold nanorod core/silver shell nanostructures (Au@Ag NRs) and human hepatocytes, HepG2 cells, and determine that Au@Ag NRs at sub-lethal doses can induce autophagy. After uptake, Au@Ag NRs mainly localize in the lysosomes where they release silver ions and promote the production of reactive oxygen species (ROS). The ROS then suppress the AKT-mTOR signaling pathway and activate autophagy. In addition, oxidative stress results in lysosomal impairment, causing decreased ability for lysosomal digestion. Moreover, oxidative stress also affects the structure and function of mitochondria, leading to the initiation of protective autophagy to eliminate the damaged mitochondrion. Our study shows that at sub-lethal dosages, silver nanomaterials may alter the physiological functions of hepatic cells by activating protective autophagy and cause potential health risks, indicating that cautious consideration of the safety of nanomaterials for certain applications is necessary.


Subject(s)
Autophagy/drug effects , Gold , Hepatocytes/metabolism , Nanoshells/chemistry , Nanotubes/chemistry , Silver , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Hep G2 Cells , Humans , Mitochondria, Liver/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Silver/chemistry , Silver/pharmacokinetics , Silver/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...