Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 99
1.
J Dent Res ; 103(6): 612-621, 2024 Jun.
Article En | MEDLINE | ID: mdl-38684484

Oral squamous cell carcinoma (OSCC) is the most common type of oral cancer, characterized by invasiveness, local lymph node metastasis, and poor prognosis. Traditional treatment and medications have limitations, making the specific inhibition of OSCC growth, invasion, and metastasis a challenge. The tumor microenvironment exhibits mildly acidity and high concentrations of H2O2, and its exploitation for cancer treatment has been widely researched across various cancers, but research in the oral cancer field is relatively limited. In this study, by loading ultra-small Prussian blue nanoparticles (USPBNPs) into mesoporous calcium-silicate nanoparticles (MCSNs), we developed an acid-responsive iron-based nanocomposite, USPBNPs@MCSNs (UPM), for the OSCC treatment. UPM demonstrated excellent dual enzyme activities, generating toxic ·OH in a mildly acidic environment, effectively killing OSCC cells and producing O2 in a neutral environment to alleviate tissue hypoxia. The results showed that UPM could effectively inhibit the proliferation, migration, and invasion of OSCC cells, as well as the growth of mice solid tumors, without obvious systemic toxicity. The mechanisms may involve UPM inducing ferroptosis of OSCC cells by downregulating the xCT/GPX4/glutathione (GSH) axis, characterized by intracellular iron accumulation, reactive oxygen species accumulation, GSH depletion, lipid peroxidation, and abnormal changes in mitochondrial morphology. Therefore, this study provides empirical support for ferroptosis as an emerging therapeutic target for OSCC and offers a valuable insight for future OSCC treatment.


Cell Proliferation , Iron , Mouth Neoplasms , Nanocomposites , Tumor Microenvironment , Nanocomposites/chemistry , Animals , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Mice , Humans , Cell Proliferation/drug effects , Tumor Microenvironment/drug effects , Cell Line, Tumor , Ferrocyanides/therapeutic use , Silicates/therapeutic use , Silicates/pharmacology , Hydrogen-Ion Concentration , Cell Movement/drug effects , Nanoparticles , Reactive Oxygen Species/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Mice, Nude , Ferroptosis/drug effects , Hydrogen Peroxide , Xenograft Model Antitumor Assays , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/pathology , Mice, Inbred BALB C
2.
Biometals ; 36(5): 1125-1140, 2023 10.
Article En | MEDLINE | ID: mdl-37222858

The similarities between thallium and potassium have suggested the use of calcium polystyrene sulfonate (CPS), an oral ion exchange resin, as a potential agent against thallium intoxication. Therefore, the study was an attempt to evaluate the efficacy of CPS and Prussian blue when given alone or in combination against thallium toxicity. The effect on binding capacity was investigated in terms of contact time, amount of CPS, influence of pH, simulated physiological solutions and interference of potassium ions. Also, rats were given single dose of thallium chloride (20 mg kg-1) and the treatment with PB and CPS was given for 28 days as CPS 30 g kg-1, orally, twice a day, PB 3 g kg-1, orally, twice a day and their combination. The effect of antidotal treatment was evaluated by calculating the thallium levels in various organs, blood, urine and feces. The results of the in vitro study indicated exceedingly quick binding in the combination of CPS and PB as compared to PB alone. Also, it was found that the binding capacity at pH 2.0 was considerably increased for PB with CPS (184.656 mg g-1) as compared to PB (37.771 mg g-1). Furthermore, statistically significant results were obtained in the in vivo study as after 7th day, thallium levels in blood of rats treated with combination were reduced by 64% as compared to control group and 52% as compared to alone PB treated group. Also, Tl retention in liver, kidney, stomach, colon and small intestine of combination treated rats was significantly reduced to 46%, 28%, 41%, 32% and 33% respectively, as compared to alone PB treated group. These findings demonstrate this as a good antidotal option against thallium intoxication.


Antidotes , Thallium , Rats , Animals , Thallium/metabolism , Antidotes/pharmacology , Antidotes/therapeutic use , Ferrocyanides/pharmacology , Ferrocyanides/therapeutic use
3.
Part Fibre Toxicol ; 20(1): 18, 2023 05 05.
Article En | MEDLINE | ID: mdl-37147710

BACKGROUND: Prussian blue (PB) nanoparticles (NPs) have been intensively investigated for medical applications, but an in-depth toxicological investigation of PB NPs has not been implemented. In the present study, a comprehensive investigation of the fate and risks of PB NPs after intravenous administration was carried out by using a mouse model and an integrated methodology of pharmacokinetics, toxicology, proteomics, and metabolomics. RESULTS: General toxicological studies demonstrated that intravenous administration of PB NPs at 5 or 10 mg/kg could not induce obvious toxicity in mice, while mice treated with a relatively high dose of PB NPs at 20 mg/kg exhibited loss of appetite and weight decrease in the first two days postinjection. Pharmacokinetic studies revealed that intravenously administered PB NPs (20 mg/kg) underwent fast clearance from blood, highly accumulated in the liver and lungs of mice, and finally cleared from tissues. By further integrated proteomics and metabolomics analysis, we found that protein expression and metabolite levels changed significantly in the liver and lungs of mice due to the high accumulation of PB NPs, leading to slight inflammatory responses and intracellular oxidative stress. CONCLUSIONS: Collectively, our integrated experimental data imply that the high accumulation of PB NPs may cause potential risks to the liver and lungs of mice, which will provide detailed references and guidance for further clinical application of PB NPs in the future.


Ferrocyanides , Nanoparticles , Ferrocyanides/administration & dosage , Ferrocyanides/therapeutic use , Ferrocyanides/toxicity , Nanoparticles/administration & dosage , Nanoparticles/therapeutic use , Nanoparticles/toxicity , Oxidative Stress , Proteomics
5.
J Control Release ; 351: 752-778, 2022 11.
Article En | MEDLINE | ID: mdl-36216173

Prussian blue (PB) nanoparticles possess excellent physicochemical properties, including imaging features, robust photothermal conversion ability, catalytic activity, surface modifiability, effective drug loading, good stability, biocompatibility and biodegradability. With the advancement of nanotechnology, diverse PB-based nanoplatforms have been developed for biomedical applications. This review systematically summarized recent studies on PB-based nanoplatforms in the treatment of tumor (photothermal therapy, photodynamic therapy, chemotherapy, immunotherapy, theranostics, etc.), cardiovascular and cerebrovascular diseases (restenosis, atherosclerosis, thrombosis, ischemic stroke, etc.), bacterial infections (photothermal sterilization, biofilms disruption, etc.), and other inflammation-related diseases (liver injury, acute pancreatitis, inflammatory bowel disease, osteoarthritis, etc.). Finally, the existing challenges associated with current studies are discussed, and the future possible research and application directions for PB-based nanoplatforms are proposed, providing paradigms for subsequent development.


Nanoparticles , Pancreatitis , Humans , Acute Disease , Ferrocyanides/therapeutic use , Ferrocyanides/chemistry , Nanoparticles/chemistry
6.
J Nanobiotechnology ; 19(1): 449, 2021 Dec 24.
Article En | MEDLINE | ID: mdl-34952587

BACKGROUND: Mono-therapeutic modality has limitations in combating metastatic lesions with complications. Although emerging immunotherapy exhibits preliminary success, solid tumors are usually immunosuppressive, leading to ineffective antitumor immune responses and immunotherapeutic resistance. The rational combination of several therapeutic modalities may potentially become a new therapeutic strategy to effectively combat cancer. RESULTS: Poly lactic-co-glycolic acid (PLGA, 50 mg) nanospheres were constructed with photothermal transduction agents (PTAs)-Prussian blue (PB, 2.98 mg) encapsulated in the core and chemotherapeutic docetaxel (DTX, 4.18 mg)/ immune adjuvant-imiquimod (R837, 1.57 mg) loaded in the shell. Tumor cell membranes were further coated outside PLGA nanospheres (designated "M@P-PDR"), which acted as "Nano-targeted cells" to actively accumulate in tumor sites, and were guided/monitored by photoacoustic (PA)/ magnetic resonance (MR) imaging. Upon laser irradiation, photothermal effects were triggered. Combined with DTX, PTT induced in situ tumor eradication. Assisted by the immune adjuvant R837, the maturation rate of DCs increased by 4.34-fold compared with that of the control. In addition, DTX polarized M2-phenotype tumor-associated macrophages (TAMs) to M1-phenotype, relieving the immunosuppressive TME. The proportion of M2-TAMs decreased from 68.57% to 32.80%, and the proportion of M1-TAMs increased from 37.02% to 70.81%. Integrating the above processes, the infiltration of cytotoxic T lymphocytes (CTLs) increased from 17.33% (control) to 35.5%. Primary tumors and metastasis were significantly inhibited when treated with "Nano-targeted cells"-based cocktail therapy. CONCLUSION: "Nano-targeted cells"-based therapeutic cocktail therapy is a promising approach to promote tumor regression and counter metastasis/recurrence.


Antineoplastic Agents/therapeutic use , Cell Membrane/chemistry , Docetaxel/chemistry , Nanoparticles/chemistry , Neoplasms/therapy , Adjuvants, Immunologic/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Docetaxel/pharmacology , Docetaxel/therapeutic use , Ferrocyanides/chemistry , Ferrocyanides/pharmacology , Ferrocyanides/therapeutic use , Humans , Imiquimod/chemistry , Imiquimod/immunology , Immunotherapy/methods , Infrared Rays , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Nude , Neoplasms/diagnostic imaging , Optical Imaging , Photothermal Therapy/methods , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry
7.
J Mater Chem B ; 9(47): 9670-9683, 2021 12 08.
Article En | MEDLINE | ID: mdl-34726228

We investigated a series of Mn2+-Prussian blue (PB) nanoparticles NazMnxFe1-x[Fe(CN)6]1-y□y·nH2O of similar size, surface state and cubic morphology with various amounts of Mn2+ synthesized through a one step self-assembly reaction. We demonstrated by a combined experimental-theoretical approach that during the synthesis, Mn2+ substituted Fe3+ up to a Mn/Na-Mn-Fe ratio of 32 at% in the PB structure, while for higher amounts, the Mn2[Fe(CN)6] analogue is obtained. For comparison, the post-synthetic insertion of Mn2+ in PB nanoparticles was also investigated and completed with Monte-Carlo simulations to probe the plausible adsorption sites. The photothermal conversion efficiency (η) of selected samples was determined and showed a clear dependence on the Mn2+amount with a maximum efficiency for a Mn/Na-Mn-Fe ratio of 10 at% associated with a dependence on the nanoparticle concentration. Evaluation of the in vitro photothermal properties of these nanoparticles performed on triple negative human breast adenocarcinoma (MDA-MB-231) cells by using continuous and pulsed laser irradiation confirm their excellent PTT efficiency permitting low dose use.


Antineoplastic Agents/therapeutic use , Ferrocyanides/therapeutic use , Manganese/chemistry , Nanoparticles/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/radiation effects , Cell Line, Tumor , Ferrocyanides/chemistry , Ferrocyanides/radiation effects , Humans , Iron/chemistry , Iron/radiation effects , Manganese/radiation effects , Nanoparticles/chemistry , Nanoparticles/radiation effects , Photochemical Processes , Photothermal Therapy , Xenograft Model Antitumor Assays , Zebrafish
8.
ACS Appl Mater Interfaces ; 13(36): 42382-42395, 2021 Sep 15.
Article En | MEDLINE | ID: mdl-34473471

Anthracycline-induced liver injury (AILI) is becoming an increasingly serious and potential clinical complication and is linked to reactive oxygen species (ROS) production and subsequent inflammatory response. Herein, we demonstrated that artificial Prussian blue nanozymes (PBZs) prevented daunorubicin-induced liver injury, a prototype of AILI, by attenuating ROS production and regulating inflammation. PBZs exhibited multienzyme activity and could scavenge ROS and free radicals. At the cellular level, PBZs could effectively eliminate ROS, suppress hepatocyte apoptosis, reduce deoxyribonucleic acid damage, and decrease the levels of inflammatory cytokines and chemokines. According to the results of the in vivo study, pretreatment with PBZs also resulted in a desirable protective effect against AILI, as indicated by both a decrease in biochemical indicator levels and hepatocyte necrosis. PBZs upregulated antioxidative genes by activating the Nrf2 pathway to reduce oxidative stress. Meanwhile, PBZs counteracted the inflammatory response based on the decreased expression levels of myeloperoxidase and F4/80 in the liver. Collectively, our findings indicate that PBZ-based nanotherapy is a novel strategy for protecting against AILI.


Anti-Inflammatory Agents/therapeutic use , Chemical and Drug Induced Liver Injury/prevention & control , Daunorubicin/toxicity , Free Radical Scavengers/therapeutic use , Inflammation/drug therapy , Nanoparticles/therapeutic use , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/toxicity , Apoptosis/drug effects , Catalysis , Chemical and Drug Induced Liver Injury/metabolism , Chemokines/metabolism , DNA/drug effects , DNA Damage/drug effects , Ferrocyanides/chemistry , Ferrocyanides/therapeutic use , Ferrocyanides/toxicity , Free Radical Scavengers/chemistry , Free Radical Scavengers/toxicity , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/toxicity , Oxidative Stress/drug effects , Povidone/chemistry , Povidone/toxicity , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
9.
ACS Appl Mater Interfaces ; 13(31): 37746-37760, 2021 Aug 11.
Article En | MEDLINE | ID: mdl-34318658

Mitochondrial dysfunction, which is directly involved in Parkinson's disease (PD), is characterized by the production of reactive oxygen species (ROS) and aberrant energy metabolism. Thus, regulating mitochondrial function might be an effective strategy to treat PD. However, the blood-brain barrier (BBB) presents a significant challenge for the intracerebral delivery of drugs. Here, we synthesized a zeolitic imidazolate framework 8-coated Prussian blue nanocomposite (ZIF-8@PB), which was encapsulated with quercetin (QCT), a natural antioxidant, to treat PD. ZIF-8@PB-QCT exhibited superior near-infrared radiation (NIR) response and penetrated through the BBB to the site of mitochondrial damage guided by the photothermal effect. In the mice model of PD, the QCT released from ZIF-8@PB-QCT significantly increased the adenosine triphosphate levels, reduced the oxidative stress levels, and reversed dopaminergic neuronal damage as well as PD-related behavioral deficits without any damage to the normal tissues. Furthermore, we explored the underlying neuroprotective mechanism of ZIF-8@PB-QCT that was mediated by activating the PI3K/Akt signaling pathway. Thus, combined with noninvasive NIR radiation, the biocompatible ZIF-8@PB-QCT nanocomposite could be used to treat neurodegenerative diseases.


Antioxidants/therapeutic use , Nanocomposites/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinson Disease, Secondary/drug therapy , Quercetin/therapeutic use , Animals , Antioxidants/chemistry , Antioxidants/pharmacokinetics , Antioxidants/toxicity , Blood-Brain Barrier/physiology , Cell Line, Tumor , Drug Liberation , Ferrocyanides/chemistry , Ferrocyanides/radiation effects , Ferrocyanides/therapeutic use , Ferrocyanides/toxicity , Humans , Imidazoles/chemistry , Imidazoles/therapeutic use , Imidazoles/toxicity , Infrared Rays , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Nanocomposites/chemistry , Nanocomposites/radiation effects , Nanocomposites/toxicity , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/toxicity , Oxidative Stress/drug effects , Quercetin/chemistry , Quercetin/pharmacokinetics , Quercetin/toxicity , Rats, Sprague-Dawley , Zeolites/chemistry , Zeolites/therapeutic use , Zeolites/toxicity
10.
Theranostics ; 11(7): 3213-3228, 2021.
Article En | MEDLINE | ID: mdl-33537083

Rationale: Acute pancreatitis (AP) is a serious acute condition affecting the abdomen and shows high morbidity and mortality rates. Its global incidence has increased in recent years. Inflammation and oxidative stress are potential therapeutic targets for AP. This study was conducted to investigate the intrinsic anti-oxidative and anti-inflammatory effects of Prussian blue nanozyme (PBzyme) on AP, along with its underlying mechanism. Methods: Prussian blue nanozymes were prepared by polyvinylpyrrolidone modification method. The effect of PBzyme on inhibiting inflammation and scavenging reactive oxygen species was verified at the cellular level. The efficacy and mechanism of PBzyme for prophylactically treating AP were evaluated using the following methods: serum testing in vivo, histological scoring following hematoxylin and eosin staining, terminal deoxynucleotidyl transferase dUTP nick end labeling fluorescence staining, polymerase chain reaction array, Kyoto Encyclopedia of Genes and Genomes analysis and Western blotting analysis. Results: The synthetic PBzyme showed potent anti-oxidative and anti-inflammatory effects in reducing oxidative stress and alleviating inflammation both in vitro and in vivo in the prophylactic treatment of AP. The prophylactic therapeutic efficacy of PBzyme on AP may involve inhibition of the toll-like receptor/nuclear factor-κB signaling pathway and reactive oxygen species scavenging. Conclusion: The single-component, gram-level mass production, stable intrinsic biological activity, biosafety, and good therapeutic efficacy suggest the potential of PBzyme in the preventive treatment of AP. This study provides a foundation for the clinical application of PBzyme.


Enzyme Therapy/methods , Nanotechnology/methods , Pancreatitis/therapy , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , China , Cytokines/metabolism , Enzymes/metabolism , Enzymes/pharmacology , Ferricyanides/chemistry , Ferricyanides/therapeutic use , Ferrocyanides/chemistry , Ferrocyanides/therapeutic use , Humans , Inflammation/drug therapy , Inflammation/pathology , Male , Mice, Inbred BALB C , NF-kappa B/drug effects , Oxidative Stress/drug effects , Pancreatitis/metabolism , Povidone/chemistry , Povidone/therapeutic use , Prussian Blue Reaction/methods , Reactive Oxygen Species/metabolism , Toll-Like Receptors/drug effects
11.
Medicine (Baltimore) ; 98(29): e16471, 2019 Jul.
Article En | MEDLINE | ID: mdl-31335706

Thallium is highly toxic and its effects are cumulative. The clinical symptoms of thallium poisoning are non-specific, thereby delaying admission and treatment. This study aimed to summarize the clinical features and treatment experience of patients with delayed admission who experience thallium poisoning.We conducted a retrospective descriptive analysis of patients in our hospital from 2008 to 2018 who had thallium poisoning and experienced a delay in hospital admission. The time from symptom onset to admission was assessed. The patients were divided into 3 groups and descriptive analyses of their clinical characteristics, including basic patient information, symptoms, laboratory test results, examination findings, treatment methods, outcomes, and follow-up information, were conducted.A total of 34 patients with thallium poisoning were included: 8 were admitted to the hospital early or with mild delay, 9 had a moderate delay, and 17 had a severely delayed admission. The time from illness onset to admission was 13 (interquartile range, 7.5-26) days. Some patients with delayed admission had significant symptoms associated with central nervous system damage, and changes in magnetic resonance images and electroencephalograms were also noted. After admission, all patients received Prussian blue treatment, and some patients with relatively high blood concentration received blood purification treatments. Following treatment, the blood and urine thallium concentrations of all patients decreased significantly, and their symptoms were alleviated.Our results show that delayed patient admission in cases of thallium poisoning is associated with greater risk of central nervous system damage. Use of Prussian blue combined with blood purification treatments might improve patients' conditions.


Delayed Diagnosis , Heavy Metal Poisoning/diagnosis , Heavy Metal Poisoning/therapy , Hospitalization , Thallium/poisoning , Time-to-Treatment , Adolescent , Adult , Antidotes/therapeutic use , Female , Ferrocyanides/therapeutic use , Heavy Metal Poisoning/blood , Heavy Metal Poisoning/urine , Hemoperfusion , Humans , Male , Middle Aged , Retrospective Studies , Thallium/blood , Thallium/urine
12.
Medicine (Baltimore) ; 98(8): e14629, 2019 Feb.
Article En | MEDLINE | ID: mdl-30813198

RATIONALE: This is the first reported severe thallium poisoning patient successfully treated with Prussian blue (PB) and plasma exchange (PE). PATIENT CONCERNS: A 42-year-old woman in a coma owing to severe thallium poisoning was admitted to our department after day 44 of poisoning. At admission, blood and urine thallium concentrations were 380.0 and 2580.0 ng/mL, respectively. DIAGNOSIS: The patient was diagnosed with toxic encephalopathy induced by thallium poisoning; in addition, she was also diagnosed with bilateral pneumonia, respiratory failure, moderate anemia, hypoproteinemia, and electrolyte imbalance based on her chest X-ray, blood gas analysis, Hb level, albumin levels, and serum electrolyte results. INTERVENTIONS: The patient was intubated and treated with PB (6600 mg/d, 15 days in total) combined with PE (once daily, 5 days in total) as well as other symptomatic supportive care measures. OUTCOMES: After treatments, her blood and urinary thallium concentrations gradually decreased and on the 13th day after admission, the blood thallium concentration decreased to 0 ng/mL. The oxygenation index gradually improved, meantime, the patient gradually regained consciousness, and on the 50th day of admission, the patient's consciousness reverted to a clear-headed state. The patient recovered mostly after 37 months of follow-up. LESSONS: Through this case, we learned that the gradual reduction in blood and urine thallium concentration and the patient's improved condition is correlated with PB and PE treatment. For patients with severe thallium poisoning, this treatment method might be effective; but the exact curative effect is unconfirmed, requiring further research to verify.


Coma/therapy , Ferrocyanides/therapeutic use , Neurotoxicity Syndromes/diagnosis , Plasma Exchange/methods , Thallium/poisoning , Adult , Coma/chemically induced , Female , Humans , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/therapy , Thallium/blood , Thallium/urine
13.
J Biomater Appl ; 33(9): 1202-1213, 2019 04.
Article En | MEDLINE | ID: mdl-30714472

To realize the photothermal therapy ability of Prussian blue-modified ferritin nanoparticles (PB-Ft NPs) and its synergistic effect with chemotherapy, PB-Ft NPs were synthesized by a simple surface double decomposition reaction. Mean sizes of ferritin and PB-Ft NPs were 10.4 nm and 12.6 nm, respectively. The obtained PB-Ft NPs were verified to have both the photothermal conversion ability of Prussian blue and the morphology of ferritin. The in vitro and in vivo photothermal therapy results confirm PB-Ft NPs can successfully inhibit the growth of murine breast cancer cell line (4T1) without any obvious side effect. Moreover, taking use of the peroxidase-like activity of PB-Ft NPs, the photothermal therapy effect of PB-Ft NPs effectively improved the curative effect of gemcitabine (GEM) via enhancing reactive oxygen species production. The obtained PB-Ft NPs can be served as a useful and safe photothermal therapy agent in breast cancer. Moreover, PB-Ft NPs-assisted photothermal therapy can be applied as an adjunctive therapy with various established cancer treatments such as chemotherapy.


Breast Neoplasms/therapy , Ferritins/therapeutic use , Ferrocyanides/therapeutic use , Nanoparticles/therapeutic use , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Combined Modality Therapy , Female , Hyperthermia, Induced , Mice , Mice, Inbred BALB C , Nanomedicine , Phototherapy , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
14.
J Mater Chem B ; 7(12): 2032-2042, 2019 03 28.
Article En | MEDLINE | ID: mdl-32254807

Development of near infrared (NIR) light-responsive nanomaterials for high performance multimodal phototherapy within a single nanoplatform is still challenging in technology and biomedicine. Herein, a new phototherapeutic nanoagent based on FDA-approved Prussian blue (PB) functionalized oxygen-deficient molybdenum oxide nanoparticles (MoO3-x NPs) is strategically designed and synthesized by a facile one-pot size/morphology-controlled process. The as-prepared PB-MoO3-x nanocomposites (NCs) with a uniform particle size of ∼90 nm and high water dispersibility exhibited strong optical absorption in the first biological window, which is induced by plasmon resonance in an oxygen-deficient MoO3-x semiconductor. More importantly, PB-MoO3-x NCs not only exhibited a high photothermal conversion efficiency of ∼63.7% and photostability but also offered a further approach for the generation of reactive oxygen species (ROS) upon singular NIR light irradiation which significantly improved the therapeutic efficiency of the PB agent. Furthermore, PB-MoO3-x NCs showed a negligible cytotoxic effect in the dark, but an excellent therapeutic effect toward two triple-negative breast cancer (TNBC) cell lines at a low concentration (20 µg mL-1) of NCs and a moderate NIR laser power density. Additionally, efficient tumor ablation and metastasis inhibition in a 4T1 TNBC mouse tumor model can also be realized by synergistic photothermal/photodynamic therapy (PTT/PDT) under a single continuous NIR wave laser. Taken together, this study paved the way for the use of a single nanosystem for multifunctional therapy.


Coloring Agents/therapeutic use , Ferrocyanides/therapeutic use , Molybdenum/therapeutic use , Nanocomposites/therapeutic use , Nanoparticles/therapeutic use , Oxides/therapeutic use , Phototherapy , Triple Negative Breast Neoplasms/therapy , Animals , Cell Line, Tumor , Coloring Agents/chemistry , Female , Ferrocyanides/chemistry , Humans , Lasers , Mice, Inbred BALB C , Molybdenum/chemistry , Nanocomposites/chemistry , Nanoparticles/chemistry , Oxides/chemistry , Reactive Oxygen Species/chemistry , Triple Negative Breast Neoplasms/pathology
15.
Mil Med Res ; 5(1): 27, 2018 08 06.
Article En | MEDLINE | ID: mdl-30086798

A wide spectrum of scenarios may lead to radiation incidents and the liberation of radioactive material. In the case of a terrorist attack by a "dirty bomb", there is a risk of mechanical and thermal trauma, external irradiation, superficial contamination and incorporation of radioactive material. The first treatment priority must be given to the care of trauma patients with life-threatening injuries, as the health effects of radiation occur with latency. Radionuclide incorporation will lead to a longer-lasting irradiation from inside the body, associated with a higher risk of stochastic radiation effects (e.g., occurrence of tumors) in the long run. It must be expected that victims with potentially incorporated radionuclides will far outnumber trauma patients. The elimination of radionuclides can be enhanced by the administration of decorporation agents such as (Ca) Diethylenetriaminepentaacetic acid (DTPA) or Prussian blue, reducing the radiological burden of the body. There is still no consensus whether decorporation treatment should be started immediately based only on a suspicion of radionuclide incorporation ("urgent approach") or if the results of internal dosimetry confirming the necessity of a treatment should be awaited, accepting the delay caused by the measurements and computations ("precautionary approach"). As the therapeutic effectiveness may be substantially decreased if treatment initiation is delayed only by several days, depending on the radionuclide, the physicochemical properties of the compounds involved and the route of absorption, we favor an "urgent approach" from a medical point of view. In doubt, it seems justified to treat victims by precaution, as the adverse effects of the medication seem minimal. However, in the case of a high number of victims, an "urgent treatment approach" may require a large number of daily doses of antidotes, and therefore, adequate investments in preparedness and antidote stockpiling are necessary.


Decontamination/methods , Nuclear Weapons , Radiation Injuries/therapy , Radiation-Protective Agents/therapeutic use , Radioactive Hazard Release , Animals , Antidotes/therapeutic use , Civil Defense , Disaster Medicine , Ferrocyanides/therapeutic use , Humans , Pentetic Acid/therapeutic use , Radioisotopes/chemistry , Radiometry , Terrorism
16.
ACS Nano ; 12(3): 2858-2872, 2018 03 27.
Article En | MEDLINE | ID: mdl-29510031

Tumor hypoxia is one of the major challenges for the treatment of tumors, as it may negatively affect the efficacy of various anticancer modalities. In this study, a tumor-targeted redox-responsive composite biocatalyst is designed and fabricated, which may combine tumor starvation therapy and low-temperature photothermal therapy for the treatment of oxygen-deprived tumors. The nanosystem was prepared by loading porous hollow Prussian Blue nanoparticles (PHPBNs) with glucose oxidase (GOx) and then coating their surface with hyaluronic acid (HA) via redox-cleavable linkage, therefore allowing the nanocarrier to bind specifically with CD44-overexpressing tumor cells while also exerting control over the cargo release profile. The nanocarriers are designed to enhance the efficacy of the hypoxia-suppressed GOx-mediated starvation therapy by catalyzing the decomposition of intratumoral hydroperoxide into oxygen with PHPBNs, and the enhanced glucose depletion by the two complementary biocatalysts may consequently suppress the expression of heat shock proteins (HSPs) after photothermal treatment to reduce their resistance to the PHPBN-mediated low-temperature photothermal therapies.


Ferrocyanides/therapeutic use , Glucose Oxidase/therapeutic use , Hyperthermia, Induced/methods , Nanoparticles/therapeutic use , Neoplasms/therapy , Animals , Drug Delivery Systems , Glucose/metabolism , Hep G2 Cells , Humans , Hydrogen Peroxide/metabolism , Mice , Mice, Nude , Nanoparticles/ultrastructure , Neoplasms/metabolism , Neoplasms/pathology , Oxygen/metabolism , Phototherapy/methods , Temperature
17.
Drug Res (Stuttg) ; 68(2): 89-99, 2018 02.
Article En | MEDLINE | ID: mdl-29036734

In the case of an attack by a "dirty bomb" with cesium-137 there is a risk of internal contamination. The excretion of cesium-137 can be enhanced by Prussian Blue (PB), and thus the committed effective dose be reduced. We analyzed the benefit and costs of PB decorporation treatment. We simulated the reduction of the radiological dose by PB treatment after cesium-137 incorporation by inhalation. The saving of life time was quantified using the monetary "value of a statistical life" (VSL). Treatment costs were based on the market price of PB in Germany. Moreover we considered the holding costs of stockpiling. The benefit of PB treatment increases with its duration up to about 90 days. If treatment initiation is delayed, the maximum achievable benefit is decreased. For a VSL of 1.646 million €, the net benefit of a 90-days treatment started 1 day after the incorporation remains positive up to a stockpiling duration of 10 years. If starting PB treatment as late as the 180th day after incorporation, the costs will surpass the benefit. We conclude that a prompt decision making and early treatment initiation greatly impacts on the medical but also economic efficiency of a PB treatment.


Cesium Radioisotopes/toxicity , Cost-Benefit Analysis/statistics & numerical data , Decontamination/economics , Ferrocyanides/economics , Strategic Stockpile/economics , Ferrocyanides/therapeutic use , Health Care Costs/statistics & numerical data , Humans , Radiation-Protective Agents/economics , Time Factors
18.
Nanomedicine (Lond) ; 11(14): 1759-67, 2016 07.
Article En | MEDLINE | ID: mdl-27389189

AIM: To engineer a novel nanoimmunotherapy comprising Prussian blue nanoparticles (PBNPs) conjugated to antigen-specific cytotoxic T lymphocytes (CTL), which leverages PBNPs for their photothermal therapy (PTT) capabilities and Epstein-Barr virus (EBV) antigen-specific CTL for their ability to traffic to and destroy EBV antigen-expressing target cells. MATERIALS & METHODS: PBNPs and CTL were independently biofunctionalized. Subsequently, PBNPs were conjugated onto CTL using avidin-biotin interactions. The resultant cell-nanoparticle construct (CTL:PBNPs) were analyzed for their physical, phenotypic and functional properties. RESULTS: Both PBNPs and CTL maintained their intrinsic physical, phenotypic and functional properties within the CTL:PBNPs. CONCLUSION: This study highlights the potential of our CTL:PBNPs nanoimmunotherapy as a novel therapeutic for treating virus-associated malignancies such as EBV+ cancers.


Coloring Agents/therapeutic use , Epstein-Barr Virus Infections/therapy , Ferrocyanides/therapeutic use , Herpesvirus 4, Human/immunology , Nanoparticles/therapeutic use , Neoplasms/therapy , Neoplasms/virology , T-Lymphocytes, Cytotoxic/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/virology , Coculture Techniques , Coloring Agents/chemistry , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/immunology , Ferrocyanides/chemistry , Humans , Immunotherapy , Jurkat Cells , Lymphocyte Activation , Nanomedicine , Nanoparticles/chemistry , Neoplasms/immunology , Phototherapy , T-Lymphocytes, Cytotoxic/chemistry
19.
Dalton Trans ; 45(44): 17581-17587, 2016 Nov 28.
Article En | MEDLINE | ID: mdl-27278267

Prussian blue type nanoparticles are exciting nano-objects that combine the advantages of molecule-based materials and nanochemistry. Here we provide a short overview focalizing on the recent advances of these nano-objects designed for biomedical applications and give an outlook on the future research orientations in this domain.


Coloring Agents/analysis , Coloring Agents/therapeutic use , Ferrocyanides/analysis , Ferrocyanides/therapeutic use , Nanoparticles/analysis , Nanoparticles/therapeutic use , Animals , Humans , Nanomedicine/methods , Nanoparticles/ultrastructure , Nanotechnology/methods , Neoplasms/diagnostic imaging , Neoplasms/therapy , Optical Imaging/methods , Tomography, Emission-Computed, Single-Photon/methods
20.
ACS Appl Mater Interfaces ; 7(21): 11575-82, 2015 Jun 03.
Article En | MEDLINE | ID: mdl-25965554

Prussian blue (PB) as a clinically adapted agent recently has drawn much attention in cancer theranostics for potential applications in magnetic resonance (MR) imaging as well as photothermal cancer treatment. In this work, we take a closer look at the imaging and therapy performance of PB agents once they are doped with Mn2+. It is found that Mn2+-doped PB nanocubes exhibit increased longitudinal relaxivity along with enhanced optical absorption red-shifted to the near-infrared (NIR) region. Those properties make PB:Mn nanocubes with appropriate surface coatings rather attractive agents for biomedical imaging and cancer therapy, which have been successfully demonstrated in our in vivo experiments for effectively tumor ablation.


Ferrocyanides/therapeutic use , Magnetic Resonance Imaging/methods , Manganese/chemistry , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Photochemotherapy/methods , Animals , Cell Line, Tumor , Contrast Media/chemistry , Ferrocyanides/chemistry , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Metal Nanoparticles/ultrastructure , Mice , Mice, Inbred BALB C , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Nanocapsules/ultrastructure , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemistry , Theranostic Nanomedicine/methods , Treatment Outcome
...