Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
Am J Hematol ; 99(6): 1077-1083, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38476079

RESUMEN

Restless legs syndrome (RLS) is a neurological disorder that can have a profound effect on sleep and quality of life. Idiopathic RLS is associated with brain iron insufficiency despite normal peripheral iron stores. There is, however, a five- to six-fold increase in prevalence of RLS in patients with iron deficiency anemia (IDA). Several open-label trials have demonstrated symptomatic improvement in RLS following treatment of IDA using oral or intravenous iron supplementation. To date, there have been no randomized double-blind controlled trials of intravenous iron compared with oral iron for the treatment of RLS patients with IDA. In the current study, oral ferrous sulfate and ferumoxytol were compared for efficacy and speed of response for treatment of RLS occurring in patients with IDA. The planned recruitment for this study was 70 patients with RLS and IDA, to be randomly assigned 1:1 to oral or intravenous iron, using double-blind, double-dummy procedures. At Week 6, the primary outcomes of Clinical Global Impression-Improvement score and change from baseline in the International Restless Legs Syndrome Study Group rating scale score were assessed. Due to challenges, performing the clinical trial during the COVID-19 pandemic, final-week data were found missing for 30 patients. As a result, in order to maintain the prespecified statistical analysis, an additional 30 patients were recruited. Both IV and oral iron were associated with a marked improvement in RLS symptoms, with no statistically significant difference between treatment groups. No serious adverse events were observed in either treatment group.


Asunto(s)
Administración Intravenosa , Anemia Ferropénica , Compuestos Ferrosos , Síndrome de las Piernas Inquietas , Humanos , Síndrome de las Piernas Inquietas/tratamiento farmacológico , Anemia Ferropénica/tratamiento farmacológico , Administración Oral , Método Doble Ciego , Masculino , Femenino , Proyectos Piloto , Persona de Mediana Edad , Compuestos Ferrosos/administración & dosificación , Compuestos Ferrosos/uso terapéutico , Compuestos Ferrosos/efectos adversos , Adulto , Anciano , Resultado del Tratamiento , Óxido Ferrosoférrico/administración & dosificación , Óxido Ferrosoférrico/uso terapéutico , Óxido Ferrosoférrico/efectos adversos , Hierro/administración & dosificación , Hierro/uso terapéutico
2.
Adv Healthc Mater ; 13(6): e2302773, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37931150

RESUMEN

Ferumoxytol, approved by the U.S. Food and Drug Administration in 2009, is one of the intravenous iron oxide nanoparticles authorized for the treatment of iron deficiency in chronic kidney disease and end-stage renal disease. With its exceptional magnetic properties, catalytic activity, and immune activity, as well as good biocompatibility and safety, ferumoxytol has gained significant recognition in various biomedical diagnoses and treatments. Unlike most existing reviews on this topic, this review primarily focuses on the recent clinical and preclinical advances of ferumoxytol in disease treatment, spanning anemia, cancer, infectious inflammatory diseases, regenerative medicine application, magnetic stimulation for neural modulation, etc. Additionally, the newly discovered mechanisms associated with the biological effects of ferumoxytol are discussed, including its magnetic, catalytic, and immunomodulatory properties. Finally, the summary and future prospects concerning the treatment and application of ferumoxytol-based nanotherapeutics are presented.


Asunto(s)
Óxido Ferrosoférrico , Inmunomodulación , Estados Unidos , Óxido Ferrosoférrico/uso terapéutico , Administración Intravenosa , Catálisis , Medicina Regenerativa
3.
Int J Nanomedicine ; 18: 4067-4100, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37525695

RESUMEN

Magnetic iron oxide nanoparticles (magnetite and maghemite) are intensively studied due to their broad potential applications in medical and biological sciences. Their unique properties, such as nanometric size, large specific surface area, and superparamagnetism, allow them to be used in targeted drug delivery and internal radiotherapy by targeting an external magnetic field. In addition, they are successfully used in magnetic resonance imaging (MRI), hyperthermia, and radiolabelling. The appropriate design of nanoparticles allows them to be delivered to the desired tissues and organs. The desired biodistribution of nanoparticles, eg, cancerous tumors, is increased using an external magnetic field. Thus, knowledge of the biodistribution of these nanoparticles is essential for medical applications. It allows for determining whether nanoparticles are captured by the desired organs or accumulated in other tissues, which may lead to potential toxicity. This review article presents the main organs where nanoparticles accumulate. The sites of their first uptake are usually the liver, spleen, and lymph nodes, but with the appropriate design of nanoparticles, they can also be accumulated in organs such as the lungs, heart, or brain. In addition, the review describes the factors affecting the biodistribution of nanoparticles, including their size, shape, surface charge, coating molecules, and route of administration. Modern techniques for determining nanoparticle accumulation sites and concentration in isolated tissues or the body in vivo are also presented.


Asunto(s)
Nanopartículas de Magnetita , Nanopartículas , Neoplasias , Humanos , Nanopartículas de Magnetita/uso terapéutico , Nanopartículas de Magnetita/química , Distribución Tisular , Compuestos Férricos/química , Nanopartículas Magnéticas de Óxido de Hierro , Óxido Ferrosoférrico/uso terapéutico , Neoplasias/tratamiento farmacológico , Nanopartículas/química , Imagen por Resonancia Magnética/métodos
4.
Nanomedicine ; 52: 102695, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37394106

RESUMEN

Chitosan-functionalized magnetite/poly(ε-caprolactone) nanoparticles were formulated by interfacial polymer disposition plus coacervation, and loaded with gemcitabine. That (core/shell)/shell nanostructure was confirmed by electron microscopy, elemental analysis, electrophoretic, and Fourier transform infrared characterizations. A short-term stability study proved the protection against particle aggregation provided by the chitosan shell. Superparamagnetic properties of the nanoparticles were characterized in vitro, while the definition of the longitudinal and transverse relaxivities was an initial indication of their capacity as T2 contrast agents. Safety of the particles was demonstrated in vitro on HFF-1 human fibroblasts, and ex vivo on SCID mice. The nanoparticles demonstrated in vitro pH- and heat-responsive gemcitabine release capabilities. In vivo magnetic resonance imaging studies and Prussian blue visualization of iron deposits in tissue samples defined the improvement in nanoparticle targeting into the tumor when using a magnetic field. This tri-stimuli (magnetite/poly(ε-caprolactone))/chitosan nanostructure could find theranostic applications (biomedical imaging & chemotherapy) against tumors.


Asunto(s)
Quitosano , Nanopartículas de Magnetita , Nanopartículas , Neoplasias , Ratones , Animales , Humanos , Óxido Ferrosoférrico/uso terapéutico , Quitosano/uso terapéutico , Medicina de Precisión , Ratones SCID , Nanopartículas de Magnetita/química , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Gemcitabina , Imagen por Resonancia Magnética/métodos
5.
Biomater Sci ; 11(15): 5337-5346, 2023 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-37366334

RESUMEN

Nanomaterials have been identified as a potential therapeutic option for targeting myeloid-derived suppressor cells (MDSCs), which are known to play a crucial role in tumor metastasis and treatment resistance. Here, we report a unique immunologically active nanomaterial composed of ferumoxytol and poly(I : C) (FP-NPs) and investigate its immunoregulatory activities on MDSCs in metastatic melanoma. In vivo assays demonstrated that FP-NPs had the ability to significantly impede the progression of metastatic melanoma and decrease the MDSC population in the lungs, spleen, and bone marrow of mice. Both in vivo and in vitro experiments revealed that FP-NPs reduced the number of granulocytic MDSCs and promoted the differentiation of monocytic MDSCs into anti-tumor M1 macrophages. Transcriptome sequencing indicated that FP-NPs significantly altered the expression of several genes involved in immunity. Analysis of Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and quantitative real-time PCR revealed that FP-NPs significantly increased the expression of the myeloid cell differentiation-related gene interferon regulatory factor 7 and activated interferon beta-related signaling pathways, which stimulated the differentiation of MDSCs into M1 macrophages. These findings suggest that FP-NPs, a unique nanomaterial with immunological properties, can induce MDSCs to differentiate into M1 macrophages, potentially offering new treatment prospects for metastatic melanoma in the future.


Asunto(s)
Melanoma , Células Supresoras de Origen Mieloide , Animales , Ratones , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/patología , Óxido Ferrosoférrico/uso terapéutico , Melanoma/tratamiento farmacológico , Diferenciación Celular , Macrófagos/metabolismo
6.
Theranostics ; 13(6): 1745-1758, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37064879

RESUMEN

Rationale: As a cancer, Glioblastoma (GBM) is a highly lethal and difficult-to-treat. With the aim of improving therapies to GBM, we developed novel and target-specific theranostic nanoparticles (TNPs) that can be selectively cleaved by cathepsin B (Cat B) to release the potent toxin monomethyl auristatin E (MMAE). Methods: We synthesized TNPs composed of a ferumoxytol-based nanoparticle carrier and a peptide prodrug with a Cat-B-responsive linker and the tubulin inhibitor MMAE. We hypothesized that intratumoral Cat B can cleave our TNPs and release MMAE to kill GBM cells. The ferumoxytol core enables in vivo drug tracking with magnetic resonance imaging (MRI). We incubated U87-MG GBM cells with TNPs or ferumoxytol and evaluated the TNP content in the cells with transmission electron microscopy and Prussian blue staining. In addition, we stereotaxically implanted 6- to 8-week-old nude mice with U87-MG with U87-MG GBM cells that express a fusion protein of Green Fluorescence Protein and firefly Luciferase (U87-MG/GFP-fLuc). We then treated the animals with an intravenous dose of TNPs (25 mg/kg of ferumoxytol, 0.3 mg/kg of MMAE) or control. We also evaluated the combination of TNP treatment with radiation therapy. We performed MRI before and after TNP injection. We compared the results for tumor and normal brain tissue between the TNP and control groups. We also monitored tumor growth for a period of 21 days. Results: We successfully synthesized TNPs with a hydrodynamic size of 41 ± 5 nm and a zeta potential of 6 ± 3 mV. TNP-treated cells demonstrated a significantly higher iron content than ferumoxytol-treated cells (98 ± 1% vs. 3 ± 1% of cells were iron-positive, respectively). We also found significantly fewer live attached cells in the TNP-treated group (3.8 ± 2.0 px2) than in the ferumoxytol-treated group (80.0 ± 14.5 px2, p < 0001). In vivo MRI studies demonstrated a decline in the tumor signal after TNP (T2= 28 ms) but not control (T2= 32 ms) injections. When TNP injection was combined with radiation therapy, the tumor signals dropped further (T2 = 24 ms). The combination therapy of radiation therapy and TNPs extended the median survival from 14.5 days for the control group to 45 days for the combination therapy group. Conclusion: The new cleavable TNPs reported in this work accumulate in GBM, cause tumor cell death, and have synergistic effects with radiation therapy.


Asunto(s)
Glioblastoma , Nanopartículas , Ratones , Animales , Glioblastoma/diagnóstico por imagen , Glioblastoma/tratamiento farmacológico , Medicina de Precisión , Óxido Ferrosoférrico/uso terapéutico , Péptido Hidrolasas , Ratones Desnudos , Imagen por Resonancia Magnética , Nanopartículas/química , Endopeptidasas , Hierro , Línea Celular Tumoral
7.
Nanomedicine ; 49: 102658, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36708910

RESUMEN

Angiogenesis plays a key role in the progression and metastasis of melanoma, and the pro-angiogenic effect of macrophages is one major reason for the failure of current anti-angiogenic therapies. Here, a nano-immunotherapy combining ferumoxytol and poly(I:C) (ferumoxytol/poly(I:C)) has been developed to boost the anti-angiogenic activities of macrophages to inhibit melanoma. Our findings demonstrated that ferumoxytol/poly(I:C) was a highly efficacious anti-tumor therapy with limited toxicity. Both in vivo and in vitro experiments indicated that this combination was successful in impeding angiogenesis. Ferumoxytol/poly(I:C) was demonstrated to reduce the viability of endothelial cells, thus hindering tube formation. Particularly, ferumoxytol/poly(I:C) was able to polarize macrophages to the M1 phenotype and decrease the expression of vascular endothelial growth factor, which in turn amplified the anti-angiogenic properties of ferumoxytol/poly(I:C). This combination of ferumoxytol/poly(I:C) nano-immunotherapy enriches the anti-angiogenic therapeutic nature of ferumoxytol and will shed new light on the treatment of melanoma.


Asunto(s)
Antiinfecciosos , Melanoma , Humanos , Óxido Ferrosoférrico/farmacología , Óxido Ferrosoférrico/uso terapéutico , Factor A de Crecimiento Endotelial Vascular , Células Endoteliales/metabolismo , Melanoma/patología , Factores de Crecimiento Endotelial Vascular , Penicilinas/uso terapéutico , Antiinfecciosos/uso terapéutico
8.
Afr Health Sci ; 23(3): 205-212, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38357109

RESUMEN

Introduction: The efficacy of iron isomaltoside ferumoxytol versus iron sucrose to treat iron deficiency anemia remains controversial. We conduct this meta-analysis to explore the influence of iron isomaltoside ferumoxytol versus iron sucrose on iron deficiency anemia. Methods: We have searched PubMed, EMbase, Web of science, EBSCO, and Cochrane library databases through March 2021 for randomized controlled trials (RCTs) assessing the effect of iron isomaltoside ferumoxytol versus iron sucrose on iron deficiency anemia. Meta-analysis was performed using the random-effect model. Results: Four RCTs involving 3892 patients were included in the meta-analysis. Overall, compared with iron sucrose for iron deficiency anemia, iron isomaltoside showed similar change of Hb (SMD=0.14; 95% CI=-0.07 to 0.35; P=0.18), Hb responder (SMD=1.41; 95% CI=0.71 to 2.81; P=0.33), serum ferritin (SMD=15.13; 95% CI=-23.45 to 53.71; P=0.44), and transferrin saturation (SMD=1.20; 95% CI=-1.08 to 3.47; P=0.30). However, iron isomaltoside further improved serum-ferritin at week 2 than iron sucrose (SMD=204.79; 95% CI=38.23 to 371.35; P=0.02). Conclusions: Iron isomaltoside ferumoxytol showed comparable efficacy to iron sucrose for the treatment of iron deficiency anemia.


Asunto(s)
Anemia Ferropénica , Disacáridos , Compuestos Férricos , Óxido Ferrosoférrico , Humanos , Anemia Ferropénica/tratamiento farmacológico , Sacarato de Óxido Férrico , Ferritinas , Óxido Ferrosoférrico/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto
9.
Sci Rep ; 12(1): 15045, 2022 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-36057729

RESUMEN

Mucopolysaccharidosis IV A (MPS IVA) is a lysosomal disorder caused by mutations in the GALNS gene. Consequently, the glycosaminoglycans (GAGs) keratan sulfate and chondroitin 6-sulfate accumulate in the lysosomal lumen. Although enzyme replacement therapy has shown essential advantages for the patients, several challenges remain to overcome, such as the limited impact on the bone lesion and recovery of oxidative profile. Recently, we validated a CRISPR/nCas9-based gene therapy with promising results in an in vitro MPS IVA model. In this study, we have expanded the use of this CRISPR/nCas9 system to several MPS IVA fibroblasts carrying different GALNS mutations. Considering the latent need to develop more safety vectors for gene therapy, we co-delivered the CRISPR/nCas9 system with a novel non-viral vector based on magnetoliposomes (MLPs). We found that the CRISPR/nCas9 treatment led to an increase in enzyme activity between 5 and 88% of wild-type levels, as well as a reduction in GAGs accumulation, lysosomal mass, and mitochondrial-dependent oxidative stress, in a mutation-dependent manner. Noteworthy, MLPs allowed to obtain similar results to those observed with the conventional transfection agent lipofectamine. Overall, these results confirmed the potential of CRISPR/nCas9 as a genome editing tool for treating MPS IVA. We also demonstrated the potential use of MLPs as a novel delivery system for CRISPR/nCas9-based therapies.


Asunto(s)
Condroitinsulfatasas , Mucopolisacaridosis , Mucopolisacaridosis IV , Nanopartículas , Condroitinsulfatasas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Óxido Ferrosoférrico/uso terapéutico , Edición Génica , Glicosaminoglicanos , Humanos , Mucopolisacaridosis/genética , Mucopolisacaridosis/terapia , Mucopolisacaridosis IV/genética , Mucopolisacaridosis IV/terapia
10.
Ren Fail ; 44(1): 94-102, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35156909

RESUMEN

PURPOSE: To evaluate the efficacy of ferumoxytol, relative to conventional iron supplement formulations, on hemoglobin levels, ferritin level, and adverse event incidence in chronic kidney disease patients. METHODS: We performed a systematic search of six academic databases (EMBASE, CENTRAL, Scopus, PubMed, Web of sciences, and MEDLINE), adhering to PRISMA guidelines. We performed a meta-analysis on relevant studies to evaluate the overall influence of ferumoxytol, relative to conventional iron supplement formulations, on hemoglobin levels, ferritin level, and treatment related treatment emergent adverse events (TEAEs) incidence in chronic kidney disease patients. RESULTS: Seven eligible studies were identified from a total of 1397 studies. These studies contained data on 3315 participants with chronic kidney disease (mean age: 59.2 ± 4.6 years). A meta-analysis revealed that ferumoxytol administration had positive effects on hemoglobin levels (Hedge's g statistic: 0.51) and ferritin level (0.88), transferrin saturation (0.39). Besides, we also report reduced incidence of treatment related TEAEs (-0.24) for patients consuming ferumoxytol as compared conventional iron supplement formulations. CONCLUSIONS: This meta-analysis provides preliminary evidence that ferumoxytol use exerts beneficial effects on the overall hematological outcomes in patients with chronic kidney disease. This study also reports improved treatment related safety profile for ferumoxytol when compared with conventional iron formulations. The findings from this study can have direct implications in forming best practice guidelines for managing anemia in patients with chronic kidney disease.


Asunto(s)
Anemia Ferropénica/tratamiento farmacológico , Óxido Ferrosoférrico/uso terapéutico , Hematínicos/uso terapéutico , Insuficiencia Renal Crónica/complicaciones , Anemia Ferropénica/etiología , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto , Diálisis Renal , Insuficiencia Renal Crónica/terapia
11.
Theranostics ; 12(2): 675-688, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34976207

RESUMEN

Background: Innate immune memory, also termed "trained immunity", is thought to protect against experimental models of infection, including sepsis. Trained immunity via reprogramming monocytes/macrophages has been reported to result in enhanced inflammatory status and antimicrobial activity against infection in sepsis. However, a safe and efficient way to induce trained immunity remains unclear. Methods: ß-glucan is a prototypical agonist for inducing trained immunity. Ferumoxytol, superparamagnetic iron oxide (SPIO) with low cytotoxicity, has been approved by FDA for clinical use. We synthesized novel nanoparticles BSNPs by coupling ß-glucan with SPIO. BSNPs were further conjugated with fluorescein for quantitative analysis and trace detection of ß-glucan on BSNPs. Inflammatory cytokine levels were measured by ELISA and qRT-PCR, and the phagocytosis of macrophages was detected by flow cytometry and confocal microscopy. The therapeutic effect of BSNPs was evaluated on the well-established sepsis mouse model induced by both clinical Escherichia coli (E. coli) and cecal ligation and puncture (CLP). Results: BSNPs were synthesized successfully with a 3:20 mass ratio of ß-glucan and SPIO on BSNPs, which were mainly internalized by macrophages and accumulated in the lungs and livers of mice. BSNPs effectively reprogrammed macrophages to enhance the production of trained immunity markers and phagocytosis toward bacteria. BSNP-induced trained immunity protected mice against sepsis caused by E. coli and CLP and also against secondary infection. We found that BSNP treatment elevated Akt, S6, and 4EBP phosphorylation, while mTOR inhibitors decreased the trained immunity markers and phagocytosis enhanced by BSNPs. Furthermore, the PCR Array analysis revealed Igf1, Sesn1, Vegfa, and Rps6ka5 as possible key regulators of mTOR signaling during trained immunity. BSNP-induced trained immunity mainly regulated cellular signal transduction, protein modification, and cell cycle by modulating ATP binding and the kinase activity. Our results indicated that BSNPs induced trained immunity in an mTOR-dependent manner. Conclusion: Our data highlight that the trained immunity of macrophages is an effective strategy against sepsis and suggest that BSNPs are a powerful tool for inducing trained immunity to prevent and treat sepsis and secondary infections.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Óxido Ferrosoférrico/uso terapéutico , Nanopartículas Magnéticas de Óxido de Hierro , Sepsis/inmunología , Animales , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/prevención & control , Femenino , Inmunidad Innata , Memoria Inmunológica , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Nanopartículas Magnéticas de Óxido de Hierro/química , Ratones , Ratones Endogámicos ICR , Fagocitosis/efectos de los fármacos , Sustancias Protectoras/uso terapéutico , Sepsis/prevención & control , beta-Glucanos/química , beta-Glucanos/uso terapéutico
12.
Theranostics ; 12(2): 796-816, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34976214

RESUMEN

Ferumoxytol is an intravenous iron oxide nanoparticle formulation that has been approved by the U.S. Food and Drug Administration (FDA) for treating anemia in patients with chronic kidney disease. In recent years, ferumoxytol has also been demonstrated to have potential for many additional biomedical applications due to its excellent inherent physical properties, such as superparamagnetism, biocatalytic activity, and immunomodulatory behavior. With good safety and clearance profiles, ferumoxytol has been extensively utilized in both preclinical and clinical studies. Here, we first introduce the medical needs and the value of current iron oxide nanoparticle formulations in the market. We then focus on ferumoxytol nanoparticles and their physicochemical, diagnostic, and therapeutic properties. We include examples describing their use in various biomedical applications, including magnetic resonance imaging (MRI), multimodality imaging, iron deficiency treatment, immunotherapy, microbial biofilm treatment and drug delivery. Finally, we provide a brief conclusion and offer our perspectives on the current limitations and emerging applications of ferumoxytol in biomedicine. Overall, this review provides a comprehensive summary of the developments of ferumoxytol as an agent with diagnostic, therapeutic, and theranostic functionalities.


Asunto(s)
Reposicionamiento de Medicamentos , Óxido Ferrosoférrico/uso terapéutico , Nanopartículas del Metal/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Aprobación de Drogas , Humanos , Hierro , Deficiencias de Hierro/tratamiento farmacológico , Imagen por Resonancia Magnética , Nanopartículas del Metal/química , Medicina de Precisión , Estados Unidos , United States Food and Drug Administration
13.
Int J Med Sci ; 18(14): 3125-3139, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34400883

RESUMEN

Background: Regulating the polarization of macrophages to antitumor M1 macrophages is a promising strategy for overcoming the immunosuppression of the tumor microenvironment for cancer therapy. Ferumoxytol (FMT) can not only serve as a drug deliver agent but also exerts anti-tumor activity. ß-glucan has immuno-modulating properties to prevent tumor growth. Thus, a nanocomposite of FMT surface-coated with ß-glucan (FMT-ß-glucan) was prepared to explore its effect on tumor suppression. Methods: Male B16F10 melanoma mouse model was established to explore the antitumor effect of FMT-ß-glucan. The viability and apoptotic rates of B16F10 cells were detected by cell counting kit-8 and Annexin-V/PI experiments. The levels of M1 markers were quantified by quantitative reverse transcription-polymerase chain reaction and enzyme linked immunosorbent assay. Phagocytic activity and intracellular reactive oxygen species (ROS) in macrophages were evaluated by the neutral red uptake assay and flow cytometry, respectively. Small interfering RNA (siRNA) transfection was applied to knock down the Dectin-1 gene in RAW 264.7 cells. Results: FMT-ß-glucan suppressed tumor growth to a greater extent and induced higher infiltration of M1 macrophages than the combination of FMT and ß-glucan (FMT+ß-glucan) in vivo. In vitro, supernatant from FMT-ß-glucan-treated RAW 264.7 cells led to lower cell viability and induced more apoptosis of B16F10 cells than that from the FMT+ß-glucan group. Moreover, FMT-ß-glucan boosted the expression of M1 type markers, and increased phagocytic activity and ROS in RAW 264.7 cells. Further research indicated that FMT-ß-glucan treatment promoted the level of Dectin-1 on the surface of RAW 264.7 cells and that knockdown of Dectin-1 abrogated the phosphorylation levels of several components in MAPK and NF-κB signaling. Conclusion: The nanocomposite FMT-ß-glucan suppressed melanoma growth by inducing the M1 macrophage-activated tumor microenvironment.


Asunto(s)
Óxido Ferrosoférrico/farmacología , Lectinas Tipo C/agonistas , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , beta-Glucanos/farmacología , Animales , Modelos Animales de Enfermedad , Óxido Ferrosoférrico/química , Óxido Ferrosoférrico/uso terapéutico , Técnicas de Silenciamiento del Gen , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Melanoma/inmunología , Melanoma/patología , Ratones , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Escape del Tumor/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , beta-Glucanos/química , beta-Glucanos/uso terapéutico
14.
Biomed Pharmacother ; 139: 111566, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33839494

RESUMEN

BACKGROUND: In recent years, magnetic nanoparticles (NMP) as novel materials have been widely used for biomedical, diagnostic and therapeutic purposes like microbial infection therapy. The purpose of this study is to synthesize PO coated iron oxide magnetic nanoparticles (Fe3O4@PO NPs) and their anti-leishmanial effects in vitro and in vivo against cutaneous leishmaniasis. METHODS: Fe3O4 magnetic nanoparticles were synthesized by the coprecipitation of Fe2 + and Fe3 + ions and used as a nanocarrier for the production of Fe3O4@PO NPs. The in vitro antileishmanial effects of PO-coated Fe3O4 NPs and Fe3O4 NPs (10-200 µg/mL) was determined against the intracellular amastigotes of Leishmania major (MRHO/IR/75/ER) and, then, examined on cutaneous leishmaniasis induced in male BALB/c mice by L. major. The rate of infectivity, production of nitric oxide (NO), and cytotoxic activates of Fe3O4 NPs and Fe3O4@PO NPs on J774-A1 macrophage cells were determined. RESULTS: The size scattering of the Fe3O4 NPs and Fe3O4@PO NPs were in the range among 1-40 and 5-55 nm, respectively. The obtained IC50 values were 62.3 ± 2.15 µg/mL, 31.3 ± 2.26 µg/mL, and 52.6 ± 2.15 µg/mL for the Fe3O4 NPs and Fe3O4@PO NPs, and MA, respectively. The results revealed that the mean number of parasites and the mean diameter of the lesions was considerably (p < 0.05) decreased in the infected mice treated with Fe3O4 NPs and Fe3O4@PO NPs. The Fe3O4 NPs and Fe3O4@PO NPs significantly (p < 0.05) prompted the production of NO as a dose-dependent manner. The promastigotes pre-incubated in Fe3O4 NPs and Fe3O4@PO NPs at the concentration of 5 µg/mL had the ability to infect only 41.7% and 28.3% of the macrophages cells. The selectivity index of greater than 10 for Fe3O4 NPs and Fe3O4@PO NPs showed its safety to the J774-A1 macrophage cells and specificity to the parasite. CONCLUSION: The results of this survey indicated the high potency of Fe3O4@PO NPs to inhibit the growth of amastigote forms of L. major as well as recovery and improvement CL induced by L. major in BALB/c mice without significant cytotoxicity. The results also indicated that, although the possible anti-leishmanial mechanisms of Fe3O4@PO NPs have not been clearly understood, however, the triggering of NO may be considered as one of the possible anti-leishmanial mechanisms of these nanoparticles. However, additional studies, in particular in clinical contexts, are mandatory.


Asunto(s)
Antiprotozoarios/uso terapéutico , Etanolaminas/química , Óxido Ferrosoférrico/uso terapéutico , Leishmaniasis Cutánea/tratamiento farmacológico , Nanopartículas de Magnetita/uso terapéutico , Piridonas/química , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Portadores de Fármacos , Combinación de Medicamentos , Óxido Ferrosoférrico/administración & dosificación , Óxido Ferrosoférrico/química , Leishmania major/efectos de los fármacos , Leishmaniasis Cutánea/parasitología , Macrófagos/parasitología , Nanopartículas de Magnetita/química , Masculino , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico/biosíntesis
15.
N Z Med J ; 134(1534): 118-127, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33927444

RESUMEN

There are several newer intravenous iron formulations to treat iron deficiency and its anaemia. Its use in the primary care setting has been infrequent compared to tertiary centres, due to historical concerns such as anaphylaxis. There is a lack of overall comparison among the intravenous formulations of iron. Compared to oral iron therapy, the newer intravenous formulations, which allow a complete or near-complete replacement in a single sitting of 15-30 minutes, have an improved safety profile with better tolerability, efficacy and effectiveness. They are suited for administration in the primary care setting. The four non-dextran formulations (ferric carboxymaltose, iron sucrose, iron isomaltoside and ferumoxytol) share an equal or near equal efficacy and safety profile. This article also outlines how to provide iron infusion safely and effectively in the community.


Asunto(s)
Anemia Ferropénica/tratamiento farmacológico , Compuestos Férricos/uso terapéutico , Óxido Ferrosoférrico/uso terapéutico , Compuestos Ferrosos/uso terapéutico , Complejo Hierro-Dextran/uso terapéutico , Humanos , Infusiones Intravenosas , Nueva Zelanda
16.
Clin Nephrol ; 95(4): 189-194, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33433318

RESUMEN

BACKGROUND: Intravenous iron is one of the main therapies for anemia management in hemodialysis-dependent patients. Data comparing the efficacy of ferumoxytol versus other parenteral iron supplements are scarce. The objective of the study was to compare the efficacy of ferumoxytol with that of sodium ferric gluconate in outpatient hemodialysis patients. MATERIALS AND METHODS: A prospective, observational study was conducted in outpatients receiving ferumoxytol 510 mg once or twice quarterly compared to sodium ferric gluconate 125 mg weekly in a single center hemodialysis center in Ontario, Canada. Patient demographics, hemoglobin levels, iron indices, iron doses, and erythropoiesis-stimulating agent (ESA) doses were collected. RESULTS: The study sample consisted of 291 observations from 173 patients. Generalized estimating equations of multiple linear regression modeling were conducted to compare the outcomes while adjusting for baseline scores. Approximately 25% of the study participants received ferumoxytol while 75% received sodium ferric gluconate. Patients treated were mainly males (58.4%), and the mean age was 68.73 (SD ± 13.03) years. Both groups did not show significant differences in their hemoglobin levels (Wald z = 0.54; p = 0.46), ESA utilization at 3 months (Wald z = 0.20; p = 0.65), and TSAT levels (Wald z = 3.45; p = 0.06). However, the iron levels (Wald z = 4.24; p = 0.04) and ferritin levels (Wald z = 5.14; p = 0.02) were higher in the ferric gluconate group (Wald z = 58.78; p ≤ 0.001), and patients who received ferumoxytol received more blood transfusions as compared to those who received sodium ferric gluconate (χ2 = 16.71; p ≤ 0.001). CONCLUSION: Both iron products maintained hemoglobin levels, but patients receiving ferumoxytol had lower iron indices and received more blood transfusions compared to patients who received sodium ferric gluconate.


Asunto(s)
Anemia , Compuestos Férricos , Óxido Ferrosoférrico , Diálisis Renal/efectos adversos , Anciano , Anciano de 80 o más Años , Anemia/tratamiento farmacológico , Anemia/etiología , Transfusión Sanguínea/estadística & datos numéricos , Femenino , Compuestos Férricos/administración & dosificación , Compuestos Férricos/uso terapéutico , Óxido Ferrosoférrico/administración & dosificación , Óxido Ferrosoférrico/uso terapéutico , Hemoglobinas/análisis , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Resultado del Tratamiento
17.
Front Immunol ; 11: 571489, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33362760

RESUMEN

Superparamagnetic iron oxide nanoparticles (SPION) are employed as diagnostics and therapeutics following intravenous delivery for the treatment of iron deficiency anemia (IDA) in adult patients with chronic kidney failure. Neutrophils are the first defense against blood borne foreign insult and recruit to vascular sites of inflammation via a sequential process that is characterized by adhesive capture, rolling, and shear resistant arrest. A primary chemotactic agonist presented on the glycocalyx of inflamed endothelium is IL-8, which upon binding to its cognate membrane receptor (CXCR1/2) activates a suite of responses in neutrophils. An early response is degranulation with accompanying upregulation of ß2-integrin (CD11/CD18) and shedding of L-selectin (CD62L) receptors, which exert differential effects on the efficiency of endothelial recruitment. Feraheme is an FDA approved SPION treatment for IDA, but its effect on the innate immune response of neutrophils during inflammation has not been reported. Here, we studied the immunomodulatory effects of Feraheme on neutrophils freshly isolated from healthy human subjects and stimulated in suspension or on inflammatory mimetic substrates with IL-8. Cells treated with Feraheme exhibited reduced sensitivity to stimulation with IL-8, indicated by reduced upregulation of membrane CD11b/CD18 receptors, high affinity (HA) CD18, and shedding of CD62L. Feraheme also inhibited N-formyl-Met-Leu-Phe (fMLP) induced reactive oxygen species production. Neutrophil rolling, arrest, and migration was assessed in vascular mimetic microfluidic channels coated with E-selectin and ICAM-1 to simulate inflamed endothelium. Neutrophils exposed to Feraheme rolled faster on E-selectin and arrested less frequently on ICAM-1, in a manner dependent upon SPION concentration. Subsequent neutrophil shape change, and migration were also significantly inhibited in the presence of Feraheme. Lastly, Feraheme accelerated clearance of cytosolic calcium flux following IL-8 stimulation. We conclude that uptake of Feraheme by neutrophils inhibits chemotactic activation and downregulates normal rolling to arrest under shear flow. The mechanism involves increased calcium clearance following chemotactic activation, which may diminish the efficiency of recruitment from the circulation at vascular sites of inflammation.


Asunto(s)
Anemia Ferropénica/inmunología , Óxido Ferrosoférrico/uso terapéutico , Inflamación/terapia , Fallo Renal Crónico/inmunología , Nanopartículas Magnéticas de Óxido de Hierro/administración & dosificación , Neutrófilos/inmunología , Anemia Ferropénica/terapia , Antígenos CD18/metabolismo , Señalización del Calcio , Degranulación de la Célula , Células Cultivadas , Humanos , Interleucina-8/metabolismo , Fallo Renal Crónico/terapia , Selectina L/metabolismo , Activación Neutrófila , Receptores de Interleucina-8A/metabolismo
18.
Nat Commun ; 11(1): 4446, 2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32895387

RESUMEN

Owing to the poor penetration depth of light, phototherapy, including photothermal and photodynamic therapies, remains severely ineffective in treating deep tissue infections such as methicillin-resistant Staphylococcus aureus (MRSA)-infected osteomyelitis. Here, we report a microwave-excited antibacterial nanocapturer system for treating deep tissue infections that consists of microwave-responsive Fe3O4/CNT and the chemotherapy agent gentamicin (Gent). This system, Fe3O4/CNT/Gent, is proven to efficiently target and eradicate MRSA-infected rabbit tibia osteomyelitis. Its robust antibacterial effectiveness is attributed to the precise bacteria-capturing ability and magnetic targeting of the nanocapturer, as well as the subsequent synergistic effects of precise microwaveocaloric therapy from Fe3O4/CNT and chemotherapy from the effective release of antibiotics in infection sites. The advanced target-nanocapturer of microwave-excited microwaveocaloric-chemotherapy with effective targeting developed in this study makes a major step forward in microwave therapy for deep tissue infections.


Asunto(s)
Nanopartículas de Magnetita/uso terapéutico , Microondas/uso terapéutico , Osteomielitis/tratamiento farmacológico , Infecciones Estafilocócicas/tratamiento farmacológico , Animales , Antibacterianos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Quimioterapia/métodos , Óxido Ferrosoférrico/uso terapéutico , Gentamicinas/uso terapéutico , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Nanotubos de Carbono , Osteomielitis/microbiología , Conejos
19.
ACS Appl Mater Interfaces ; 12(17): 19285-19294, 2020 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-32249558

RESUMEN

Magnetite (Fe3O4) nanoparticles have been extensively used in noninvasive cancer treatment, for example, magnetic hyperthermia (MH) and chemodynamic therapy (CDT). However, how to achieve a highly efficient MH-CDT synergistic therapy based only on a single component of Fe3O4 still remains a challenge. Herein, hollow Fe3O4 mesocrystals (MCs) are constructed via a modified solvothermal method. Owing to the distinctive magnetic property of the mesocrystalline structure, Fe3O4 MCs show excellent magnetothermal conversion efficiency with a specific absorption rate of 722 w g-1 at a Fe concentration of 0.6 mg mL-1, much higher than that of Fe3O4 polycrystals (PCs). Moreover, Fe3O4 MCs also exhibit higher peroxidase-like activity than Fe3O4 PCs, which may be ascribed to the higher ratio of Fe2+/Fe3+ and more oxygen defects in the Fe3O4 MCs. Detailed in vivo results confirm that Fe3O4 MCs can instantly initiate CDT by producing the detrimental •OH, and such boosted reactive oxygen levels not only induces cell apoptosis but also reduces the expression of heat shock proteins, thus enabling low-temperature-mediated MH. More importantly, the in situ rising temperature resulted from MH in turn facilitates CDT, thus achieving a self-augmented synergistic effect between MH and CDT.


Asunto(s)
Antineoplásicos/uso terapéutico , Óxido Ferrosoférrico/uso terapéutico , Hipertermia Inducida/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Catálisis , Femenino , Óxido Ferrosoférrico/química , Radical Hidroxilo/metabolismo , Fenómenos Magnéticos , Ratones Endogámicos BALB C , Neoplasias/patología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Sci Rep ; 10(1): 1171, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31980695

RESUMEN

The use of magnetic fluid hyperthermia (MFH) for cancer therapy has shown promise but lacks suitable methods for quantifying exogenous irons such as superparamagnetic iron oxide (SPIO) nanoparticles as a source of heat generation under an alternating magnetic field (AMF). Application of quantitative susceptibility mapping (QSM) technique to prediction of SPIO in preclinical models has been challenging due to a large variation of susceptibility values, chemical shift from tissue fat, and noisier data arising from the higher resolution required to visualize the anatomy of small animals. In this study, we developed a robust QSM for the SPIO ferumoxytol in live mice to examine its potential application in MFH for cancer therapy. We demonstrated that QSM was able to simultaneously detect high level ferumoxytol accumulation in the liver and low level localization near the periphery of tumors. Detection of ferumoxytol distribution in the body by QSM, however, required imaging prior to and post ferumoxytol injection to discriminate exogenous iron susceptibility from other endogenous sources. Intratumoral injection of ferumoxytol combined with AMF produced a ferumoxytol-dose dependent tumor killing. Histology of tumor sections corroborated QSM visualization of ferumoxytol distribution near the tumor periphery, and confirmed the spatial correlation of cell death with ferumoxytol distribution. Due to the dissipation of SPIOs from the injection site, quantitative mapping of SPIO distribution will aid in estimating a change in temperature in tissues, thereby maximizing MFH effects on tumors and minimizing side-effects by avoiding unwanted tissue heating.


Asunto(s)
Compuestos Férricos/análisis , Óxido Ferrosoférrico/análisis , Hipertermia Inducida , Nanopartículas/análisis , Adenocarcinoma/diagnóstico por imagen , Adenocarcinoma/patología , Adenocarcinoma/terapia , Animales , Línea Celular Tumoral , Medios de Contraste , Compuestos Férricos/farmacocinética , Compuestos Férricos/uso terapéutico , Óxido Ferrosoférrico/farmacocinética , Óxido Ferrosoférrico/uso terapéutico , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos NOD , Nanopartículas/uso terapéutico , Tomografía Computarizada por Tomografía de Emisión de Positrones , Neoplasias de la Próstata/patología , Radioisótopos , Radiofármacos , Tejido Subcutáneo , Distribución Tisular , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Circonio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...