Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Angew Chem Int Ed Engl ; : e202410919, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38995663

RESUMEN

Despite numerous screening tools for colorectal cancer (CRC), 25% of patients are diagnosed with advanced disease.  Novel diagnostic technologies that are early, accurate, and rapid are imperative to assess the therapeutic efficacy of clinical drugs and identify new biomarkers of treatment response. Here Raman spectroscopy (RS) was used to track metabolic reprogramming in KRAS-mutant HCT116 and SW837 cells, and KRAS wild-type CC cells. RS combined with multivariate analysis methods distinguished nonresponsive, partially responsive, and responsive cells treated with cetuximab, a monoclonal antibody for EGFR inhibition, sotorasib, a clinically approved KRAS inhibitor, and various doses of trametinib, an inhibitor of the MAPK pathway. Cells treated with a combination of subtoxic doses of trametinib and BKM120, an inhibitor of the PI3K pathway, showed a synergistic response between the two pathways. Using a supervised machine learning regression model, we established a scoring methodology trained to a priori predict therapeutic response to new treatment combinations. RS metabolites were verified with mass spectrometry, and enrichment pathways were identified, including amino acid, purine, and nicotinate and nicotinamide metabolism that differentiated monotherapy from combination therapy. Our approach may ultimately be applicable to patient-derived primary cells and cultures of patient tumors to predict effective drugs for individualized care.

2.
Biomater Sci ; 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39041781

RESUMEN

Reactive oxygen species (ROS) drive processes in various pathological conditions serving as an attractive target for therapeutic strategies. This review highlights the development and use of ROS-dependent prodrug-based nanoscale carriers that has transformed many biomedical applications. Incorporating prodrugs into nanoscale carriers not only improves their stability and solubility but also enables site-specific drug delivery ultimately enhancing the therapeutic effectiveness of the nanoscale carriers. We critically examine recent advances in ROS-responsive nanoparticulate platforms, encompassing liposomes, polymeric nanoparticles, and inorganic nanocarriers. These platforms facilitate precise control over drug release upon encountering elevated ROS levels at disease sites, thereby minimizing off-target effects and maximizing therapeutic efficiency. Furthermore, we investigate the potential of combination therapies in which ROS-activated prodrugs are combined with other therapeutic agents and underscore their synergistic potential for treating multifaceted diseases. This comprehensive review highlights the immense potential of ROS-dependent prodrug-based nanoparticulate systems in revolutionizing biomedical applications; such nanoparticulate systems can facilitate selective and controlled drug delivery, reduce toxicity, and improve therapeutic outcomes for ROS-associated diseases.

3.
ACS Nano ; 18(19): 12117-12133, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38648373

RESUMEN

Ulcerative colitis is a chronic condition in which a dysregulated immune response contributes to the acute intestinal inflammation of the colon. Current clinical therapies often exhibit limited efficacy and undesirable side effects. Here, programmable nanomicelles were designed for colitis treatment and loaded with RU.521, an inhibitor of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. STING-inhibiting micelles (SIMs) comprise hyaluronic acid-stearic acid conjugates and include a reactive oxygen species (ROS)-responsive thioketal linker. SIMs were designed to selectively accumulate at the site of inflammation and trigger drug release in the presence of ROS. Our in vitro studies in macrophages and in vivo studies in a murine model of colitis demonstrated that SIMs leverage HA-CD44 binding to target sites of inflammation. Oral delivery of SIMs to mice in both preventive and delayed therapeutic models ameliorated colitis's severity by reducing STING expression, suppressing the secretion of proinflammatory cytokines, enabling bodyweight recovery, protecting mice from colon shortening, and restoring colonic epithelium. In vivo end points combined with metabolomics identified key metabolites with a therapeutic role in reducing intestinal and mucosal inflammation. Our findings highlight the significance of programmable delivery platforms that downregulate inflammatory pathways at the intestinal mucosa for managing inflammatory bowel diseases.


Asunto(s)
Colitis Ulcerosa , Proteínas de la Membrana , Micelas , Nucleotidiltransferasas , Animales , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/patología , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/inducido químicamente , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Humanos , Ratones Endogámicos C57BL , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo
4.
Bioeng Transl Med ; 9(1): e10595, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38193120

RESUMEN

Preeclampsia is a life-threatening pregnancy disorder. Current clinical assays cannot predict the onset of preeclampsia until the late 2nd trimester, which often leads to poor maternal and neonatal outcomes. Here we show that Raman spectroscopy combined with machine learning in pregnant patient plasma enables rapid, highly sensitive maternal metabolome screening that predicts preeclampsia as early as the 1st trimester with >82% accuracy. We identified 12, 15 and 17 statistically significant metabolites in the 1st, 2nd and 3rd trimesters, respectively. Metabolic pathway analysis shows multiple pathways corresponding to amino acids, fatty acids, retinol, and sugars are enriched in the preeclamptic cohort relative to a healthy pregnancy. Leveraging Pearson's correlation analysis, we show for the first time with Raman Spectroscopy that metabolites are associated with several clinical factors, including patients' body mass index, gestational age at delivery, history of preeclampsia, and severity of preeclampsia. We also show that protein quantification alone of proinflammatory cytokines and clinically relevant angiogenic markers are inadequate in identifying at-risk patients. Our findings demonstrate that Raman spectroscopy is a powerful tool that may complement current clinical assays in early diagnosis and in the prognosis of the severity of preeclampsia to ultimately enable comprehensive prenatal care for all patients.

5.
Biomater Sci ; 11(21): 7188-7202, 2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37750339

RESUMEN

Magnetic hyperthermia has attracted considerable attention for efficient cancer therapy because of its noninvasive nature, deep tissue penetration, and minimal damage to healthy tissues. Herein, we have fused cancer cell membrane fragments with lipids and cloaked them on magnetic nanorings to form targeted Fe nanorings (TF) for tumor-targeted magnetic hyperthermia-induced tumor ablation. In our approach, cell membrane fragments from cancer cells were fused with lipids to form vesicles, which could efficiently encapsulate magnetic nanorings, thereby forming TF. We observed that TF have high tumor uptake via homotypic targeting, where cancer cells take up TF through membrane fusion. Under an external alternating magnetic field (AMF), TF accumulated in the tumors are heated, driving magnetic-hyperthermia-induced tumor cell death. Our in vitro studies show that self-targeting TF efficiently localized in cancer cells and induced cell death with an AMF, which was shown by a live/dead assay. Our findings demonstrate the potential of TF in tumor ablation, thereby making them promising and efficient nanosystems for tumor-targeted theranostics.


Asunto(s)
Hipertermia Inducida , Nanopartículas de Magnetita , Línea Celular Tumoral , Membrana Celular , Fenómenos Magnéticos , Lípidos , Campos Magnéticos
6.
ACS Appl Mater Interfaces ; 15(32): 38185-38200, 2023 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-37549133

RESUMEN

Preterm birth (PTB) is the leading cause of infant deaths globally. Current clinical measures often fail to identify women who may deliver preterm. Therefore, accurate screening tools are imperative for early prediction of PTB. Here, we show that Raman spectroscopy is a promising tool for studying biological interfaces, and we examine differences in the maternal metabolome of the first trimester plasma of PTB patients and those that delivered at term (healthy). We identified fifteen statistically significant metabolites that are predictive of the onset of PTB. Mass spectrometry metabolomics validates the Raman findings identifying key metabolic pathways that are enriched in PTB. We also show that patient clinical information alone and protein quantification of standard inflammatory cytokines both fail to identify PTB patients. We show for the first time that synergistic integration of Raman and clinical data guided with machine learning results in an unprecedented 85.1% accuracy of risk stratification of PTB in the first trimester that is currently not possible clinically. Correlations between metabolites and clinical features highlight the body mass index and maternal age as contributors of metabolic rewiring. Our findings show that Raman spectral screening may complement current prenatal care for early prediction of PTB, and our approach can be translated to other patient-specific biological interfaces.


Asunto(s)
Nacimiento Prematuro , Embarazo , Humanos , Femenino , Recién Nacido , Nacimiento Prematuro/diagnóstico , Nacimiento Prematuro/prevención & control , Primer Trimestre del Embarazo , Espectrometría Raman , Metabolómica
7.
Anal Chem ; 95(35): 13172-13184, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37605298

RESUMEN

Resistance to clinical therapies remains a major barrier in cancer management. There is a critical need for rapid and highly sensitive diagnostic tools that enable early prediction of treatment response to allow accurate clinical decisions. Here, Raman spectroscopy was employed to monitor changes in key metabolites as early predictors of response in KRAS-mutant colorectal cancer (CRC) cells, HCT116, treated with chemotherapies. We show at the single cell level that HCT116 is resistant to cetuximab (CTX), the first-line treatment in CRC, but this resistance can be overcome with pre-sensitization of cells with oxaliplatin (OX). In combination treatment of CTX + OX, sequential delivery of OX followed by CTX rather than simultaneous administration of drugs was observed to be critical for effective therapy. Our results demonstrated that metabolic changes are well aligned to cellular mechanical changes where Young's modulus decreased after effective treatment, indicating that both changes in mechanical properties and metabolism in cells are likely responsible for cancer proliferation. Raman findings were verified with mass spectrometry (MS) metabolomics, and both platforms showed changes in lipids, nucleic acids, and amino acids as predictors of resistance/response. Finally, key metabolic pathways enriched were identified when cells are resistant to CTX but downregulated with effective treatment. This study highlights that drug-induced metabolic changes both at the single cell level (Raman) and ensemble level (MS) have the potential to identify mechanisms of response to clinical cancer therapies.


Asunto(s)
Antifibrinolíticos , Neoplasias , Humanos , Espectrometría Raman , Metabolómica , Aminoácidos , Cetuximab/farmacología , Oxaliplatino/farmacología
8.
Chem Rev ; 123(13): 8297-8346, 2023 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-37318957

RESUMEN

Omics technologies have rapidly evolved with the unprecedented potential to shape precision medicine. Novel omics approaches are imperative toallow rapid and accurate data collection and integration with clinical information and enable a new era of healthcare. In this comprehensive review, we highlight the utility of Raman spectroscopy (RS) as an emerging omics technology for clinically relevant applications using clinically significant samples and models. We discuss the use of RS both as a label-free approach for probing the intrinsic metabolites of biological materials, and as a labeled approach where signal from Raman reporters conjugated to nanoparticles (NPs) serve as an indirect measure for tracking protein biomarkers in vivo and for high throughout proteomics. We summarize the use of machine learning algorithms for processing RS data to allow accurate detection and evaluation of treatment response specifically focusing on cancer, cardiac, gastrointestinal, and neurodegenerative diseases. We also highlight the integration of RS with established omics approaches for holistic diagnostic information. Further, we elaborate on metal-free NPs that leverage the biological Raman-silent region overcoming the challenges of traditional metal NPs. We conclude the review with an outlook on future directions that will ultimately allow the adaptation of RS as a clinical approach and revolutionize precision medicine.


Asunto(s)
Medicina de Precisión , Espectrometría Raman , Medicina de Precisión/métodos , Proteómica/métodos , Metabolómica/métodos , Biomarcadores/metabolismo
9.
Investig Clin Urol ; 64(2): 182-188, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36882178

RESUMEN

PURPOSE: This study aimed to develop hyaluronic acid (HA)-based, retinoic acid (RA)-containing nanomicelles and to investigate the effects of these newly developed nanomicelles on regeneration of the vaginal epithelium and aquaporin 3 (AQP3) expression in a murine menopause model. MATERIALS AND METHODS: The HA-based, RA-loaded nanomicelles were developed, and the RA-loading rate, encapsulation efficiency, and hydrodynamic diameter were measured. Female BALB/c mice (8 weeks; n=30) were divided into control and experimental groups. Menopause was established in the experimental group by removing both ovaries. The experimental group was further divided into an ovariectomy group, an HA-C18 vehicle group, and an HA-C18-RA group (2.5 µg per mouse); vaginal administration of HA-C18 or HA-C18-RA was performed once daily. After 4 weeks of treatment, murine vaginal tissue was removed, and histological analysis was performed. RESULTS: Three drug-loaded nanomicelles were synthesized: the RA content in HA-C18-RA-10, HA-C18-RA-20, and HA-C18-RA-30 was 3.13%, 2.52%, and 16.67%, respectively, and the RA encapsulation efficiency was 95.57%, 83.92%, and 93.24%, respectively. In the experimental versus control group, serum estrogen levels were significantly reduced, and the vaginal mucosal epithelial layer was significantly thinner. After 4 weeks of treatment, the thickness of the vaginal mucosal epithelial layer and AQP3 expression was increased in the HA-C18-RA group compared with the HA-C18 vehicle group. CONCLUSIONS: The newly developed HA-based nanomicelles containing RA resulted in vaginal epithelial recovery and increased AQP3 expression. The results may contribute to the development of functional vaginal lubricants or moisturizers for the treatment of vaginal dryness.


Asunto(s)
Ácido Hialurónico , Retinoides , Femenino , Animales , Ratones , Ácido Hialurónico/farmacología , Tretinoina , Epitelio , Menopausia , Ratones Endogámicos BALB C
10.
Small ; 19(29): e2204293, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36965074

RESUMEN

The in vivo dynamics of nanoparticles requires a mechanistic understanding of multiple factors. Here, for the first time, the surprising breakdown of functionalized gold nanostars (F-AuNSs) conjugated with antibodies and 64 Cu radiolabels in vivo and in artificial lysosomal fluid ex vivo, is shown. The short-term biodistribution of F-AuNSs is driven by the route of systemic delivery (intravenous vs intraperitoneal) and long-term fate is controlled by the tissue type in vivo. In vitro studies including endocytosis pathways, intracellular trafficking, and opsonization, are combined with in vivo studies integrating a milieu of spectroscopy and microcopy techniques that show F-AuNSs dynamics is driven by their physicochemical properties and route of delivery. F-AuNSs break down into sub-20 nm broken nanoparticles as early as 7 days postinjection. Martini coarse-grained simulations are performed to support the in vivo findings. Simulations suggest that shape, size, and charge of the broken nanoparticles, and composition of the lipid membrane depicting various tissues govern the interaction of the nanoparticles with the membrane, and the rate of translocation across the membrane to ultimately enable tissue clearance. The fundamental study addresses critical gaps in the knowledge regarding the fate of nanoparticles in vivo that remain a bottleneck in their clinical translation.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Oro/química , Distribución Tisular , Nanopartículas/química , Nanopartículas del Metal/química
11.
Biomater Sci ; 11(1): 298-306, 2022 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-36448579

RESUMEN

In an immunosuppressive tumor microenvironment, tumor-associated macrophages (TAMs) are the most abundant cells displaying pro-tumorigenic M2-like phenotypes, encouraging tumor growth and influencing the development of resistance against conventional therapies. TAMs are highly malleable. They can be repolarized into tumoricidal M1-like cells. In this study, we report the synthesis of novel co-operative immuno-photodynamic nanoparticles involving TAM self-targeting acrylic acid grafted mannan (a polysaccharide) conjugated with the chlorin e6 (Ce6) photosensitizer and then loaded with resiquimod (R848), a toll-like receptor (TLR7/8) agonist. The mannan conjugated Ce6 loaded with R848 (MCR) as bioconjugate nanoparticles demonstrated selective targeting of anti-inflammatory M2-like cells. Using photodynamic therapy they were repolarized to pro-inflammatory M1-like cells with combined effects of reactive oxygen species (ROS)-triggered intracellular signaling and a small-molecule immunostimulant. The MCR also demonstrated a TAM-directed adaptive immune response, inhibited tumor growth, and prevented metastasis. Our results indicate that these MCR nanoparticles can effectively target TAMs and modulate them for cancer immunotherapy.


Asunto(s)
Nanopartículas , Neoplasias , Fotoquimioterapia , Humanos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Mananos , Macrófagos Asociados a Tumores , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/uso terapéutico , Fotoquimioterapia/métodos , Neoplasias/tratamiento farmacológico , Línea Celular Tumoral , Microambiente Tumoral
12.
Artículo en Inglés | MEDLINE | ID: mdl-36006784

RESUMEN

Natural killer (NK) cells are an important component of the tumor immunosurveillance; activated NK cells can recognize and directly lyse tumor cells eliciting a potent antitumor immune response. Due to their intrinsic ability to unleash cytotoxicity against tumor cells, NK cell-based adoptive cell therapies have gained rapid clinical significance, and many clinical trials are ongoing. However, priming and activating NK cells, infiltration of activated NK cells in the immunosuppressive tumor microenvironment, and tracking the infiltrated NK cells in the tumors remain a critical challenge. To address these challenges, NK cells have been successfully interfaced with nanomaterials where the morphology, composition, and surface characteristics of nanoparticles (NPs) were leveraged to enable longitudinal tracking of NK cells in tumors or deliver therapeutics to prime NK cells. Distinct from other published reviews, in this tutorial review, we summarize the recent findings in the past decade where NPs were used to label NK cells for immunoimaging or deliver treatment to activate NK cells and induce long-term immunity against tumors. We discuss the NP properties that are key to surmounting the current challenges in NK cells and the different strategies employed to advance NK cells-based diagnostics and therapeutics. We conclude the review with an outlook on future directions in NP-NK cell hybrid interfaces, and overall clinical impact and patient response to such interfaces that need to be addressed to enable their clinical translation.

13.
Vaccines (Basel) ; 10(5)2022 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-35632493

RESUMEN

In December 2019, an unknown viral infection emerged and quickly spread worldwide, resulting in a global pandemic. This novel virus caused severe pneumonia and acute respiratory distress syndrome caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). It has caused 6.25 millions of deaths worldwide and remains a major concern for health, society, and the economy. As vaccination is one of the most efficient ways to combat this pandemic, different vaccines were developed in a short period. This review article discusses how coronavirus affected the top nations of the world and the vaccines being used for the prevention. Amongst the vaccines, some vaccines have already been approved, and some have been involved in clinical studies. The article also provides insight into different COVID-19 vaccine platforms, their preparation, working, efficacy, and side effects.

14.
J Nanobiotechnology ; 20(1): 205, 2022 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-35477452

RESUMEN

BACKGROUND: The reactive oxygen species (ROS) and inflammation, a critical contributor to tissue damage, is well-known to be associated with various disease. The kidney is susceptible to hypoxia and vulnerable to ROS. Thus, the vicious cycle between oxidative stress and renal hypoxia critically contributes to the progression of chronic kidney disease and finally, end-stage renal disease. Thus, delivering therapeutic agents to the ROS-rich inflammation site and releasing the therapeutic agents is a feasible solution. RESULTS: We developed a longer-circulating, inflammation-sensing, ROS-scavenging versatile nanoplatform by stably loading catalase-mimicking 1-dodecanethiol stabilized Mn3O4 (dMn3O4) nanoparticles inside ROS-sensitive nanomicelles (PTC), resulting in an ROS-sensitive nanozyme (PTC-M). Hydrophobic dMn3O4 nanoparticles were loaded inside PTC micelles to prevent premature release during circulation and act as a therapeutic agent by ROS-responsive release of loaded dMn3O4 once it reached the inflammation site. CONCLUSIONS: The findings of our study demonstrated the successful attenuation of inflammation and apoptosis in the IRI mice kidneys, suggesting that PTC-M nanozyme could possess promising potential in AKI therapy. This study paves the way for high-performance ROS depletion in treating various inflammation-related diseases.


Asunto(s)
Lesión Renal Aguda , Lesión Renal Aguda/tratamiento farmacológico , Animales , Catalasa , Femenino , Humanos , Hipoxia , Inflamación/tratamiento farmacológico , Masculino , Ratones , Estrés Oxidativo , Especies Reactivas de Oxígeno
16.
J Korean Med Sci ; 36(49): e333, 2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34931497

RESUMEN

BACKGROUND: Chronic kidney disease (CKD) is a global health problem, and there is no permanent treatment for reversing kidney failure; thus, early diagnosis and effective treatment are required. Gene therapy has outstanding potential; however, the lack of safe gene delivery vectors, a reasonable transfection rate, and kidney targeting ability limit its application. Nanoparticles can offer innovative ways to diagnose and treat kidney diseases as they facilitate targetability and therapeutic efficacy. METHODS: Herein, we developed a proximal renal tubule-targeting gene delivery system based on alternative copolymer (PS) of sorbitol and polyethyleneimine (PEI), modified with vimentin-specific chitobionic acid (CA), producing PS-conjugated CA (PSC) for targeting toward vimentin-expressing cells in the kidneys. In vitro studies were used to determine cell viability, transfection efficiency, serum influence, and specific uptake in the human proximal renal tubular epithelial cell line (HK-2). Finally, the targeting efficiency of the prepared PSC gene carriers was checked in a murine model of Alport syndrome. RESULTS: Our results suggested that the prepared polyplex showed low cytotoxicity, enhanced transfection efficiency, specific uptake toward HK-2 cells, and excellent targeting efficiency toward the kidneys. CONCLUSION: Collectively, from these results it can be inferred that the PSC can be further evaluated as a potential gene carrier for the kidney-targeted delivery of therapeutic genes for treating diseases.


Asunto(s)
Nanopartículas/química , Plásmidos/genética , Vimentina/genética , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Disacáridos/química , Colorantes Fluorescentes/química , Humanos , Riñón/metabolismo , Riñón/patología , Ratones , Nanopartículas/toxicidad , Plásmidos/química , Plásmidos/metabolismo , Polietileneimina/química , Polímeros/química , Azúcares Ácidos/química , Transfección/métodos , Vimentina/metabolismo
17.
Adv Healthc Mater ; 10(21): e2100907, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34541833

RESUMEN

Triple-negative breast cancer (TNBC) features immunologically "cold" tumor microenvironments with limited cytotoxic T lymphocyte (CTL) infiltration. Although ablation therapies have demonstrated modulation of "cold" TNBC tumors to inflamed "hot" tumors, recruitment of myeloid derived suppressor cells (MDSCs) at the tumors post ablation therapies prevents the infiltration of CTLs and challenge the antitumor potentials of T-cell therapies. Here, a thermal ablation immunotherapy strategy is developed to prevent the immune suppressive effects of MDSCs during photothermal ablation and induce a durable systemic antitumor immunity to eradicate TNBC tumors. An injectable pluronic F127/hyaluronic acid (HA)-based hydrogel embedded with manganese dioxide (BM) nanoparticles and TLR7 agonist resiquimod (R848) (BAGEL-R848), is synthesized to induce in situ laser-assisted gelation of the hydrogel and achieve desired ablation temperatures at a low laser-exposure time. Upon 808-nm laser irradiation, a significant reduction in the tumor burden is observed in BAGEL-R848-injected 4T1 tumor-bearing mice. The ablation induced immunogenic cell death and sustained release of R848 from BAGEL-R848 promotes dendritic cell maturation and reduced MDSCs localization in tumors. In addition, inflammatory M1 macrophages and CD8+IFN+ CTL are enriched in distant tumors in bilateral 4T1 tumor model, preventing metastatic tumor growth and signifying the potential of BAGEL-R848 to treat TNBC.


Asunto(s)
Nanopartículas , Neoplasias de la Mama Triple Negativas , Animales , Calor , Humanos , Inmunidad , Inmunoterapia , Ratones , Neoplasias de la Mama Triple Negativas/terapia , Microambiente Tumoral
18.
J Nanobiotechnology ; 19(1): 109, 2021 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-33865397

RESUMEN

BACKGROUND: Recently, we developed hydrophobically modified glycol chitosan (HGC) nanomicelles loaded with tacrolimus (TAC) (HGC-TAC) for the targeted renal delivery of TAC. Herein, we determined whether the administration of the HGC-TAC nanomicelles decreases kidney injury in a model of lupus nephritis. Lupus-prone female MRL/lpr mice were randomly assigned into three groups that received intravenous administration of either vehicle control, an equivalent dose of TAC, or HGC-TAC (0.5 mg/kg TAC) weekly for 8 weeks. Age-matched MRL/MpJ mice without Faslpr mutation were also treated with HGC vehicle and used as healthy controls. RESULTS: Weekly intravenous treatment with HGC-TAC significantly reduced genetically attributable lupus activity in lupus nephritis-positive mice. In addition, HGC-TAC treatment mitigated renal dysfunction, proteinuria, and histological injury, including glomerular proliferative lesions and tubulointerstitial infiltration. Furthermore, HGC-TAC treatment reduced renal inflammation and inflammatory gene expression and ameliorated increased apoptosis and glomerular fibrosis. Moreover, HGC-TAC administration regulated renal injury via the TGF-ß1/MAPK/NF-κB signaling pathway. These renoprotective effects of HGC-TAC treatment were more potent in lupus mice compared to those of TAC treatment alone. CONCLUSION: Our study indicates that weekly treatment with the HGC-TAC nanomicelles reduces kidney injury resulting from lupus nephritis by preventing inflammation, fibrosis, and apoptosis. This advantage of a new therapeutic modality using kidney-targeted HGC-TAC nanocarriers may improve drug adherence and provide treatment efficacy in lupus nephritis mice.


Asunto(s)
Quitosano/farmacología , Quitosano/uso terapéutico , Nefritis Lúpica/tratamiento farmacológico , Micelas , Tacrolimus/farmacología , Tacrolimus/uso terapéutico , Animales , Apoptosis , Quitosano/química , Femenino , Fibrosis/patología , Expresión Génica , Interacciones Hidrofóbicas e Hidrofílicas , Inflamación , Riñón/lesiones , Riñón/patología , Nefritis Lúpica/patología , Ratones , Ratones Endogámicos MRL lpr , FN-kappa B/metabolismo , Transducción de Señal
20.
ACS Appl Mater Interfaces ; 13(4): 4844-4852, 2021 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-33486952

RESUMEN

The combination of photothermal therapy (PTT) and toll-like receptor (TLR)-mediated immunotherapy can elicit antitumor immunity and modulate the immunosuppressive tumor microenvironment (TME). Unlike other TLRs, TLR-5 is a promising target for immune activation, as its expression is well-maintained even during immunosenescence. Here, we developed a unique tumor microenvironment-regulating immunosenescence-independent nanostimulant consisting of TLR-5 adjuvant Vibrio vulnificus flagellin B (FlaB) conjugated onto the surface to an IR 780-loaded hyaluronic acid-stearylamine (HIF) micelles. These HIF micelles induced immune-mediated cell death via PTT when irradiated with a near-infrared laser. In comparison with PTT alone, the combination of in situ-generated tumor-associated antigens produced during PTT and the immune adjuvant FlaB demonstrated enhanced vaccine-like properties and modulated the TME by suppressing immune-suppressive regulatory cells (Tregs) and increasing the fraction of CD103+ migratory dendritic cells, which are responsible for trafficking tumor antigens to draining lymph nodes (DLNs). This combinatorial strategy (i.e., applying a TLR-5 adjuvant targeted to immunosenescence-independent TLR-5 and the in situ photothermal generation of tumor-associated antigens) is a robust system for next-generation immunotherapy and could even be applied in elderly patients, thus broadening the clinical scope of immunotherapy strategies.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Flagelina/uso terapéutico , Inmunoterapia , Nanopartículas/uso terapéutico , Neoplasias/terapia , Terapia Fototérmica , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Animales , Línea Celular Tumoral , Femenino , Flagelina/administración & dosificación , Flagelina/inmunología , Células HEK293 , Humanos , Inmunosenescencia/efectos de los fármacos , Inmunosenescencia/efectos de la radiación , Inmunoterapia/métodos , Rayos Infrarrojos/uso terapéutico , Ratones Endogámicos C57BL , Nanopartículas/administración & dosificación , Neoplasias/inmunología , Neoplasias/patología , Terapia Fototérmica/métodos , Receptor Toll-Like 5/antagonistas & inhibidores , Receptor Toll-Like 5/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de la radiación , Vibrio vulnificus/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...