Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
J Mater Chem B ; 8(1): 65-77, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31768514

ABSTRACT

Gold nanostars (AuNSs), with unique physicochemical properties, are thought to be a promising agent for photothermal therapy (PTT). In this study, we prepared PEGylated gold nanostars (pAuNSs) using the HEPES-reduction method. The high photothermal conversion efficiency (∼80%) and photothermal stability of pAuNSs were demonstrated in vitro and in vivo. 111In-DTPA-pAuNSs were prepared as a radioactive surrogate for the biodistribution studies of pAuNSs. In both microSPECT/CT images and the biodistribution study, the tumor-to-muscle (T/M) ratio reached a maximum at 24 h post intravenous injection of 111In-DTPA-pAuNSs. The high linear correlation between the 111In radioactivity and the gold content in the tumors (R2 0.86-0.99) indicated that 111In-DTPA-pAuNSs were appropriate for noninvasively tracking pAuNSs in vivo after systemic administration. Histological examination after silver enhancement staining clearly illustrated that the accumulated pAuNSs in the tumors were mainly located on the luminal surface of vessels. The mice bearing a SKOV-3 xenograft exhibited remarkable therapeutic efficacy with negligible organ damage after receiving pAuNS-mediated photothermal therapy. Our findings suggested that pAuNSs, together with their radioactive surrogate 111In-DTPA-pAuNSs, are promising for applications in image-guided photothermal therapy.


Subject(s)
Gold/pharmacokinetics , Metal Nanoparticles/therapeutic use , Neoplasms/therapy , Phototherapy/methods , Polyethylene Glycols/pharmacokinetics , Theranostic Nanomedicine/methods , Animals , Cell Line, Tumor , Female , Gold/therapeutic use , Humans , Mice , Mice, Inbred BALB C
2.
ACS Appl Mater Interfaces ; 11(43): 39624-39632, 2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31573175

ABSTRACT

To advance anti-tumor efficiency and lessen the adverse effect caused by nanodrug residues in the body, a smart nanoagent system is developed and successfully used in intracellular ATP imaging and in vivo chemo-photothermal synergetic therapy. The nanoagent system is facilely prepared using a DNA complex to modify gold nanoparticles (AuNPs). The DNA complex is formed by three oligonucleotides (ATP aptamer, rC-DNA, and rG-DNA). The CG-rich structure in a ternary DNA complex could be exploited for payload of chemotherapeutic medicine doxorubicin (DOX), thus making efficient DOX transport into the tumor site possible. In tumor cells, especially in acidic organelles (e.g., endosome and lysosome), DOX could be rapidly released via the dual stimuli of overexpressed ATP and pH. What is more, the specific recognition of a fluorescently labeled aptamer strand to ATP can achieve the intracellular ATP imaging. pH-controlled reversible folding and unfolding of intermolecular i-motif formed by C-rich strands can lead to intracellular in situ assembly of AuNP aggregates with high photothermal conversion efficiency and promote relatively facile renal clearance of AuNPs through the disassociation of the aggregates in extracellular environments. Experiments in vivo and vitro present feasibility for a synergetic chemo-photothermal therapy. Such an in situ reversible assembly strategy of a chemo-photothermal agent also presents a new paradigm for a smart and highly efficient disease treatment with reduced side effects.


Subject(s)
Adenosine Triphosphate/metabolism , Doxorubicin , Gold , Hyperthermia, Induced , Metal Nanoparticles , Molecular Imaging , Neoplasms, Experimental , Phototherapy , Animals , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Female , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , HeLa Cells , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/therapy , Xenograft Model Antitumor Assays
3.
Colloids Surf B Biointerfaces ; 184: 110496, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31525600

ABSTRACT

A greener approach for the design of surface plasmon resonant gold nanoparticles has been obtained with a hydrosoluble fraction of an endemic asteraceae medicinal plant. This medicinal plant is originated from Indian Ocean and demonstrates its bioreducing activity in the design of stable green nanomedicine in aqueous media. This article describes the preclinical assessment of the efficacy of these novel nanocandidates on murine model by intratumoral and intravenous injections. It definitely demonstrates two key points in the treatment of cancer: 1) optimization of the tumor microenvironment targeting by specific ligands for a limited damage on healthy tissue, 2) the need to screen the specific irradiation dose (time, power) taking into account the type of tumor.


Subject(s)
Gold/chemistry , Green Chemistry Technology/methods , Metal Nanoparticles/chemistry , Surface Plasmon Resonance/methods , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Asteraceae/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Gold/administration & dosage , Gold/pharmacokinetics , Humans , Injections, Intralesional , Injections, Intravenous , Light , Metal Nanoparticles/administration & dosage , Mice , Nanomedicine/methods , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Phototherapy/methods , Proof of Concept Study , Tumor Burden/drug effects , Tumor Burden/radiation effects
4.
IET Nanobiotechnol ; 13(6): 634-639, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31432798

ABSTRACT

In present study, the effective penetration of radiofrequency (RF) induced gold decorated iron oxide nanoparticles (GS@IONPs) hyperthermia was investigated. The effective penetration depth of RF also the damage potency of hyperthermia was evaluated during histopathology observations which were done on the chicken breast tissue and hepatocellular carcinoma (HCC) models. The thermal damages are well- documented in our previous cellular study which was engaged with potency of RF hyperthermia in Epithelial adenocarcinoma (MCF-7) and fibroblast (L-929) cells deaths [1]. In recent work, PEGylated iron oxide nanoparticles (IONPs) were used as base platform for gold magnetic nanoparticles (GS@IONPs) formation. The 144.00015 MHz, 180W RF generator was applied for stimulating the nanoparticles. The chicken breast tissue and the hepatocellular tumor model was considered in the experimental section. In histology studies, the structural changes also the effective penetration depth of RF induced nanoparticles was observed through microscopic monitoring of the tissue slices in histology observations (Gazi medical school). The highest damage level was seen in 8.0 µm tissue slices where lower damages were seen in depth of 1.0 cm and more inside tissue. The histology observations clarified the effective penetration depth of RF waves and irreversible damages in the 2.0 cm inside the tissue.


Subject(s)
Gold/pharmacokinetics , Hyperthermia, Induced , Metal Nanoparticles , Radio Waves , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Chickens , Drug Liberation , Gold/chemistry , Histological Techniques/methods , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Metal Nanoparticles/chemistry , Radiofrequency Therapy , Tissue Distribution
5.
ACS Appl Mater Interfaces ; 11(32): 28621-28630, 2019 Aug 14.
Article in English | MEDLINE | ID: mdl-31293148

ABSTRACT

Multiple amino acid (glutamine and lysine)-modified gold nanoparticles a with pH-switchable zwitterionic surface were fabricated through coordination bonds using ferrous iron (Fe2+) as bridge ions, which are able to spontaneously and selectively assemble in tumor cells for accurate tumor therapy combining enzyme-triggered photothermal therapy and H2O2-dependent catalytic medicine. These gold nanoparticles showed electric neutrality at pH 7.4 (hematological system) to prevent endocytosis of normal cells, which could be positively charged at pH 6.8 (tumor microenvironment) to promote the endocytosis of tumor cells to these nanoparticles, performing great tumor selectivity. After cell uptake, the specific enzyme (transglutaminase) in tumor cells would catalyze the polymerization of glutamine and lysine to cause the intracellular assembly of these gold nanoparticles, resulting in an excellent photothermal property for accurate tumor therapy. Moreover, the Fe2+ ion could decompose excess hydrogen peroxide (H2O2) in tumor cells via the Fenton reaction, resulting in a large amount of hydroxyl radicals (·OH). These radicals would also cause tumor cell damage. This synergetic therapy associating with high tumor selectivity generated an 8-fold in vitro cytotoxicity against tumor cells compared with normal cells under 48 h incubation with 10 min NIR irradiation. Moreover, in vivo data from tumor-bearing nude mice models showed that tumors can be completely inhibited and gradually eliminated after multimode treatment combining catalytic medicine and photothermal therapy for 3 weeks. This system takes advantage of three tumor microenvironment conditions (low pH, enzyme, and H2O2) to trigger the therapeutic actions, which is a promising platform for cancer therapy that achieved prolonged circulation time in the blood system, selective cellular uptake, and accurate tumor therapy in multiple models.


Subject(s)
Gold , Hyperthermia, Induced , Melanoma, Experimental , Metal Nanoparticles , Neoplasm Proteins/metabolism , Phototherapy , Transglutaminases/metabolism , Amino Acids/chemistry , Amino Acids/pharmacokinetics , Amino Acids/pharmacology , Animals , Cell Line, Tumor , Coated Materials, Biocompatible , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Endocytosis/drug effects , Female , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Humans , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
6.
Biomater Sci ; 7(5): 2009-2022, 2019 Apr 23.
Article in English | MEDLINE | ID: mdl-30839987

ABSTRACT

In this work, we took advantage of a one-pot reaction to prepare tumor-targeting nanoparticles (Au@T), which could respond to the intracellular acidic environment and form aggregates to enhance the retention effect of nanoparticles in tumor cells. Au@T is composed of gold nanoparticles (Au NPs) modified with 4-mercaptobenzoic acid (MCBA), p-hydroxythiophenol (HTP), LA (lipoic acid)-PEG2K-OCH3 and LA-PEG2K-biotin. During blood circulation, Au@T remains well dispersed, making it inconspicuous. Then, with the help of active targeted transport, much more Au@T becomes internalized at the tumor site. After being internalized by tumor cells, Au@T aggregates under the condition of pH = 6.0, thereby improving the retention effect of Au@T, stymieing exocytosis and reducing the amount of nanoparticles returned to the blood stream. Furthermore, the in vivo experimental results showed that aggregated Au@T exhibits excellent photothermal effects, with a tumor inhibition rate of 86.40%. The computed tomography (CT) value was found to be 1.5 times higher than that of the control group (Au@Bio), as Au@Bio was unable to aggregate in tumor cells. In conclusion, this work provides a simple method for synthesizing a type of gold nanoparticles (Au@T) with promising potential for tumor diagnosis and treatment through enhancing the retention effect in tumor cells.


Subject(s)
Gold/chemistry , Gold/therapeutic use , Metal Nanoparticles/chemistry , Theranostic Nanomedicine , Cell Line, Tumor , Chemistry Techniques, Synthetic , Gold/metabolism , Gold/pharmacokinetics , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Materials Testing , Phototherapy , Tissue Distribution , Tomography, X-Ray Computed
7.
Biomaterials ; 197: 380-392, 2019 03.
Article in English | MEDLINE | ID: mdl-30703743

ABSTRACT

Systemic lupus erythematosus (SLE) constitutes an autoimmune disease characterized by the breakdown of tolerance to self-antigens, sustained production of pathogenic autoantibodies, and damage to multiple organs and tissues. Nanoparticle (NP)-based therapeutics have demonstrated efficacy in attenuating the progression of SLE. However, investigations of nano-drugs that address the crucial initiating factor in the pathogenesis of SLE; e.g., inefficient clearance of apoptotic cells by phagocytes and consequent accumulation of self-antigens, have seldom been reported. Here, an apoptotic cell-mimicking gold nanocage (AuNC)-based nano drug carrier capable of correcting the impaired clearance of apoptotic cells in SLE was rationally designed and generated by conjugating phosphatidylserine (PS) on the surface of liposome-coated AuNCs for liver X receptor (LXR) agonist T0901317 delivery. Notably, PS-lipos-AuNC@T0901317 could efficiently enhance apoptotic cell clearance by elevating the expression of Mer, one of the pivotal phagocytosis-associated receptors on macrophages, resulting in decreased production of anti-dsDNA autoantibodies, reduced inflammatory response, and alleviation of kidney damage in lupus model mice. Additionally, PS-lipos-AuNC could be tracked by photoacoustic imaging for nano drug carrier biodistribution. By addressing the crucial pathogenic factor of SLE, the NP-based delivery system in this study is envisioned to provide a promising strategy to treat this complex and challenging disease.


Subject(s)
Apoptosis , Drug Delivery Systems , Gold/administration & dosage , Hydrocarbons, Fluorinated/administration & dosage , Liver X Receptors/agonists , Lupus Erythematosus, Systemic/drug therapy , Nanocapsules/administration & dosage , Sulfonamides/administration & dosage , Animals , Autoantibodies/analysis , Cytokines/metabolism , Disease Progression , Drug Evaluation, Preclinical , Female , Gold/pharmacokinetics , Hydrocarbons, Fluorinated/therapeutic use , Hydrocarbons, Fluorinated/toxicity , Liposomes/administration & dosage , Lupus Nephritis/drug therapy , Lupus Nephritis/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Phosphatidylserines , Sulfonamides/therapeutic use , Sulfonamides/toxicity , Tissue Distribution , c-Mer Tyrosine Kinase/biosynthesis , c-Mer Tyrosine Kinase/genetics
8.
ACS Appl Mater Interfaces ; 11(7): 6777-6788, 2019 Feb 20.
Article in English | MEDLINE | ID: mdl-30668088

ABSTRACT

Recently, rodlike nanomaterials with specific aspect ratio for efficient cellular uptake have received enormous attention. For functional nanomaterials, such as photothermal agents, large surface areas for their rod-shaped exterior that increase the amount of light absorbed would lead to a higher absorption coefficient as well as drug-loading property. In this project, we coated rodlike mesoporous silica with gold nanoshells (MSNR@Au hybrid), modifying them with ultrasmall gadolinium (Gd)-chelated supramolecular photosensitizers, TPPS4 (MSNR@Au-TPPS4(Gd)), which could be applied to near-infrared fluorescence/multispectral optoacoustic tomography/computed tomography/magnetic resonance imaging and imaging-guided remotely controlled photothermal (PTT)/photodynamic (PDT) combined antitumor therapy. Gold nanoshells, as a perfect PTT agent, were used to assemble the rodlike mesoporous silica nanoparticles with larger superficial area and higher drug loading, thus obtaining the MSNR@Au hybrid. HS-ß-CD, which was used as the host, was adsorbed on the gold nanoshell (MSNR@Au-ß-CD) to link TPPS4(Gd) through the host-guest reaction, thus forming CD-TPPS4 supramolecular photosensitizers (supraPSs). Compared with conventional PSs, supraPSs have host screens, which could reduce the self-aggregation of TPPS4, and consequently generate 1O2 with high efficiency. The in vivo quadmodal imaging of MSNR@Au-TPPS4(Gd) nanoparticles revealed an intensive tumor uptake effect after injection. The in vivo antitumor efficacy further testified that the synergistic therapy, which was more efficient than any other monotherapy, exhibited an excellent tumor inhibition therapeutic effect. As a result, this encourages to further explore multifunctional theranostic nanoparticles based on gold shells for combined cancer therapy.


Subject(s)
Contrast Media , Gold , Hyperthermia, Induced/methods , Nanoshells , Neoplasms, Experimental , Photochemotherapy , Photosensitizing Agents , Silicon Dioxide , Tomography, X-Ray Computed , Animals , Cell Line, Tumor , Contrast Media/chemistry , Contrast Media/pharmacokinetics , Contrast Media/pharmacology , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Humans , Mice, Inbred BALB C , Mice, Nude , Nanoshells/chemistry , Nanoshells/therapeutic use , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/therapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacokinetics , Silicon Dioxide/pharmacology , Theranostic Nanomedicine/methods
9.
ACS Appl Mater Interfaces ; 11(2): 1951-1956, 2019 Jan 16.
Article in English | MEDLINE | ID: mdl-30571091

ABSTRACT

Thrombus diseases, induced by blood stasis or vascular embolization normally, frequently occur with high disability and mortalities worldwide. At present, drug thrombolysis, a primary clinical therapy for blood clot lysis, could increase the lethal risk for hemorrhage when thrombolysis agents are overused in the whole body. Therefore, a novel and advanced therapy for blood clot lysis, based on remote physical signals, is helpful for assisting clinical therapy. Here, we used the localized light-Au-hyperthermia (LAH) treatment, induced by gold nanorods (Au NRs) irradiated with near-infrared light (808 nm), for precise, rapid, and drug-free blood clot lysis. The LAH technology was first introduced in the murine hematoma model and the murine myocardial infarction model for blood clot lysis. Compared with traditional therapy, LAH was assured to shorten the time of detumescence in the murine hematoma model owing to their precise and localized hyperthermia. Meanwhile, we also discovered that LAH was a benefit to vascular recanalization in the murine myocardial infarction model. In addition, the Au NRs used in LAH present ideal biocompatibility in the murine model, which endows it to be suitable for blood clot lysis in vivo.


Subject(s)
Fibrinolytic Agents , Gold , Hyperthermia, Induced/methods , Metal Nanoparticles , Nanotubes/chemistry , Thrombolytic Therapy/methods , Animals , Disease Models, Animal , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacokinetics , Fibrinolytic Agents/pharmacology , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Rats
10.
Acta Biomater ; 83: 400-413, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30465921

ABSTRACT

Multi-stimuli-responsive theranostic nanoplatform integrating functions of both imaging and multimodal therapeutics holds great promise for improving diagnosis and therapeutic efficacy. In this study, we reported a pH, glutathione (GSH) and hyaluronidase (HAase) triple-responsive nanoplatform for HER2 and CD44 dual-targeted and fluorescence imaging-guided PDT/PTT dual-therapy against HER2-overexpressed breast cancer. The nanoplatform was fabricated by functionalizing gold nanorods (GNRs) with hyaluronic acid (HA) bearing pendant hydrazide and thiol groups via Au-S bonds, and subsequently chemically conjugating 5-aminolevulinic acid (ALA), Cy7.5 and anti-HER2 antibody onto HA moiety for PDT, fluorescence imaging and active targeting, respectively. The resulting versatile nanoplatform GNR-HA-ALA/Cy7.5-HER2 had uniform sizes, favorable dispersibility, as well as pH, GSH and HAase triple-responsive drug release manner. In vitro studies demonstrated that HER2 and CD44 receptor-mediated dual-targeting strategy could significantly enhance the cellular uptake of GNR-HA-ALA/Cy7.5-HER2. Under near-infrared (NIR) irradiation, MCF-7 cells could efficiently generate reactive oxygen species (ROS) and heat, and be more efficiently killed by a combination of PDT and PTT as compared with individual therapy. Pharmacokinetic and biodistribution studies showed that the nanoplatform possessed a circulation half-life of 1.9 h and could be specifically delivered to tumor tissues with an accumulation ratio of 12.8%. Upon the fluorescence imaging-guided PDT/PTT treatments, the tumors were completely eliminated without obvious side effects. The results suggest that the GNR-HA-ALA/Cy7.5-HER2 holds great potential for breast cancer therapy. STATEMENT OF SIGNIFICANCE: A combination of photodynamic therapy (PDT) and photothermal therapy (PTT) is emerging as a promising cancer treatment strategy. However, its therapeutic efficacy is compromised by the nonspecific delivery and unintended release of photo-responsive agents. Herein, we developed a multifunctional theranostic nanoplatform GNR-HA-ALA/Cy7.5-HER2 with pH, glutathione and hyaluronidase triple-responsive drug release for HER2 and CD44 dual-targeted and fluorescence imaging-guided PDT/PTT therapy against breast cancer. We demonstrated that HER2 and CD44 receptors-mediated dual-targeting strategy significantly enhanced the cellular uptake of GNR-HA-ALA/Cy7.5-HER2. We also demonstrated that the combined PDT/PTT treatment had significantly superior antitumor effect than PDT or PTT alone both in vitro and in vivo. Therefore, GNR-HA-ALA/Cy7.5-HER2 could serve as a promising nanoplatform for HER2-positive breast cancer therapy.


Subject(s)
Breast Neoplasms , Gold , Hyaluronic Acid , Hyperthermia, Induced , Metal Nanoparticles , Nanotubes/chemistry , Photochemotherapy , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/pharmacokinetics , Aminolevulinic Acid/pharmacology , Animals , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Female , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacokinetics , Hyaluronic Acid/pharmacology , MCF-7 Cells , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
11.
ACS Appl Mater Interfaces ; 11(1): 278-287, 2019 Jan 09.
Article in English | MEDLINE | ID: mdl-30520633

ABSTRACT

Heretofore, conjugated polymers (CPs) attract considerable attention in photothermal therapy (PTT). Although various CPs with different structures have been reported, the suboptimal circulation persistence and biodistribution limit their efficacy in tumor treatment. Human serum albumin (HSA), an endogenous plasma protein, has been widely functioned as a carrier for therapeutic agents. Herein, we construct nanocomplex C16 pBDP@HSA nanoparticles (NPs) from hydrophobic 4,4-difluoro-4-bora-3 a,4 a-diaza- s-indacene (BODIPY)-containing CPs and HSA, which exhibit robust stability in physiological conditions and excellent photothermal activity upon irradiation. The high photothermal conversion efficiency of 37.5%, higher than that of other reported PTT agents such as gold nanorods, phosphorus quantum dots, and 2D materials, results in the potent photocytotoxicity toward cancer cells. Simultaneously, C16 pBDP@HSA NPs' capabilities of near-infrared fluorescence and photoacoustic imaging can provide guidance to the PTT. The outstanding inhibition of tumor growth results from great photothermal activity, the benefited accumulation in tumor, and optimal timing of treatment. To the best of our knowledge, this is the first study which combines the BODIPY-based CPs and HSA in one nanostructure and finds application in cancer treatment. Moreover, this article also offers a new strategy for other insoluble macromolecules to explore more biomedical applications.


Subject(s)
Gold , Hyperthermia, Induced , Metal Nanoparticles , Nanotubes/chemistry , Neoplasms , Photoacoustic Techniques , Phototherapy , Serum Albumin, Human , Boron Compounds/chemistry , Boron Compounds/pharmacokinetics , Boron Compounds/pharmacology , Cell Line, Tumor , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Humans , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Serum Albumin, Human/chemistry , Serum Albumin, Human/pharmacokinetics , Serum Albumin, Human/pharmacology
12.
Acta Biomater ; 83: 414-424, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30366131

ABSTRACT

Nanoparticles are emerging as a new therapeutic modality due to their high stability, precise targeting, and high biocompatibility. Branched Au-Ag nanoparticles with polydopamine coating (Au-Ag@PDA) have strong near-infrared absorbance and no cytotoxicity but high photothermal conversion efficiency. However, the photothermal activity of Au-Ag@PDA in vivo and in vitro has not been reported yet, and the mechanism underlying the effects of photothermal nanomaterials is not clear. Therefore, in this study, the colorectal cancer cell line HCT-116 and nude mice xenografts were used to observe the photothermal effects of Au-Ag@PDA in vivo and in vitro. The results suggest that Au-Ag@PDA NPs significantly inhibited cell proliferation and induced apoptosis in colorectal cancer cells. Moreover, Au-Ag@PDA NP-mediated photothermal therapy inhibited the growth of tumors at doses of 50 and 100 µg in vivo. The mechanisms through which Au-Ag@PDA NPs induced colorectal cancer cell death involved multiple pathways, including caspase-dependent and -independent apoptosis, mitochondrial damage, lysosomal membrane permeability, and autophagy. Thus, our findings suggest that Au-Ag@PDA NPs could be used as potential antitumor agents for photothermal ablation of colorectal cancer cells.


Subject(s)
Apoptosis , Coated Materials, Biocompatible , Colorectal Neoplasms , Gold , Hyperthermia, Induced , Indoles , Metal Nanoparticles , Phototherapy , Polymers , Silver , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacokinetics , Coated Materials, Biocompatible/pharmacology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , HCT116 Cells , Humans , Indoles/chemistry , Indoles/pharmacokinetics , Indoles/pharmacology , Male , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Polymers/chemistry , Polymers/pharmacokinetics , Polymers/pharmacology , Silver/chemistry , Silver/pharmacokinetics , Silver/pharmacology , Xenograft Model Antitumor Assays
13.
ACS Nano ; 12(7): 6597-6611, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29969226

ABSTRACT

We report sub-100 nm optical/magnetic resonance (MR)/X-ray contrast-bearing theranostic nanoparticles (TNPs) for interventional image-guided photothermal therapy (PTT) of solid tumors. TNPs were composed of Au@Gd2O3:Ln (Ln = Yb/Er) with X-ray contrast (∼486 HU; 1014 NPs/mL, 0.167 nM) and MR contrast (∼1.1 × 108 mM-1 S-1 at 9.4 T field strength). Although TNPs are deposited in tumors following systemic administration via enhanced permeation and retention effect, the delivered dose to tumors is typically low; this can adversely impact the efficacy of PTT. To overcome this limitation, we investigated the feasibility of site-selective hepatic image-guided delivery of TNPs in rats bearing colorectal liver metastasis (CRLM). The mesenteric vein of tumor-bearing rats was catheterized, and TNPs were infused into the liver by accessing the portal vein for site-selective delivery. The uptake of TNPs with hepatic delivery was compared with systemic administration. MR imaging confirmed that delivery via the hepatic portal vein can double the CRLM tumor-to-liver contrast compared with systemic administration. Photothermal ablation was performed by inserting a 100 µm fiber-optic carrying 808 nm light via a JB1, 3-French catheter for 3 min under DynaCT image guidance. Histological analysis revealed that the thermal damage was largely confined to the tumor region with minimal damage to the adjacent liver tissue. Transmission electron microscopy imaging validated the stability of core-shell structure of TNPs in vivo pre- and post-PTT. TNPs comprising Gd-shell-coated Au nanorods can be effectively employed for the site-directed PTT of CRLM by leveraging interventional radiology methods.


Subject(s)
Colorectal Neoplasms/pathology , Gadolinium/therapeutic use , Gold/therapeutic use , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Nanoparticles/therapeutic use , Theranostic Nanomedicine/methods , Animals , Cell Line, Tumor , Contrast Media/administration & dosage , Contrast Media/pharmacokinetics , Contrast Media/therapeutic use , Gadolinium/administration & dosage , Gadolinium/pharmacokinetics , Gold/administration & dosage , Gold/pharmacokinetics , Humans , Hyperthermia, Induced/methods , Liver/blood supply , Liver/pathology , Liver Neoplasms/diagnosis , Liver Neoplasms/pathology , Magnetic Resonance Imaging/methods , Nanoparticles/administration & dosage , Phototherapy/methods , Radiology, Interventional/methods , Rats , Rats, Wistar
14.
J Nanosci Nanotechnol ; 18(10): 6791-6798, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29954495

ABSTRACT

In the ancient traditional Indian Ayurvedic system of natural healing, gold nanoparticles (Swarna Bhasma, gold ash) have been used for its therapeutic benefits as far back as 2500 B.C. Ayurvedic medicinal preparations are complex mixtures that include many plant-derived products and metals. Bhasmas date as far back as the 8th century and are made by samskaras (processings), such as shodhana (purification and potentiation), jarana (roasting), and marana (incineration, trituration) in the presence of plant products, including juices and concoctions. Previous studies characterized the physical properties of gold ash, and the mechanisms of its entry into human cells, but only preliminary data exist on its toxicity. Before using nanoparticles for therapeutic application, it is extremely important to study their toxicity and cellular internalization. In the present study, various imaging techniques were used to investigate Swarna Bhasma's (gold nanopowder) toxicity in both cancerous and noncancerous cells (HeLa and HFF-1) and to characterize its spectral properties. The results showed that gold ash particles had no impact on the cellular viability of both HeLa and HFF-1 cells, even at high concentrations or long incubation times. Moreover, it was found that the internalization level of Swarna Bhasma to cells may be improved by mechanical breaking of the large aggregates into smaller agglomerates. Hyperspectral images revealed that after breaking, the small agglomerates have different spectral properties in cells, compared to the original aggregates, suggesting that size of particles is instrumental for the subcellular interaction with human cells.


Subject(s)
Gold/pharmacology , Gold/pharmacokinetics , Latex/pharmacology , Latex/pharmacokinetics , Arsenic/adverse effects , Arsenic/pharmacokinetics , Arsenic/pharmacology , Calotropis/adverse effects , Cell Line , Cell Survival/drug effects , Drug Combinations , Gold/adverse effects , HeLa Cells , Humans , Latex/adverse effects , Lead/adverse effects , Lead/pharmacokinetics , Lead/pharmacology , Medicine, Ayurvedic , Metal Nanoparticles/adverse effects , Particle Size
15.
Drug Dev Ind Pharm ; 44(10): 1679-1684, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29939766

ABSTRACT

OBJECTIVE: It is difficult to identify the gold nanoparticles (AuNPs) intracellularly due to their non-fluorescent nature. Although gold can quench the fluorescence of any fluorophore, hence it is also difficult to combine gold with a fluorophore such as a semiconductor quantum dots (QDs). The aim of this study was to prepare a single fluorescent stable AuNPs combined with QDs (QDs-Au-NPs) which can be easily detected intracellularly. METHODS: QDs-Au-NPs were prepared via a simple one-step process through controlling the spacing between them using polyethylene glycol (PEG) as space linker in the form of PEGylated QDs. Furthermore, the applicability of this system was evaluated after coating the particles with somatostatin citrate, SST, to active target somatostatin receptors (SSTRs), and identification of the internalized particles via confocal laser scanning spectroscopy. RESULTS: The results showed that the produced Au shell has a thickness of 2.0 ± 0.2 nm and QDs-Au-NPs showed the same fluorescence intensity compared to the unmodified QDs. Additionally, a stable monodisperse QDs-Au-NPs coated with SST were prepared after coating with 11-Mercaptoundecanoic acid. Moreover, cellular uptake study in Human Caucasian breast adenocarcinoma cell lines showed that QDs-Au-SST-NPs could be detected easily using the confocal microscope. In addition, they showed a significant (p ≤ .05) internalization per cell compared to untreated QDs-Au-NPs as detected by flow cytometry. CONCLUSION: It could be concluded that the produced QDs-Au-NPs has a strong fluorescence property like QDs which enable them to be easily detected after cells internalization.


Subject(s)
Fluorescent Dyes/chemical synthesis , Gold/chemistry , Metal Nanoparticles/chemistry , Drug Evaluation, Preclinical/methods , Fluorescent Dyes/pharmacokinetics , Gold/pharmacokinetics , Humans , MCF-7 Cells
16.
Acta Biomater ; 75: 334-345, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29885528

ABSTRACT

Effects of nanosized drug delivery systems on cancer are often compromised due to their low drug loadings, premature drug release and multi-drug resistance (MDR). Herein, we reported a glutathione detonated and pH responsive nano-cluster of Au nanorods (AuNRs) with chemotherapeutic doxorubicin (DOX) and pre-chemosensitizer polycurcumin to treat MCF-7/ADR cells. The nano-cluster was prepared by self-assembling of AuNRs conjugated with DOX and amphiphilic poly(curcumin-co-dithiodipropionic acid)-b-biotinylated poly(ethylene glycol) via an emulsion/solvent evaporation technique, termed AuNR Cluster. The AuNR Cluster had a high drug loading (31.5% DOX), presenting much better aqueous solubility and stability at physiological pH than their individual AuNRs. The AuNR Cluster could be detonated to be their individual AuNRs at an intracellular concentration level of glutathione (GSH) (5 mM) and triggered to release DOX at an acidic pH (pH 6.8 or 5.0), which effectively facilitated cellular uptake of DOX (607 vs 356 a.u. for AuNRs at 12 h) and inhibited DOX efflux (471.33 vs 39.17 a.u. for free DOX at 24 h). The IC50 value of DOX against MCF-7/ADR cells for AuNR Cluster was 4.15 µg/mL, much lower than that for free DOX (90.97 µg/mL). The AuNR Cluster took much more photothermal effects than their corresponding AuNRs and presented enhanced anti-tumor effect (IC50: 2.61 µg/mL) under 808 nm laser irradiation. STATEMENT OF SIGNIFICANCE: Nano-sized drug delivery systems for anti-MDR cancer is still a challenging task. Herein, AuNR Cluster was self-assembled by individual AuNRs via emulsion/solvent evaporation technique, having a structure consisting of hydrophobic DOX/PCDA-AuNR core and hydrophilic biotin-PEG chain shell. AuNR Cluster is detonated to disintegrate and yield its individual AuNRs at an intracellular concentration level of glutathione (5 mM) and triggered to release DOX at an acidic pH (6.8 or 5.0). In comparison with its individual AuNRs, AuNR Cluster has better water solubility and stability, greater photothermal effects under NIR irradiation, bigger cytotoxicity against MCF-7/ADR cells. AuNR Cluster is expected to be a potential nanomedicine for treatment of MDR cancer.


Subject(s)
Doxorubicin , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Glutathione/metabolism , Gold , Nanotubes/chemistry , Neoplasms , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Female , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Neoplasms/drug therapy , Neoplasms/pathology
17.
Acta Biomater ; 72: 287-294, 2018 05.
Article in English | MEDLINE | ID: mdl-29578086

ABSTRACT

We report sub-100 nm metal-shell (Au) dielectric-core (BaTiO3) nanoparticles with bimodal imaging abilities and enhanced photothermal effects. The nanoparticles efficiently absorb light in the near infrared range of the spectrum and convert it to heat to ablate tumors. Their BaTiO3 core, a highly ordered non-centrosymmetric material, can be imaged by second harmonic generation, and their Au shell generates two-photon luminescence. The intrinsic dual imaging capability allows investigating the distribution of the nanoparticles in relation to the tumor vasculature morphology during photothermal ablation. Our design enabled in vivo real-time tracking of the BT-Au-NPs and observation of their thermally-induced effect on tumor vessels. STATEMENT OF SIGNIFICANCE: Photothermal therapy induced by plasmonic nanoparticles has emerged as a promising approach to treating cancer. However, the study of the role of intratumoral nanoparticle distribution in mediating tumoricidal activity has been hampered by the lack of suitable imaging techniques. This work describes metal-shell (Au) dielectric-core (BaTiO3) nanoparticles (abbreviated as BT-Au-NP) for photothermal therapy and bimodal imaging. We demonstrated that sub-100 nm BT-Au-NP can efficiently absorb near infrared light and convert it to heat to ablate tumors. The intrinsic dual imaging capability allowed us to investigate the distribution of the nanoparticles in relation to the tumor vasculature morphology during photothermal ablation, enabling in vivo real-time tracking of the BT-Au-NPs and observation of their thermally-induced effect on tumor vessels.


Subject(s)
Adenocarcinoma/therapy , Barium Compounds , Gold , Hyperthermia, Induced , Mammary Neoplasms, Experimental/therapy , Nanoparticles , Phototherapy , Titanium , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Barium Compounds/chemistry , Barium Compounds/pharmacokinetics , Barium Compounds/pharmacology , Cell Line, Tumor , Female , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Titanium/chemistry , Titanium/pharmacokinetics , Titanium/pharmacology
18.
J Mater Sci Mater Med ; 28(9): 143, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28819929

ABSTRACT

This paper explores the adhesion of biosynthesized gold nanoparticles (AuNPs) and gold (Au) nanoparticle/prodigiosin (PG) drug nanoparticles to breast cancer cells (MDA-MB-231 cells). The AuNPs were synthesized in a record time (less than 30 s) from Nauclea latifolia leaf extracts, while the PG was produced via bacterial synthesis with Serratia marcescens sp. The size distributions and shapes of the resulting AuNPs were characterized using transmission electron microscopy (TEM), while the resulting hydrodynamic diameters and polydispersity indices were studied using dynamic light scattering (DLS). Atomic Force Microscopy (AFM) was used to study the adhesion between the synthesized gold nanoparticles (AuNPs)/LHRH-conjugated AuNPs and triple negative breast cancer cells (MDA-MB-231 cells), as well as the adhesion between LHRH-conjugated AuNP/PG drug and MDA-MB-231 breast cancer cells. The adhesion forces between LHRH-conjugated AuNPs and breast cancer cells are shown to be five times greater than those between AuNPs and normal breast cells. The increase in adhesion is shown to be due to the over-expression of LHRH receptors on the surfaces of MDA-MB-231 breast cancer cells, which was revealed by confocal immuno-fluorescence microscopy. The implications of the results are then discussed for the selective and specific targeting and treatment of triple negative breast cancer.


Subject(s)
Gold/pharmacokinetics , Metal Nanoparticles , Prodigiosin/pharmacokinetics , Triple Negative Breast Neoplasms/metabolism , Adsorption , Antineoplastic Agents/administration & dosage , Cell Adhesion , Cell Line, Tumor , Combined Modality Therapy , Drug Delivery Systems , Female , Gold/chemistry , Humans , Hyperthermia, Induced/methods , Metal Nanoparticles/chemistry , Microscopy, Atomic Force , Prodigiosin/administration & dosage , Prodigiosin/chemistry , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/physiopathology , Triple Negative Breast Neoplasms/therapy
19.
Acta Biomater ; 59: 170-180, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28629893

ABSTRACT

It is a great challenge to combat multidrug resistant (MDR) cancer effectively. To address this issue, we developed a new near-infrared (NIR) triggered chemotherapeutic agent doxorubicin (DOX) and photosensitizer indocyanine green (ICG) co-release system by aid of NIR induced photothermal effect of gold nanocages (AuNCs) and temperature sensitive phase-change property of 1-tetradecanol at its melting point of 39°C, which could simultaneously exerted chemo/photothermal/photodynamic treatment on MDR human breast cancer MCF-7/ADR cells. This nano-sized system was constructed by filling the interior of AuNCs with DOX, ICG and 1-tetradecanol, and modifying the surface with biotinylated poly (ethylene glycol) via Au-S bonds, termed as DOX/ICG@biotin-PEG-AuNC-PCM. The DOX and ICG co-release from DOX/ICG@biotin-PEG-AuNC-PCM was much faster in PBS at 40°C or under 808nm NIR irradiation at 2.5W/cm2 than at 37°C (e.g. 67.27% or 80.31% vs. 5.57% of DOX, 76.08% vs. 3.83% of ICG for 20min). The flow cytometry and confocal laser scanning microscopy (CLSM) results showed, the AuNCs were taken up by MCF-7/ADR cells via endocytosis, thus enhancing DOX uptake; the biotin on AuNCs facilitated this endocytosis; NIR irradiation caused the heating of the AuNCs, triggering the DOX and ICG co-release and enhancing the distribution of DOX in nuclei, the released ICG generated ROS to take photodynamic therapy. Due to the above unique properties, DOX/ICG@biotin-PEG-AuNC-PCM exerted excellent anti-tumor effects under NIR irradiation, its IC50 against MCF-7/ADR cells was very low, only 0.48µg/mL, much smaller than that of free DOX (74.51µg/mL). STATEMENT OF SIGNIFICANCE: A new near-infrared (NIR) triggered chemotherapeutic agent doxorubicin (DOX) and photosensitizer indocyanine green (ICG) co-release system by aid of NIR induced photothermal effect of gold nanocages (AuNCs) and temperature sensitive phase-change property of 1-tetradecanol at its melting point of 39°C, was prepared, termed as DOX/ICG@biotin-PEG-AuNC-PCM, which could simultaneously exerted chemo/photothermal/photodynamic treatment on MDR human breast cancer MCF-7/ADR cells. DOX/ICG@biotin-PEG-AuNC-PCM exerted excellent anti-tumor effects under NIR irradiation, its IC50 against MCF-7/ADR cells was very low, only 0.48µg/mL, much smaller than that of free DOX (74.51µg/mL).


Subject(s)
Doxorubicin , Drug Delivery Systems/methods , Hyperthermia, Induced/methods , Indocyanine Green , Photochemotherapy/methods , Photosensitizing Agents , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Humans , Indocyanine Green/chemistry , Indocyanine Green/pharmacokinetics , Indocyanine Green/pharmacology , MCF-7 Cells , Metal Nanoparticles/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology
20.
Toxicol Sci ; 158(1): 23-35, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28402537

ABSTRACT

Many physiologically based pharmacokinetic (PBPK) models for environmental chemicals, drugs, and nanomaterials have been developed to aid risk and safety assessments using acslX. However, acslX has been rendered sunset since November 2015. Alternative modeling tools and tutorials are needed for future PBPK applications. This forum article aimed to: (1) demonstrate the performance of 4 PBPK modeling software packages (acslX, Berkeley Madonna, MATLAB, and R language) tested using 2 existing models (oxytetracycline and gold nanoparticles); (2) provide a tutorial of PBPK model code conversion from acslX to Berkeley Madonna, MATLAB, and R language; (3) discuss the advantages and disadvantages of each software package in the implementation of PBPK models in toxicology, and (4) share our perspective about future direction in this field. Simulation results of plasma/tissue concentrations/amounts of oxytetracycline and gold from different models were compared visually and statistically with linear regression analyses. Simulation results from the original models were correlated well with results from the recoded models, with time-concentration/amount curves nearly superimposable and determination coefficients of 0.86-1.00. Step-by-step explanations of the recoding of the models in different software programs are provided in the Supplementary Data. In summary, this article presents a tutorial of PBPK model code conversion for a small molecule and a nanoparticle among 4 software packages, and a performance comparison of these software packages in PBPK model implementation. This tutorial helps beginners learn PBPK modeling, provides suggestions for selecting a suitable tool for future projects, and may lead to the transition from acslX to alternative modeling tools.


Subject(s)
Gold/pharmacokinetics , Metal Nanoparticles/chemistry , Models, Biological , Oxytetracycline/pharmacokinetics , Animals , Dogs , Gold/blood , Gold/chemistry , Oxytetracycline/blood , Swine , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL