Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Colloid Interface Sci ; 670: 297-310, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38763026

ABSTRACT

Fenton/Fenton-like reaction induced chemical dynamic therapy (CDT) has been widely recognized in tumor therapy. Due to the low efficiency of conversion from high-valent metal ions (M(n+1)+) to low-valent ions (Mn+) in the Fenton/Fenton-like catalytic process, enhancing the conversion efficiency safely and effectively would create a great opportunity for the clinical application of CDT. In the study, a universal nanoreactor (NR) consisting of liposome (Lip), tumor cell membrane (CM), and bis(2,4,5-trichloro-6-carboxyphenyl) oxalate (CPPO) is developed to tackle this challenge. The CPPO was first discovered to decompose under weak acidity and H2O2 conditions to generate carboxylic acids (R'COOH) and alcohols (R'OH) with reducibility, which will reduce M(n+1)+ to Mn+ and magnify the effect of CDT. Furthermore, glucose oxidase (GOx) was introduced to decompose glucose in tumor and generate H2O2 and glucose acid, which promote the degradation of CPPO, further strengthening the efficiency of CDT, leading to a butterfly effect. This demonstrated that the butterfly effect triggered by NR and GOx encourages Fenton/Fenton-like reactions of Fe3O4 and MoS2, thereby enhancing the tumor inhibition effect. The strategy of combining GOx and CPPO to strengthen the Fenton/Fenton-like reaction is a universal strategy, which provides a new and interesting perspective for CPPO in the application of CDT, reflecting the exquisite integration of Fenton chemistry and catalytic medicine.


Subject(s)
Hydrogen Peroxide , Hydrogen Peroxide/chemistry , Humans , Iron/chemistry , Liposomes/chemistry , Glucose Oxidase/chemistry , Glucose Oxidase/metabolism , Animals , Surface Properties , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Oxalates/chemistry , Mice , Particle Size , Cell Survival/drug effects
2.
J Nanobiotechnology ; 22(1): 227, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711078

ABSTRACT

BACKGROUND: Elevated interstitial fluid pressure within tumors, resulting from impaired lymphatic drainage, constitutes a critical barrier to effective drug penetration and therapeutic outcomes. RESULTS: In this study, based on the photosynthetic characteristics of algae, an active drug carrier (CP@ICG) derived from Chlorella pyrenoidosa (CP) was designed and constructed. Leveraging the hypoxia tropism and phototropism exhibited by CP, we achieved targeted transport of the carrier to tumor sites. Additionally, dual near-infrared (NIR) irradiation at the tumor site facilitated photosynthesis in CP, enabling the breakdown of excessive intratumoral interstitial fluid by generating oxygen from water decomposition. This process effectively reduced the interstitial pressure, thereby promoting enhanced perfusion of blood into the tumor, significantly improving deep-seated penetration of chemotherapeutic agents, and alleviating tumor hypoxia. CONCLUSIONS: CP@ICG demonstrated a combined effect of photothermal/photodynamic/starvation therapy, exhibiting excellent in vitro/in vivo anti-tumor efficacy and favorable biocompatibility. This work provides a scientific foundation for the application of microbial-enhanced intratumoral drug delivery and tumor therapy.


Subject(s)
Chlorella , Drug Carriers , Photosynthesis , Animals , Mice , Cell Line, Tumor , Drug Carriers/chemistry , Humans , Combined Modality Therapy , Photochemotherapy/methods , Neoplasms/therapy , Antineoplastic Agents/pharmacology , Mice, Inbred BALB C , Drug Delivery Systems/methods , Indocyanine Green/pharmacokinetics , Indocyanine Green/chemistry , Female
3.
J Colloid Interface Sci ; 663: 1064-1073, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38458046

ABSTRACT

Doxorubicin (DOX) is widely used in clinic as a broad-spectrum chemotherapy drug, which can enhance the efficacy of chemodynamic therapy (CDT) by interfering tumor-related metabolize to increase H2O2 content. However, DOX can induce serious cardiomyopathy (DIC) due to its oxidative stress in cardiomyocytes. Eliminating oxidative stress would create a significant opportunity for the clinical application of DOX combined with CDT. To address this issue, we introduced sodium ascorbate (AscNa), the main reason is that AscNa can be catalyzed to produce H2O2 by the abundant Fe3+ in the tumor site, thereby enhancing CDT. While the content of Fe3+ in heart tissue is relatively low, so the oxidation of AscNa had tumor specificity. Meanwhile, due to its inherent reducing properties, AscNa could also eliminate the oxidative stress generated by DOX, preventing cardiotoxicity. Due to the differences between myocardial tissue and tumor microenvironment, a novel nanomedicine was designed. MoS2 was employed as a carrier and CDT catalyst, loaded with DOX and AscNa, coating with homologous tumor cell membrane to construct an acid-responsive nanomedicine MoS2-DOX/AscNa@M (MDA@M). In tumor cells, AscNa enhances the synergistic therapy of DOX and MoS2. In cardiomyocytes, AscNa could effectively reduce the cardiomyopathy induced by DOX. Overall, this study enhanced the clinical potential of chemotherapy synergistic CDT.


Subject(s)
Cardiomyopathies , Neoplasms , Humans , Cardiotoxicity/drug therapy , Cardiotoxicity/etiology , Cardiotoxicity/prevention & control , Nanomedicine , Hydrogen Peroxide/metabolism , Molybdenum/metabolism , Doxorubicin/pharmacology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Cardiomyopathies/chemically induced , Cardiomyopathies/drug therapy , Cardiomyopathies/pathology , Ascorbic Acid/pharmacology , Cell Line, Tumor , Neoplasms/metabolism , Tumor Microenvironment
4.
ACS Nano ; 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38330150

ABSTRACT

The practical efficacy of nanomedicines for treating solid tumors is frequently low, predominantly due to the elevated interstitial pressure within such tumors that obstructs the penetration of nanomedicines. This increased interstitial pressure originates from both liquid and solid stresses related to an undeveloped vascular network and excessive fibroblast proliferation. To specifically resolve the penetration issues of nanomedicines for tumor treatment, this study introduces a holistic "dual-faceted" approach. A treatment platform predicated on the WS2/Pt Schottky heterojunction was adopted, and flexocatalysis technology was used to disintegrate tumor interstitial fluids, thus producing oxygen and reactive oxygen species and effectively mitigating the interstitial fluid pressure. The chemotherapeutic agent curcumin was incorporated to further suppress the activity of cancer-associated fibroblasts, minimize collagen deposition in the extracellular matrix, and alleviate solid stress. Nanomedicines achieve homologous targeting by enveloping the tumor cell membrane. It was found that this multidimensional strategy not only alleviated the high-pressure milieu of the tumor interstitium─which enhanced the efficiency of nanomedicine delivery─but also triggered tumor cell apoptosis via the generated reactive oxygen species and modulated the tumor microenvironment. This, in turn, amplified immune responses, substantially optimizing the therapeutic impacts of nanomedicines.

5.
Nanoscale Horiz ; 9(3): 365-383, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38230559

ABSTRACT

Precision drug delivery and multimodal synergistic therapy are crucial in treating diverse ailments, such as cancer, tissue damage, and degenerative diseases. Electrodes that emit electric pulses have proven effective in enhancing molecule release and permeability in drug delivery systems. Moreover, the physiological electrical microenvironment plays a vital role in regulating biological functions and triggering action potentials in neural and muscular tissues. Due to their unique noncentrosymmetric structures, many 2D materials exhibit outstanding piezoelectric performance, generating positive and negative charges under mechanical forces. This ability facilitates precise drug targeting and ensures high stimulus responsiveness, thereby controlling cellular destinies. Additionally, the abundant active sites within piezoelectric 2D materials facilitate efficient catalysis through piezochemical coupling, offering multimodal synergistic therapeutic strategies. However, the full potential of piezoelectric 2D nanomaterials in drug delivery system design remains underexplored due to research gaps. In this context, the current applications of piezoelectric 2D materials in disease management are summarized in this review, and the development of drug delivery systems influenced by these materials is forecast.


Subject(s)
Nanostructures , Nanostructures/therapeutic use , Nanostructures/chemistry , Electricity , Mechanical Phenomena , Drug Delivery Systems
6.
Small ; : e2309487, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38197548

ABSTRACT

Cellular senescence, a vulnerable state of growth arrest, has been regarded as a potential strategy to weaken the resistance of tumor cells, leading to dramatic improvements in treatment efficacy. However, a selective and efficient strategy for inducing local tumor cellular senescence has not yet been reported. Herein, piezoelectric catalysis is utilized to reduce intracellular NAD+ to NADH for local tumor cell senescence for the first time. In detail, a biocompatible nanomedicine (BTO/Rh-D@M) is constructed by wrapping the piezoelectric BaTiO3 /(Cp* RhCl2 )2 (BTO/Rh) and doxorubicin (DOX) in the homologous cytomembrane with tumor target. After tumors are stimulated by ultrasound, negative and positive charges are generated on the BTO/Rh by piezoelectric catalysis, which reduce the intracellular NAD+ to NADH for cellular senescence and oxidize H2 O to reactive oxygen species (ROS) for mitochondrial damage. Thus, the therapeutic efficacy of tumor immunogenic cell death-induced chemo-immunotherapy is boosted by combining cellular senescence, DOX, and ROS. The results indicate that 23.9% of the piezoelectric catalysis-treated tumor cells senesced, and solid tumors in mice disappeared completely after therapy. Collectively, this study highlights a novel strategy to realize cellular senescence utilizing piezoelectric catalysis and the significance of inducing tumor cellular senescence to improve therapeutic efficacy.

7.
Article in English | MEDLINE | ID: mdl-38016813

ABSTRACT

In nanocatalytic medicine, drugs can be transformed into toxic components through highly selective and highly specific catalytic reactions in the tumor microenvironment, avoiding toxic side effects on normal tissues. Due to the coexistence of Ce3+ and Ce4+, CeO2 is endowed with dual nanozyme activities. Herein, CeO2 nanoparticles served as templates to construct a biomimetic nanodrug delivery system (C/CeO2@M) by electrostatic adsorption of carbon quantum dots (CQDs) and coating a homologous tumor cytomembrane. After homologous targeting to tumors, the CQDs emitted 350-600 nm light under 660 nm laser irradiation by upconversion luminescence, which caused a CeO2-mediated photocatalytic reaction to generate reactive oxygen species. The catalase-like activity of CeO2-enabled converting excess H2O2 to O2, which not only alleviated tumor hypoxia and promoted intratumor drug delivery but also provided substrates for subsequent catalytic reactions. Meanwhile, the phosphatase activity of CeO2 could consume adenosine triphosphate (ATP) to block the energy supply for tumor cells, thus limiting cell proliferation and metastasis. The strategy of energy restriction and photocatalysis of dual nanozyme stimulation offers great potentials in enhancing drug penetration and eradicating solid tumors.

8.
World J Gastroenterol ; 29(4): 670-681, 2023 Jan 28.
Article in English | MEDLINE | ID: mdl-36742173

ABSTRACT

Colon cancer has attracted much attention due to its annually increasing incidence. Conventional chemotherapeutic drugs are unsatisfactory in clinical application because of their lack of targeting and severe toxic side effects. In the past decade, nanomedicines with multimodal therapeutic strategies have shown potential for colon cancer because of their enhanced permeability and retention, high accumulation at tumor sites, co-loading with different drugs, and comb-ination of various therapies. This review summarizes the advances in research on various nanomedicine-based therapeutic strategies including chemotherapy, radiotherapy, phototherapy (photothermal therapy and photodynamic therapy), chemodynamic therapy, gas therapy, and immunotherapy. Additionally, the therapeutic mechanisms, limitations, improvements, and future of the above therapies are discussed.


Subject(s)
Colonic Neoplasms , Neoplasms , Photochemotherapy , Humans , Neoplasms/therapy , Nanomedicine , Phototherapy , Drug Delivery Systems , Colonic Neoplasms/drug therapy
9.
ACS Nano ; 16(11): 18376-18389, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36355037

ABSTRACT

Currently, one of the main reasons for the ineffectiveness of tumor treatment is that the abnormally high tumor interstitial pressure (TIP) hinders the delivery of drugs to the tumor center and promotes intratumoral cell survival and metastasis. Herein, we designed a "nanomotor" by in situ growth of Ag2S nanoparticles on the surface of ultrathin WS2 to fabricate Z-scheme photocatalytic drug AWS@M, which could rapidly enter tumors by splitting water in interstitial liquid to reduce TIP, along with O2 generation. Moreover, the O2 would be further converted to reactive oxygen species (ROS), accompanied by increased local temperature of tumors, and the combination of ROS with thermotherapy could eliminate the deep tumor cells. Therefore, the "nanomotor'' could effectively reduce the TIP levels of cervical cancer and pancreatic cancer (degradation rates of 40.2% and 36.1%, respectively) under 660 nm laser irradiation, further enhance intratumor drug delivery, and inhibit tumor growth (inhibition ratio 95.83% and 87.61%, respectively), and the related mechanism in vivo was explored. This work achieves efficiently photocatalytic water-splitting in tumor interstitial fluid to reduce TIP by the nanomotor, which addresses the bottleneck problem of blocking of intratumor drug delivery, and provides a general strategy for effectively inhibiting tumor growth.


Subject(s)
Hyperthermia, Induced , Nanoparticles , Neoplasms , Humans , Reactive Oxygen Species/metabolism , Drug Delivery Systems , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Water , Cell Line, Tumor
10.
Biomaterials ; 290: 121816, 2022 11.
Article in English | MEDLINE | ID: mdl-36201946

ABSTRACT

The absence of lymphatic vessels in tumors leads to the retention of interstitial fluid, and the formation of an inverse pressure difference between the tumor and blood vessels hinders drug delivery deep into the tumor, which leads to tumor recurrence and metastasis. Therefore, we designed a novel strategy to downregulate tumor interstitial fluid pressure (TIFP) by water splitting in the tumor interstitium based on piezoelectric catalysis nanomedicine. First, the chemotherapeutic drug doxorubicin (DOX) was loaded on the piezoelectric catalytic material MoS2 and then encapsulated with tumor cell membrane (CM) to obtain MD@C. MD@C could not only target the tumor through homologous targeting but, more importantly, also triggered piezoelectric catalytic water splitting under ultrasound (US) stimulation; as a result, the TIFPs of U14 and PAN02 tumor-bearing mice were reduced to 57.14% and 45.5%, respectively, and the tumor inhibition rates of MD@C were 96.75% and 99.21%, which increased the perfusion of blood-derived drugs in the tumors. Moreover, the hydroxyl radicals generated by piezoelectric catalysis could effectively inhibit the growth of tumors in combination with DOX. Consequently, the piezoelectric catalytic water splitting strategy of MD@C can enhance drug delivery, providing a new universal platform for the treatment of solid malignant tumors.


Subject(s)
Nanoparticles , Neoplasms , Mice , Animals , Molybdenum , Doxorubicin/therapeutic use , Doxorubicin/pharmacology , Nanomedicine , Neoplasms/drug therapy , Neoplasms/pathology , Catalysis , Water , Cell Line, Tumor , Nanoparticles/therapeutic use
11.
J Mater Chem B ; 9(18): 3925-3934, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33942817

ABSTRACT

Nanozymes, as a kind of artificial mimic enzymes, have superior catalytic capacity and stability. As lack of O2 in tumor cells can cause resistance to drugs, we designed drug delivery liposomes (MnO2-PTX/Ce6@lips) loaded with catalase-like nanozymes of manganese dioxide nanoparticles (MnO2 NPs), paclitaxel (PTX) and chlorin e6 (Ce6) to consume tumor's native H2O2 and produce O2. Based on the catalysis of MnO2 NPs, a large amount of oxygen was produced by MnO2-PTX/Ce6@lips to burst the liposomes and achieve a responsive release of the loaded drug (paclitaxel), and the released O2 relieved the chemoresistance of tumor cells and provided raw materials for photodynamic therapy. Subsequently, MnO2 NPs were decomposed into Mn2+ in an acidic tumor environment to be used as contrast agents for magnetic resonance imaging. The MnO2-PTX/Ce6@lips enhanced the efficacy of chemotherapy and photodynamic therapy (PDT) in bearing-tumor mice, even achieving complete cure. These results indicated the great potential of MnO2-PTX/Ce6@lips for the modulation of the TME and the enhancement of chemotherapy and PDT along with MRI tracing in the treatment of tumors.


Subject(s)
Nanostructures/therapeutic use , Neoplasms/drug therapy , Theranostic Nanomedicine , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Survival/drug effects , Chlorophyllides , Contrast Media/chemistry , Humans , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Light , Liposomes/chemistry , Magnetic Resonance Imaging , Manganese Compounds/chemistry , Mice , Nanoparticles/chemistry , Nanostructures/chemistry , Nanostructures/toxicity , Neoplasms/diagnostic imaging , Oxides/chemistry , Oxygen/chemistry , Oxygen/metabolism , Paclitaxel/chemistry , Paclitaxel/metabolism , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/metabolism , Photosensitizing Agents/therapeutic use , Porphyrins/chemistry
12.
ACS Nano ; 15(6): 10488-10501, 2021 06 22.
Article in English | MEDLINE | ID: mdl-34018736

ABSTRACT

Because of the deficiency of lymphatic reflux in the tumor, the retention of tumor interstitial fluid causes aggravation of the tumor interstitial pressure (TIP), which leads to unsatisfactory tumor penetration of nanomedicine. It is the main inducement of tumor recurrence and metastasis. Herein, we design a pyroelectric catalysis-based "Nano-lymphatic" to decrease the TIP for enhanced tumor penetration and treatments. It realizes photothermal therapy and decomposition of tumor interstitial fluid under NIR-II laser irradiation after reaching the tumor, which reduces the TIP for enhanced tumor penetration. Simultaneously, reactive oxygen species generated during the pyroelectric catalysis can further damage deep tumor stem cells. The results indicate that the "Nano-lymphatic" relieves 52% of TIP, leading to enhanced tumor penetration, which effectively inhibits the tumor proliferation (93.75%) and recurrence. Our finding presents a rational strategy to reduce TIP by pyroelectric catalysis for enhanced tumor penetration and improved treatments, which is of great significance for drug delivery.


Subject(s)
Nanoparticles , Neoplasms , Catalysis , Cell Line, Tumor , Drug Delivery Systems , Humans , Hydrodynamics , Neoplasms/drug therapy , Phototherapy
13.
Biomater Sci ; 9(6): 2313-2321, 2021 Mar 21.
Article in English | MEDLINE | ID: mdl-33556159

ABSTRACT

The gaseous microenvironment (GME) of tumors is rapidly becoming a new concern for nanotechnology-mediated oncotherapy. Here, we constructed a tumor/near-infrared (NIR) light-responsive nanoplatform to generate O2 and NO for remodeling the GME of tumors and phototherapy. The biocompatible and pyrolytic polydopamine was used to load indocyanine green, NONOate, and MnO2 NPs as a nanoenzyme (PINM). Then, HA was modified on the PINM to form the final nanoplatform (PINMH). PINMH can target tumors favorably due to the modification of HA. Under the NIR light irradiation, PINM converts the light and O2 to hyperpyrexia (58.5 °C) and cytotoxic 1O2. MnO2 NPs catalyze the H2O2 overexpressed in tumors to O2, which increases the amount of 1O2. Moreover, NONOate decomposes to NO (100 µM) under hyperpyrexia, thus leading to the gas therapy. The results verified that the responsive nanoplatform with precise gaseous regulation and phototherapy exhibited a superior anti-tumor effect (V/V0 = 1.2) and biosafety. In addition, PINMH can be tracked in real-time via magnetic resonance imaging. In this study, an intelligent nano-platform integrated with diagnosis and treatment was developed, which used the phototherapy technology to reshape GME and achieve good anti-tumor effects, aiming to provide an innovative and reasonable strategy for the development of tumor treatment.


Subject(s)
Hyperthermia, Induced , Nanoparticles , Gases , Hydrogen Peroxide , Manganese Compounds , Oxides
14.
Chem Commun (Camb) ; 56(72): 10533-10536, 2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32780059

ABSTRACT

Herein, cobalt-involved redox in a magnetically responsive drug-loaded nanocatalyst (PTX/Co-Lips@Fe3O4) was used to convert Fe(iii) to Fe(ii) for enhancing tumor ferrotherapy for the first time. Moreover, this work highlighted an "all in one" strategy: (1) targeting, chemotherapy, and ferrotherapy in one nanomedicine, and (2) a decrease in GSH quantity, increase in the quantity of efficient catalytic ions, and use of a magnetic field, all in one tumor ferrotherapy enhancement approach.


Subject(s)
Cobalt/chemistry , Drug Carriers/chemistry , Ferric Compounds/chemistry , Ferric Compounds/therapeutic use , Magnetic Phenomena , Nanomedicine/methods , Nanostructures/chemistry , Catalysis , Glutathione/metabolism , HeLa Cells , Humans , Oxidation-Reduction
15.
Nanomedicine (Lond) ; 15(9): 871-890, 2020 04.
Article in English | MEDLINE | ID: mdl-32223505

ABSTRACT

Aim: To synthesize a drug-delivery system with chemo-photothermal function and magnetic targeting, to validate its antitumor effect. Materials & methods: Parthenolide (PTL), employing chemotherapy and indocyanine green (ICG) providing phototherapy, were encased separately in the lipid and aqueous phases of liposomes (Lips). The Fe3O4 nanoparticles (MNPs), endowing magnetic targeting, were modified on the surface of Lips. The antitumor effects were investigated in vitro and in vivo. Results: ICG-PTL-Lips@MNPs showed outstanding synergistic antitumor efficacy in vitro and in vivo. Especially, after 14-day treatment, the tumor volumes decreased significantly and the biotoxicity was very low. Conclusion: The designed ICG-PTL-Lips@MNPs possess synergistic effects of chemotherapy, photothermal and targeting therapy, which are expected to provide an alternative way to further improve antitumor efficacy.


Subject(s)
Antineoplastic Agents , Indocyanine Green , Metal Nanoparticles , Photochemotherapy , Sesquiterpenes , Animals , Cell Line, Tumor , Ferric Compounds , Liposomes , Phototherapy
16.
ACS Appl Mater Interfaces ; 12(8): 8978-8988, 2020 Feb 26.
Article in English | MEDLINE | ID: mdl-32020804

ABSTRACT

Cells in the center of solid tumors have always been an abyss untouched by treatments because of their deep location and increased drug resistance. Herein, we designed a rational strategy for sequential intra-intercellular delivery of nanomedicine to deep sites of drug-resistant solid tumors. In our formulation, dopamine and hemoglobin were polymerized to form a smart nanocarrier (PDA/Hb). Subsequently, the doxorubicin and nitric oxide donor were connected on the surface of PDA/Hb to obtain D/N-PDA/Hb. Ultimately, the hyaluronic acid was combined with D/N-PDA/Hb to form D/N-PDA/Hb@HA. Concretely, acidic and neutral environments of tumor cells were treated as a switch to turn on or off the drug release of a nanodrug. Meanwhile, the generation of nitric oxide in situ was exploited to favor the lysosomal escape of nanocarriers and overcome the drug resistance of deep solid tumor cells. The results indicated that the nanodrug based on sequential intra-intercellular delivery showed exciting penetration efficiency and resistance reversal of solid tumors. Conventional nanodrug delivery was highly dependent on the enhanced permeability and retention (EPR) effect and limited by tumorous interstitial fluid pressure. Plenty of drugs stayed on the surface of solid tumors, and the infiltrated drugs were inefficient due to strict resistance. To conquer this dilemma, this work proposed a new mechanism reversing the EPR effect for drug delivery, leading to better penetration and resistance reversal of solid tumors.


Subject(s)
Doxorubicin , Drug Carriers , Drug Resistance, Neoplasm/drug effects , Nanoparticles , Neoplasms, Experimental/drug therapy , Animals , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacology , Female , HeLa Cells , Humans , Mice , Nanomedicine , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Xenograft Model Antitumor Assays
17.
Nanotechnology ; 31(3): 035603, 2020 Jan 17.
Article in English | MEDLINE | ID: mdl-31557747

ABSTRACT

A key challenge in developing an ethanol oxidation reaction is nontoxic fabrication of highly active stable and low-cost catalysts. Here we design a green synthetic strategy of AgPd bimetallic nanosphere by a dual-template cascade method. The Pd nanoshell is firstly prepared using Vapreotide acetate as a primary template, and then the Ag nanoshell acts as a secondary template for the distribution of AgPd alloy nanoparticles. The AgPd nanoparticles have core-shell structures and various sizes, and their shell thicknesses are tuned by controlling the amount of PdCl2. The six different samples are prepared, named AgPd-1, AgPd-2, AgPd-3, AgPd-4, AgPd-5, and AgPd-6, respectively. The mass current density of AgPd-5, is higher 3.87 times that of commercial Pd/C, and exhibits the best ethanol oxidation reaction activity and long-term stability. The main reasons are that the AgPd-5 possessed excellent specific surface area due to their rough structure, and Ag can remove more CO-like species. This is the first time a Vapreotide acetate/Ag-template method has been used to synthesize a AgPd core-shell structure, which would have broad application prospects for direct ethanol fuel cells.

18.
Theranostics ; 9(10): 2897-2909, 2019.
Article in English | MEDLINE | ID: mdl-31244931

ABSTRACT

Inaccessibility of deep-seated malignant cells in the central region of tumors and uncontrollable tumor recurrence represent a significant challenge for conventional synergistic cancer therapy. Herein, we designed a novel nanoplatform based on hierarchical drug release for deep cascade cancer therapy including localized photothermal therapy, systematic chemotherapy, and elicited immune responses. Methods: The first-step chemotherapy could be carried out by polydopamine (PDA) releasing doxorubicin (DOX) in the specific microenvironment of lysosomes (pH 5.5). The branched gold nanoshells and PDA converted the light to heat efficiently to accomplish the second-step photothermal therapy and collapsed biomimetic vesicles (BVs) to release paclitaxel (PTX), which promoted the third-step of chemotherapy and triggered immune responses. Results: After 10 days of treatment, there were no obvious residual tumors in tumor-bearing mice. Significantly, 10 days after stopping treatment, mice in the drug immune-therapeutic group showed little tumor recurrence (1.5 times) compared to substantial recurrence (20 times) in the conventional treatment group. Conclusion: The hierarchical drug release and cascade therapeutic modality enhance the penetration of drugs deep into the tumor tissue and effectively inhibit recurrence. This cascade therapeutic modality provides a novel approach for more effective cancer therapy.


Subject(s)
Carcinoma/therapy , Drug Delivery Systems , Drug Therapy/methods , Hyperthermia, Induced/methods , Immunotherapy/methods , Nanocomposites/administration & dosage , Phototherapy/methods , Animals , Antineoplastic Agents/administration & dosage , Disease Models, Animal , Doxorubicin/administration & dosage , Drug Liberation , Gold/administration & dosage , HeLa Cells , Humans , Immunologic Factors/administration & dosage , Mice , Models, Biological , Neoplasm Transplantation , Paclitaxel/administration & dosage , Photosensitizing Agents/administration & dosage , Transplantation, Heterologous , Treatment Outcome
19.
J Mater Chem B ; 7(4): 598-610, 2019 01 28.
Article in English | MEDLINE | ID: mdl-32254793

ABSTRACT

With the rapid development of photothermal therapy (PTT) in cancer treatment, it is necessary to obtain effective plasma-responsive tunable photothermal transducing agents. Inspired by the peptide-directed hierarchical mineralized Ag nanocages (Ag NCs), scientists designed a new duel-template cascade preparation method, and novel unique multi-branched gold nanoshells (BGSs) were successfully prepared under mild conditions using green strategy. The length, density and diameter of the branches were tuned, which led to the adjustment of the surface plasma response of the nanostructure. Because of the hierarchical structure and anisotropic surface, an obvious red shift of the local surface plasmon resonance spectrum was observed for the branched Au nanoshells. The excellent photothermal conversion efficiency (70.9%) and photo-induced heating responsive curves proved the superior photothermal conversion performance and photothermal stability of BGSs. The in vitro and in vivo results indicated that the heat generated by the intense NIR absorption of BGSs can selectively destroy cancer cells under laser irradiation. The nanostructures with ultrastrong absorption have promising prospects in tumor therapy.


Subject(s)
Nanoshells , Neoplasms/therapy , Phototherapy/methods , Animals , Cell Survival/drug effects , Gold/chemistry , HEK293 Cells , HeLa Cells , Humans , Mice , Nanoshells/chemistry , Nanoshells/therapeutic use , Physical Phenomena , Silver/chemistry
20.
Nanomedicine ; 15(1): 142-152, 2019 01.
Article in English | MEDLINE | ID: mdl-30300749

ABSTRACT

A multifunctional antitumor drug delivery system was synthesized based on graphene oxide (GO) for near-infrared (NIR) light controlling chemotherapeutic/photothermal (PTT) /photodynamic (PDT) trimodal synergistic therapy. The system named ICG-Wed-GO was formed by co-loading wedelolactone (Wed) and indocyanine green (ICG) on the surface of GO through π-π stacking interaction. Under NIR laser irradiation, ICG-Wed-GO could effectively absorb and transform optical energy to heat, generate reactive oxygen species (ROS) to ablating and damage tumor cells. The temperature of ICG-Wed-GO solution reached up to 79.4 °C in 10 min with NIR irradiation. In in vitro and in vivo study, ICG-Wed-GO showed excellent antitumor effect. After 14-day treatment of ICG-Wed-GO with NIR laser irradiation, the tumor disappeared completely on tumor-bearing mice. The low biotoxicity of ICG-Wed-GO was also proved. The system achieved the synergistic trimodal chemotherapeutic/photothermal/photodynamic treatment and demonstrated excellent antitumor effect, which is expected to have a greater potential for cancer therapy.


Subject(s)
Coumarins/chemistry , Drug Delivery Systems , Graphite/chemistry , Hyperthermia, Induced , Indocyanine Green/administration & dosage , Photochemotherapy , Uterine Cervical Neoplasms/therapy , Animals , Coloring Agents/administration & dosage , Coloring Agents/chemistry , Female , Indocyanine Green/chemistry , Mice , Reactive Oxygen Species , Tumor Cells, Cultured , Uterine Cervical Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...