Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Biomater Sci ; 11(22): 7327-7338, 2023 Nov 07.
Article in English | MEDLINE | ID: mdl-37847063

ABSTRACT

Local radio-therapy combined with immunotherapy has attracted great interest in controlling local tumors. In this study, we have developed membrane-cloaked manganese dioxide nanoparticles displaying anti-PD-L1 antibodies as targeted immuno-radio-enhancers. Mediated by anti-PD-L1 antibodies (aPD-L1) expressed on cell membranes, this kind of membrane-coated nanosystem can selectively deliver cytosine-phosphate-guanine (CpG)-loaded MnO2 nanoparticles (NPs) and induce systemic anti-tumor immunities, thereby achieving favorable immuno/radio-therapeutic outcomes. Through expressing various functional proteins onto cellular membranes, the new class of membrane-camouflaged nanovehicles can be endowed with a wide variety of artificial functionalities such as enzymatic catalytic capabilities and specific targeting. This versatile nanoplatform, in general, enables the targeted delivery of theranostics, opening a new avenue for personalized nanomedicine.


Subject(s)
Nanoparticles , Neoplasms , Humans , Manganese Compounds/therapeutic use , Nanomedicine , Oxides/therapeutic use , Neoplasms/drug therapy , Drug Carriers/therapeutic use , Immunotherapy , Cell Line, Tumor
2.
J Mater Chem B ; 11(42): 10108-10120, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37853796

ABSTRACT

Bacterial biofilm-associated infectious diseases remain serious menaces to human health. Recently, photodynamic therapy (PDT) has become a prospective strategy for combating biofilm infection. However, anaerobic conditions in a biofilm greatly inhibit its therapeutic efficacy. Here, a nanozyme-reinforced injectable hydrogel is prepared using Ca2+-crosslinked sodium alginate incorporated with photosensitizer-loaded MnO2 nanosheets and CaO2 nanoparticles for O2 self-sufficient PDT to eradicate biofilm infection. In our design, CaO2 reacts with water to produce locally concentrated H2O2, which could be catalyzed by MnO2 nanosheets (catalase-mimic nanozymes) to generate O2 and greatly relieve the hypoxic conditions in the biofilm, thus significantly strengthening PDT efficacy. In vitro assays confirmed that the hybrid hydrogel not only exhibits high-performance bactericidal activity in combating both Gram-positive Staphylococcus aureus and Gram-negative Escherichia coli but also shows great efficacy in eliminating biofilm infection. Moreover, benefiting from its good syringeability, the hybrid hydrogel is prone to fit irregular wounds and exhibits high efficiency in promoting wound healing in a biofilm-infected mice model. Besides, no obvious toxicity is detected in the hybrid hydrogel. Overall, we envision that our designed hydrogel could provide a prospective solution for combating biofilm-associated infections.


Subject(s)
Photochemotherapy , Staphylococcal Infections , Mice , Animals , Humans , Hydrogels/pharmacology , Hydrogels/therapeutic use , Manganese Compounds/therapeutic use , Hydrogen Peroxide/therapeutic use , Oxides/therapeutic use , Staphylococcal Infections/drug therapy , Biofilms
3.
J Mater Chem B ; 11(29): 6889-6895, 2023 07 26.
Article in English | MEDLINE | ID: mdl-37377123

ABSTRACT

Nanozyme-based tumour catalytic therapy has attracted widespread attention in recent years, but the therapeutic efficacy is limited due to the trapping of hydroxyl radicals (˙OH) by endogenous glutathione (GSH) in the tumour microenvironment (TME). Zr/Ce-MOFs/DOX/MnO2 is constructed in this work to serve as a new kind of nanozyme for combination chemotherapy and catalytic treatment. Zr/Ce-MOFs can produce ˙OH in a mimic TME, and the MnO2 on the surface could deplete the GSH, further promoting the ˙OH generation. The pH/GSH dual stimulation accelerates the release of anticancer drug doxorubicin (DOX) in tumour tissue for enhanced tumour chemotherapy. Moreover, Mn2+ produced by the reaction of Zr/Ce-MOFs/DOX/MnO2 and GSH can be used as the contrast agent for T1-MRI. The potential antitumour effect of Zr/Ce-MOFs/DOX/MnO2 is demonstrated by in vitro and in vivo cancer treatment tests. This work thus provides a new nanozyme-based platform for enhanced combination chemotherapy and catalytic treatment for tumours.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Manganese Compounds/pharmacology , Manganese Compounds/therapeutic use , Oxides/pharmacology , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Doxorubicin , Tumor Microenvironment
4.
Pharm Res ; 40(5): 1271-1282, 2023 May.
Article in English | MEDLINE | ID: mdl-36991228

ABSTRACT

PURPOSE: The effect of monotherapy in cancer is frequently influenced by the tumor's unique hypoxic microenvironment, insufficient drug concentration at the treatment site, and tumour cells' increased drug tolerance. In this work, we expect to design a novel therapeutic nanoprobe with the ability to solve these problems and improve the efficacy of antitumor therapy. METHODS: We have prepared a hollow manganese dioxide nanoprobes loaded with photosensitive drug IR780 for the photothermal/photodynamic/chemodynamic co-therapy of liver cancer. RESULTS: The nanoprobe demonstrates efficient thermal transformation ability under a single laser irradiation, and under the synergistic influence of photo heat, accelerates the Fenton/ Fenton-like reaction efficiency based on Mn2+ ions to produce more ·OH under the synergistic effect of photo heat. Moreover, the oxygen released under the degradation of manganese dioxide further promotes the ability of photosensitive drugs to produce singlet oxygen (ROS). The nanoprobe has been found to efficiently destroy tumour cells in vivo and in vitro experiments when used in combination with photothermal/photodynamic/ chemodynamic modes of treatment under laser irradiation. CONCLUSION: In all, this research shows that a therapeutic strategy based on this nanoprobe could be a viable alternative for cancer treatment in the near future.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Nanoparticles , Neoplasms , Photochemotherapy , Humans , Manganese Compounds/pharmacology , Manganese Compounds/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Oxides , Liver Neoplasms/drug therapy , Neoplasms/drug therapy , Cell Line, Tumor , Tumor Microenvironment
5.
Angew Chem Int Ed Engl ; 62(23): e202302525, 2023 06 05.
Article in English | MEDLINE | ID: mdl-36930411

ABSTRACT

Carbon monoxide (CO) is an endogenous signaling molecule with broad therapeutic effects. Here, a multifunctional X-ray-triggered carbon monoxide (CO) and manganese dioxide (MnO2 ) generation nanoplatform based on metal carbonyl and scintillating nanoparticles (SCNPs) is reported. Attributed to the radioluminescent characteristic of SCNPs, UV-responsive Mn2 (CO)10 is not only indirectly activated to release CO by X-ray but can also be degraded into MnO2 . A high dose of CO can be used as a glycolytic inhibitor for tumor suppression; it will also sensitize tumor cells to radiotherapy. Meanwhile MnO2 , as the photolytic byproduct of Mn2 (CO)10 , has both glutathione (GSH) depletion and Fenton-like Mn2+ delivery properties to produce highly toxic hydroxyl radical (⋅OH) in tumors. Thus, this strategy can realize X-ray-activated CO release, GSH depletion, and ⋅OH generation for cascade cancer radiosensitization. Furthermore, X-ray-activated Mn2+ in vivo demonstrates an MRI contrast effect, making it a potential theranostic nanoplatform.


Subject(s)
Nanoparticles , Neoplasms , Humans , Manganese Compounds/pharmacology , Manganese Compounds/therapeutic use , Oxides/pharmacology , Carbon Monoxide/pharmacology , Carbon Monoxide/therapeutic use , X-Rays , Neoplasms/drug therapy , Neoplasms/metabolism , Cell Line, Tumor , Glutathione/metabolism , Hydrogen Peroxide/therapeutic use
6.
ACS Appl Bio Mater ; 5(12): 5865-5876, 2022 12 19.
Article in English | MEDLINE | ID: mdl-36410719

ABSTRACT

Immunogenic cell death (ICD) induced by treatment modalities like chemotherapy, radiotherapy, and photothermal and photodynamic therapy has shown great potential to improve the low response rate of various solid tumors in cancer immunotherapy. However, extensive studies have revealed that the efficacy of cancer treatment is limited by the hypoxia and immunosuppression in the tumor microenvironment (TME). To address these challenges, a hypoxia alleviated and one phototriggered thermal/dynamic nanoplatform based on MnO2@PDA/ICG-BSA (MPIB) is developed for oxygen (O2) self-supply enhanced cancer phototherapy (PT). First, MnO2 transfers intracellular overexpression H2O2 into O2 in the acidic TME through its catalase-like activity to improve the hypoxia and also provide O2 for the following photodynamic therapy. Then, under single NIR-808 nm light irradiation (called the "phototherapeutic window"), excellent photothermal and photodynamic performance of the MPIB is activated for combined PT. Finally, assisted with immune adjuvant cytosine-phospho-guanine, obvious ICD and systemic antitumor immunity was elicited in PT-treated mice and demonstrated significant growth inhibition on distant tumors. This MPIB-based nanoplatform highlights the promise to overcome the limitations of hypoxia and also challenges of immunosuppressive tumor microenvironments for improved cancer immunotherapy.


Subject(s)
Manganese Compounds , Neoplasms , Mice , Animals , Manganese Compounds/therapeutic use , Immunogenic Cell Death , Hydrogen Peroxide/therapeutic use , Oxides/therapeutic use , Immunotherapy , Neoplasms/therapy , Oxygen/therapeutic use , Hypoxia/therapy , Tumor Microenvironment
7.
J Control Release ; 352: 793-812, 2022 12.
Article in English | MEDLINE | ID: mdl-36343761

ABSTRACT

As a promising cancer treatment, photodynamic therapy (PDT) still achieved limited clinical success due to the severe hypoxia and programmed death ligand-1 (PD-L1) over-expressed immunosuppression tumor microenvironment. At present, few methods have been proven to solve these two defects simply and effectively by a single drug or nano-system simultaneously. To ameliorate this situation, we designed and constructed MB@Bu@MnO2 nanoparticles with two-step oxygen regulation ability and PD-1/PD-L1 axis cascade-disruption capacity via a biomineralization method. In such a nanosystem, manganese dioxide albumin (MnO2@Alb) was used as the drug carrier, Butformin (Bu) as mitochondria-associated oxidative phosphorylation (OXPHOS) disruption agent with PD-L1 depression and oxygen reversion ability, and methylene blue (MB) as PDT drug with programmed cell death protein 1 (PD-1) inhibition capacity. Owing to the tumor-responsive capacity of MB@Bu@MnO2 nanoparticles, Bu and MB were selectively delivered and released in tumors. Then, the tumor hypoxia was dramatically reversed by Bu inhibited oxygen consumption, and MnO2 improved oxygen generation. Following this, the reactive oxygen species (ROS) generation was enhanced by MB@Bu@MnO2 nanoparticles mediated PDT owing to the reversed tumor hypoxia. Furthermore, the immunosuppression microenvironment was also obviously reversed by MB@Bu@MnO2 nanoparticles enhanced immunogenic cell death (ICD) and PD-1/PD-L1 axis cascade-disruption, which then enhanced T cell infiltration and improved its tumor cell killing ability. Finally, the growth of solid tumors was significantly depressed by MB@Bu@MnO2 nanoparticles mediated PDT. All in all, this well-designed nanosystem could solve the defects of traditional PDT via PD-1/PD-L1 axis dual disruption and reversing tumor hypoxia by two-step oxygen regulation.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Humans , B7-H1 Antigen , Cell Line, Tumor , Depression , Hypoxia/drug therapy , Manganese Compounds/therapeutic use , Methylene Blue/therapeutic use , Neoplasms/drug therapy , Oxides/therapeutic use , Oxygen/metabolism , Photosensitizing Agents/therapeutic use , Programmed Cell Death 1 Receptor
8.
J Mater Chem B ; 10(22): 4274-4284, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35583909

ABSTRACT

Specific tumor-responsive capabilities and efficient synergistic therapeutic performance are the keys to effective tumor treatment. Herein, AuNRs@SiO2-RB@MnO2 was developed as a new type of tumor-responsive nanotheranostic for multimodal imaging and synergistic chemodynamic/photothermal therapy. In AuNRs@SiO2-RB@MnO2, the SiO2 layer wraps the AuNRs, providing light absorption in the second near-infrared (NIR-II) region. The SiO2 layer also adsorbs the MnO2 nanosheets, which have Fenton-like activity, resulting in a fluorescent sensing platform based on the fluorescence quenching properties of MnO2 for rhodamine B dye. The fluorescence can be recovered by the consumption of MnO2 by glutathione, which simultaneously produces Mn2+ in the tumor region. The recovery of fluorescence reflects the consumption of glutathione and the increase in Mn2+, which produces hydroxyl radicals via Fenton-like reaction in the tumor microenvironment to realize chemodynamic therapy. Meanwhile, the AuNRs are a good photothermal reagent that can effectively absorb NIR-II light and convert it into heat energy to kill tumor cells via photothermal therapy. The NIR-II absorption performance of the AuNRs provides good photoacoustic imaging and deep photothermal performance, which is favorable for efficient NIR-II photoacoustic imaging-guided photothermal therapy. As a result, the AuNRs@SiO2-RB@MnO2 nanotheranostic exhibits outstanding imaging and synergistic chemodynamic/photothermal therapeutic performance for tumor imaging and treatment.


Subject(s)
Neoplasms , Theranostic Nanomedicine , Glutathione/therapeutic use , Humans , Manganese Compounds/therapeutic use , Multimodal Imaging , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Oxides/therapeutic use , Silicon Dioxide/therapeutic use , Theranostic Nanomedicine/methods , Tumor Microenvironment
9.
ACS Appl Mater Interfaces ; 14(24): 27634-27650, 2022 Jun 22.
Article in English | MEDLINE | ID: mdl-35638645

ABSTRACT

Peri-implantitis is a major cause of dental implant failure. Bacterial biofilm contamination on the implant induces surrounding bone resorption and soft tissue inflammation, leading to severe deterioration of oral health. However, conventional biofilm removal procedures, such as mechanical decontamination and antiseptic application, are not effective enough to induce reosseointegration on decontaminated implant surfaces. This is due to (1) incomplete decontamination of the biofilm from inaccessible areas and (2) physicochemical alteration of implant surfaces caused by decontamination procedures. Herein, a safe and effective therapeutic approach for peri-implantitis is developed, which involves decontamination of implant-bound biofilms using the kinetic energy of microsized oxygen bubbles generated from the catalytic reaction between hydrogen peroxide (H2O2) and manganese oxide (MnO2) nanozyme sheet-doped silica diatom microparticles (Diatom Microbubbler, DM). Rapidly moving microsized DM particles are able to penetrate narrow spaces between implant screws, exerting just the right amount of force to entirely destroy biofilms without harming the surrounding mucosa or implant surfaces, as opposed to conventional antiseptics such as chlorhexidine or 3% H2O2 when used alone. Consequently, decontamination with DM facilitates successful reosseointegration on the peri-implantitis-affected implant surface. In summary, our new DM-based therapeutic approach will become a promising alternative to resolve clinically challenging aspects of peri-implantitis.


Subject(s)
Anti-Infective Agents, Local , Dental Implants , Diatoms , Peri-Implantitis , Humans , Hydrogen Peroxide , Manganese Compounds/therapeutic use , Oxides/pharmacology , Oxides/therapeutic use , Peri-Implantitis/drug therapy , Peri-Implantitis/microbiology
10.
Adv Sci (Weinh) ; 9(17): e2200005, 2022 06.
Article in English | MEDLINE | ID: mdl-35484709

ABSTRACT

Sonodynamic therapy (SDT) typically suffers from compromised anticancer efficacy owing to the low reactive oxygen species (ROS) yield and complicated tumor microenvironment (TME) which can consume ROS and support the occurrence and development of tumors. Herein, ultrathin-FeOOH-coated MnO2 nanospheres (denoted as MO@FHO) as sonosensitizers which can not only facilitate ultrasound (US)-triggered ROS but also tune the TME by hypoxia alleviation, H2 O2 consumption as well as glutathione (GSH) depletion are designed. The FeOOH coating will boost the production yield of singlet oxygen (1 O2 ) and hydroxyl radicals (• OH) by inhibiting the recombination of US-initiated electron-hole pairs and Fenton-like reaction, respectively. Additionally, the catalase-like and GSH peroxidase-like activities of MO@FHO nanospheres enable them to break the TME equilibrium via hypoxia alleviation and GSH depletion. The combination of high ROS yield and fundamental destruction of TME equilibrium results in satisfactory antitumor outcomes, as demonstrated by the high tumor suppression efficacy of MO@FHO on MDA-MB-231-tumor-bearing mice. No obvious toxicity is detected to normal tissues at therapeutic doses in vivo. The capability to modulate the ROS production and TME simultaneously can afford new probability for the development of advanced sonosensitizers for synergistic comprehensive cancer therapy.


Subject(s)
Neoplasms , Tumor Microenvironment , Animals , Glutathione/therapeutic use , Hypoxia , Manganese Compounds/pharmacology , Manganese Compounds/therapeutic use , Mice , Neoplasms/therapy , Oxides/pharmacology , Oxides/therapeutic use , Reactive Oxygen Species
11.
J Mater Chem B ; 10(9): 1343-1358, 2022 03 02.
Article in English | MEDLINE | ID: mdl-35129557

ABSTRACT

Bacterial infection has received substantial attention and poses a serious threat to human health. Although antibiotics can effectively fight against bacterial infection, the occurrence of antibiotic resistance has become increasingly serious in recent years, which tremendously hinders its clinical application. Consequently, it is urgent to explore novel strategies to achieve efficacious treatment of bacterial diagnosis and detection. Manganese dioxide (MnO2) nanomaterial has been extensively reported in tumor therapy. Nevertheless, there are few antibacterial reviews of MnO2. Herein, we will discuss the applications of MnO2 in the detection and treatment of bacterial infection, including photodynamic therapy, immunotherapy, improvement of hypoxia, dual-modal combination therapy, reactive oxygen species scavenging, magnetic resonance imaging, optical application of acoustic imaging, and so forth. This review is expected to provide meaningful guidance on further research of MnO2 nanomaterial for antibacterial applications.


Subject(s)
Bacterial Infections , Nanostructures , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/diagnostic imaging , Bacterial Infections/drug therapy , Humans , Manganese Compounds/pharmacology , Manganese Compounds/therapeutic use , Nanostructures/therapeutic use , Oxides/pharmacology , Oxides/therapeutic use
12.
Drug Deliv Transl Res ; 12(11): 2678-2692, 2022 11.
Article in English | MEDLINE | ID: mdl-35061221

ABSTRACT

The limited tissue penetration depth and tumor hypoxic microenvironment have become the two pivotal obstacles that alleviate the antineoplastic efficacy in tumor photodynamic therapy (PDT). In the research, MnO2-decorated upconversion nanoparticles (UCSMn) have been designed to generate certain oxygen within the solid tumor, and also increase the light penetrating depth due to the optical conversion ability derived from upconversion nanoparticles. Furthermore, upconversion nanoparticles as the inner core are coated by mesoporous silica for the loading of curcumin as photosensitizer and chemotherapeutics, and then a MnO2 shell is proceeding to grow via redox method. When reaching the tumor tissue, the MnO2 nanoshells of UCSMn could be rapidly degraded into manganese ions (Mn2+) owing to the reaction with H2O2 in acidic tumor microenvironment, meanwhile producing oxygen and facilitating curcumin release. Once the tumor is illuminated by 980 nm light, the upconversion nanoparticles can transform the infrared light to visible light of 450 nm and 475.5 nm, which can be efficiently absorbed by curcumin, and then produce singlet oxygen to induce tumor cell apoptosis. Curcumin played a dual role which can not only be acted as a photosensitizer, but also a chemotherapeutic agent to further reinforce the antitumor activity. In short, the intelligent nanostructure has the potential to overcome the above-mentioned shortcomings existed in PDT and eventually do work well in the hypoxia tumors. MnO2-decorated upconversion nanoparticle to solve the tissue penetration and tumor hypoxic microenvironment for tumor photodynamic therapy.


Subject(s)
Antineoplastic Agents , Curcumin , Nanoparticles , Neoplasms , Photochemotherapy , Antineoplastic Agents/chemistry , Cell Line, Tumor , Humans , Hydrogen Peroxide , Manganese/therapeutic use , Manganese Compounds/chemistry , Manganese Compounds/therapeutic use , Nanoparticles/chemistry , Neoplasms/drug therapy , Oxides/chemistry , Oxides/therapeutic use , Oxygen/metabolism , Oxygen/therapeutic use , Photochemotherapy/methods , Photosensitizing Agents , Silicon Dioxide/chemistry , Singlet Oxygen/metabolism , Singlet Oxygen/therapeutic use
13.
Photodiagnosis Photodyn Ther ; 37: 102684, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34923155

ABSTRACT

Photodynamic therapy (PDT) and photothermal therapy (PTT) are both promising therapeutic approaches for cancer. Unfortunately, the anticancer efficiency of PDT is restricted by the hypoxic tumor microenvironment and the performance of the photosensitizer (PS) while the efficiency of PTT is limited by the penetration depth of NIR light, making it difficult to further improve the efficiency of the treatment. In this paper, we strategically proposed a multifunctional nano-platform based on g-C3N4 and loaded with CuS and MnO2 nanoparticals. Interestingly, the obtained F127@CNs-CuS/MnO2 nano-platform with high singlet oxygen quantum yield and excellent photothermal performance were used in synergistic PTT and PDT therapy to cope with the limitation of single mode cancer treatment under irradiation and has greatly improved the treatment effect. Additionally, MnO2 nanoparticles loaded on the CNs surface could not only generate oxygen to ameliorate hypoxia in the tumor environment by reacting with H2O2 in tumor cells, but also react with the over-expressed reduced glutathione (GSH) in cancer cells to further improve the synergistic therapeutic effect. In the in vitro hepatocarcinoma cell inactivation experiment, the maximum cell inactivation efficiency of the PDT, PTT and PDT/PTT synergistic treatment group reached at 65% (F127@CNs-MnO2), 69.2% (CNs-MnO2) and 88.6% (F127@CNs-MnO2) respectively, which means that the F127@CNs-CuS/MnO2-mediated PTT/PDT synergy anticancer treatment was more effective than single mode therapy. In summary, the innovative multifunctional nanoplatform F127@CNs-CuS/MnO2 used for synergistic PTT and PDT treatment has greatly improved the inactivation efficiency of cancer cells and has provided a new scheme for the treatment of hypoxic tumors.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Copper , Humans , Hydrogen Peroxide/therapeutic use , Manganese Compounds/pharmacology , Manganese Compounds/therapeutic use , Neoplasms/drug therapy , Oxides/pharmacology , Oxygen , Photochemotherapy/methods , Tumor Microenvironment
14.
ACS Appl Mater Interfaces ; 13(46): 55559-55568, 2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34783244

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory joint disease. Antioxidative treatment combined with chemotherapy holds great promise for RA treatment, and the ability to efficiently deliver drugs and antioxidants to the RA synovial joint is highly desired. Herein, we developed a programmable polymeric microneedle (MN) platform for transdermal delivery of methotrexate (MTX) and reactive oxygen species (ROS) scavengers for RA treatment. The biodegradable MNs made of polyvinylpyrrolidone (PVP) were incorporated with polydopamine/manganese dioxide (termed PDA@MnO2) and MTX. After insertion into skin tissue, the MNs degraded, thus enabling release of loaded MTX and PDA@MnO2. The PDA@MnO2 could be utilized as an MRI contrast agent in the RA synovial microenvironment. It also acted as a robust antioxidant to remove ROS and decrease RA inflammation, which when combined with the MTX-mediated chemotherapy led to an ideal outcome for RA treatments in a murine model. This work not only represents a valuable MN-assisted RA therapeutic agent transdermal delivery approach but also opens a new avenue for chemotherapy and antioxidative synergistic treatment of RA.


Subject(s)
Antioxidants/therapeutic use , Arthritis, Rheumatoid/drug therapy , Polymers/therapeutic use , 3T3 Cells , Administration, Cutaneous , Animals , Antioxidants/administration & dosage , Arthritis, Rheumatoid/metabolism , Drug Therapy, Combination , Indoles/administration & dosage , Indoles/therapeutic use , Manganese Compounds/administration & dosage , Manganese Compounds/therapeutic use , Methotrexate/administration & dosage , Methotrexate/therapeutic use , Mice , Oxides/administration & dosage , Oxides/therapeutic use , Particle Size , Polymers/administration & dosage , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Surface Properties
15.
ACS Appl Mater Interfaces ; 13(24): 28650-28661, 2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34124895

ABSTRACT

Novel and effective radiosensitizers that can enhance radiosensitivity of tumor tissues and increase the local radiation dose are highly desirable. In this work, templated by bovine serum albumin (BSA), Bi2Se3-MnO2 nanocomposites (Bi2Se3-MnO2@BSA) were fabricated via biomineralization, while Bi2Se3 nanodots act as radiosensitizers to increase the local radiation dosage because of their strong X-ray attenuation ability, and MnO2 with catalase-like activity can increase the oxygen concentration in tumors by triggering the decomposition of tumor endogenous H2O2 so as to improve the hypoxia-associated radioresistance of tumors. Owing to the interaction of the two components in the interface, Bi2Se3-MnO2@BSA showed promoted catalytic activity compared to MnO2@BSA, favoring tumor radiotherapy (RT) sensitization. BSA templating enabled the nanocomposites with high colloidal stability and biocompatibility as well as satisfactory tumor targeting both in vitro and in vivo; thus, an enhanced RT efficacy was obtained. Moreover, the proposed Bi2Se3-MnO2@BSA exhibited excellent performances in computerized tomography and magnetic resonance imaging. Thus, this work provides a tumor microenvironment-responsive multifunctional theranostic nanoagent with an improved performance for imaging-guided tumor RT sensitization.


Subject(s)
Antineoplastic Agents/therapeutic use , Bismuth/therapeutic use , Manganese Compounds/therapeutic use , Nanocomposites/therapeutic use , Neoplasms/drug therapy , Oxides/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Selenium Compounds/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/radiation effects , Bismuth/chemistry , Catalysis/radiation effects , Cattle , Cell Line, Tumor , Contrast Media/chemical synthesis , Contrast Media/radiation effects , Contrast Media/therapeutic use , Female , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Manganese Compounds/chemistry , Manganese Compounds/radiation effects , Mice, Inbred BALB C , Nanocomposites/chemistry , Nanocomposites/radiation effects , Neoplasms/diagnostic imaging , Oxides/chemistry , Oxides/radiation effects , Oxygen/metabolism , Precision Medicine , Radiation-Sensitizing Agents/chemical synthesis , Radiation-Sensitizing Agents/radiation effects , Selenium Compounds/chemistry , Selenium Compounds/radiation effects
16.
Article in English | MEDLINE | ID: mdl-33908171

ABSTRACT

Tumor microenvironment (TME) is generally featured by low pH values, high glutathione (GSH) concentrations, overproduced hydrogen peroxide (H2 O2 ), and severe hypoxia. These characteristics could provide an interior environment for origination and residence of tumor cells and would lead to tumor progression, metastasis, and drug resistance. Therefore, the development of TME-responsive smart nanosystems has shown significant potential to enhance the efficacy of current cancer treatments. Manganese dioxide (MnO2 )-based nanosystems have attracted growing attentions for applications in cancer treatment as an excellent TME-responsive theranostic platform, due to their tunable structures/morphologies, pH responsive degradation, and excellent catalytic activities. In this review, we mainly summarize the strategies of MnO2 and its nanocomposites to modulate TME, such as tumor hypoxia relief, excessive GSH depletion, glucose consumption, and tumor immunosuppressive microenvironment moderation. Such MnO2 -based TME modulation would be beneficial for a wide range of cancer therapies including photodynamic therapy, radiotherapy, sonodynamic therapy, chemodynamic therapy, starvation therapy, and immunotherapy. Next, some representative designs of MnO2 -based nanoplatforms in other tumor therapies are highlighted. Moreover, we will discuss the challenges and future perspectives of these MnO2 -based nanosystems for enhanced tumor treatment. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.


Subject(s)
Multifunctional Nanoparticles , Nanoparticles , Neoplasms , Photochemotherapy , Cell Line, Tumor , Humans , Manganese Compounds/therapeutic use , Neoplasms/drug therapy , Oxides/therapeutic use , Theranostic Nanomedicine , Tumor Microenvironment
17.
ACS Appl Mater Interfaces ; 13(13): 14928-14937, 2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33759491

ABSTRACT

In order to achieve safe and high-efficient photodynamic therapy (PDT), it was a powerful strategy of constructing O2-generated nanozyme with intelligent "off/on" modulation and enhancement. Herein, a kind of H2O2-responsive nanozyme was developed for off/on modulation and enhancement of magnetic resonance (MR) imaging and PDT, in which great amounts of gold nanoclusters (AuNCs) were loaded into mesoporous silica to form nanoassembly, and manganese dioxide (MnO2) nanosheets were wrapped as switching shield shell (AuNCs@mSiO2@MnO2). In a neutral physiological environment, stable MnO2 shells eliminated singlet oxygen (1O2) generation to switch off PDT and MR imaging. However, in an acidic tumor microenvironment, the MnO2 shell reacted with H2O2, in which MnO2 degradation switched on MR imaging and PDT, and the generated O2 further enhanced PDT. H2O2-responsive MnO2 degradation brought about excellent MR imaging with a longitudinal relaxation rate of 25.31 mM-1 s-1, and simultaneously sufficient O2 generation guaranteed a 74% high 1O2 yield. Under the irradiation of a 635 nm laser, the viability of MDA-MB-435 cells was reduced to 4%, and the tumors completely disappeared, demonstrating strong PDT performance. Therefore, H2O2-responsive AuNCs@mSiO2@MnO2 nanozyme showed excellent off/on modulation and enhancement of MR imaging and PDT and was a promising intelligent nanoprobe for safe and high-efficiency theranostics.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Gold/therapeutic use , Manganese Compounds/therapeutic use , Nanoparticles/therapeutic use , Oxides/therapeutic use , Photosensitizing Agents/therapeutic use , Animals , Cell Line, Tumor , Female , Gold/chemistry , Humans , Hydrogen Peroxide/metabolism , Magnetic Resonance Imaging , Manganese Compounds/chemistry , Mice , Mice, Nude , Nanoparticles/chemistry , Oxides/chemistry , Photochemotherapy , Photosensitizing Agents/chemistry
18.
Biomater Sci ; 9(7): 2709-2720, 2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33616151

ABSTRACT

Carbon monoxide (CO) based gas therapy has been an emerging strategy for cancer treatment. However, the uncontrolled release of CO and limited therapeutic efficacy of monotherapy are two major obstacles for clinical application. To overcome these issues, human serum albumin (HSA) nanoparticles combined with manganese dioxide (MnO2) were developed to deliver a photosensitizer (IR780) and CO donor (MnCO) for a synergistic therapy combining CO gas therapy and phototherapy. The nanoparticles (HIM-MnO2) formed catalyze hydrogen peroxide to produce oxygen for hypoxia relief. With laser irradiation, it can increase the generation of reactive oxygen species for the enhancement of photodynamic therapy (PDT). Furthermore, the generated heat of photothermal therapy (PTT) induced by nanoparticles could trigger the release of CO to achieve a therapeutic window for enhanced gas therapy. Due to the co-localization of IR780 in mitochondria, HIM-MnO2 could accumulate in mitochondria for the synergistic therapy combining CO gas therapy and phototherapy, and could oxidize the mitochondrial membrane and induce more apoptosis. After intravenous injection into tumor bearing mice, HIM-MnO2 could accumulate at tumor sites and with laser irradiation, tumor growth was significantly inhibited due to the enhanced PDT, PTT, and CO gas therapy. This study provides a strategy with oxygen generating and thermal-responsive CO release to combine phototherapy and CO gas therapy for cancer treatment.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Animals , Carbon Monoxide , Cell Line, Tumor , Manganese Compounds/therapeutic use , Mice , Mitochondria , Neoplasms/drug therapy , Oxides/therapeutic use , Oxygen
19.
Drug Metab Rev ; 53(4): 592-603, 2021 11.
Article in English | MEDLINE | ID: mdl-33561356

ABSTRACT

In the modern age, the struggle to generate appropriate bio-based materials and nano-scaled colloidal particulates for developed application domains, has already resulted in remarkable attempts in the advancement of regulated size and shape, anisotropy, and characteristics of nanostructures. The bottom-up development strategies of components are among the most important science areas throughout nanotechnology, in which the designed building blocks are often utilized to generate novel structures by random self-assembly. In biomedical applications, Janus nanoparticles (JNPs) are necessary. This is due to their effective stimulus-responsive properties, tunable structure, biocompatibility, containing two surfaces with various hydrophobic characteristics and distinct functional groups. Featuring two parts with differing hydrophobicity has been the most critical aspect of the Janus amphiphilic particles. Development of JNPs has been afforded, using imaging agents (e.g. gold (AU) for photoacoustic imaging processing (PAI), silver for surface-enhanced Raman scattering (SERS), and Fe3O4 and MnO2 to magnetic resonance imaging (MRI)). It is also to be mentioned that a number of other properties become salient - properties such as integration imaging factors into JNPs (like quantum dots, fluorescent dyes), multiple imaging methods for screening and diagnosis application can indeed be accomplished. Janus nanostructures have been promising platforms for bioengineering as therapeutic carriers, drug delivery vehicles, and biosensor equipment; they may also be employed for the transport of bioactive hydrophilic and hydrophobic materials. The main production approaches and major advancement of JNPs in the biomedical sector and cancer therapy will be described in this paper.


Subject(s)
Multifunctional Nanoparticles , Nanoparticles , Neoplasms , Gold/chemistry , Humans , Manganese Compounds/therapeutic use , Neoplasms/drug therapy , Oxides/therapeutic use
20.
Theranostics ; 10(20): 9132-9152, 2020.
Article in English | MEDLINE | ID: mdl-32802183

ABSTRACT

Photodynamic therapy (PDT) is a promising strategy in cancer treatment that utilizes photosensitizers (PSs) to produce reactive oxygen species (ROS) and eliminate cancer cells under specific wavelength light irradiation. However, special tumor environments, such as those with overexpression of glutathione (GSH), which will consume PDT-mediated ROS, as well as hypoxia in the tumor microenvironment (TME) could lead to ineffective treatment. Moreover, PDT is highly light-dependent and therefore can be hindered in deep tumor cells where light cannot easily penetrate. To solve these problems, we designed oxygen-dual-generating nanosystems MnO2@Chitosan-CyI (MCC) for enhanced phototherapy. Methods: The TME-sensitive nanosystems MCC were easily prepared through the self-assembly of iodinated indocyanine green (ICG) derivative CyI and chitosan, after which the MnO2 nanoparticles were formed as a shell by electrostatic interaction and Mn-N coordinate bonding. Results: When subjected to NIR irradiation, MCC offered enhanced ROS production and heat generation. Furthermore, once endocytosed, MnO2 could not only decrease the level of GSH but also serve as a highly efficient in situ oxygen generator. Meanwhile, heat generation-induced temperature increase accelerated in vivo blood flow, which effectively relieved the environmental tumor hypoxia. Furthermore, enhanced PDT triggered an acute immune response, leading to NIR-guided, synergistic PDT/photothermal/immunotherapy capable of eliminating tumors and reducing tumor metastasis. Conclusion: The proposed novel nanosystems represent an important advance in altering TME for improved clinical PDT efficacy, as well as their potential as effective theranostic agents in cancer treatment.


Subject(s)
Immunologic Factors/metabolism , Nanoparticles/therapeutic use , Tumor Hypoxia/drug effects , Tumor Hypoxia/physiology , Tumor Microenvironment/physiology , Animals , Cell Line , Cell Line, Tumor , Female , Glutathione/metabolism , Humans , Indocyanine Green/metabolism , Male , Manganese Compounds/therapeutic use , Mice , Mice, Inbred BALB C , Oxides/therapeutic use , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Phototherapy/methods , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Theranostic Nanomedicine/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...