Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Int J Pharm ; 642: 123139, 2023 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-37311499

RESUMEN

Recent work has highlighted that amorphous solid dispersions (ASDs) containing delamanid (DLM) and an enteric polymer, hypromellose phthalate (HPMCP), appear to be susceptible to crystallization during immersion in simulated gastric fluids. The goal of this study was to minimize contact of the ASD particles with the acidic media via application of an enteric coating to tablets containing the ASD intermediate, and improve the subsequent drug release at higher pH conditions. DLM ASDs were prepared with HPMCP and formulated into a tablet that was then coated with a methacrylic acid copolymer. Drug release was studied in vitro using a two-stage dissolution test where the pH of the gastric compartment was altered to reflect physiological variations. The medium was subsequently switched to simulated intestinal fluid. The gastric resistance time of the enteric coating was probed over the pH range of 1.6-5.0. The enteric coating was found to be effective at protecting the drug against crystallization in pH conditions where HPMCP was insoluble. Consequently, the variability in drug release following gastric immersion under pH conditions reflecting different prandial states was notably reduced when compared to the reference product. These findings support closer examination of the potential for drug crystallization from ASDs in the gastric environment where acid-insoluble polymers may be less effective as crystallization inhibitors. Further, addition of a protective enteric coating appears to provide a promising remediation strategy to prevent crystallization at low pH environments, and may mitigate variability associated with prandial state that arises due to pH changes.


Asunto(s)
Polímeros , Polímeros/química , Solubilidad , Liberación de Fármacos , Comprimidos , Cristalización
2.
Mol Pharm ; 20(6): 3170-3186, 2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-37220082

RESUMEN

Weakly acid polymers with pH-responsive solubility are being used with increasing frequency in amorphous solid dispersion (ASD) formulations of drugs with low aqueous solubility. However, drug release and crystallization in a pH environment where the polymer is insoluble are not well understood. The aim of the current study was to develop ASD formulations optimized for release and supersaturation longevity of a rapidly crystallizing drug, pretomanid (PTM), and to evaluate a subset of these formulations in vivo. Following screening of several polymers for their ability to inhibit crystallization, hypromellose acetate succinate HF grade (HPMCAS-HF; HF) was selected to prepare PTM ASDs. In vitro release studies were conducted in simulated fasted- and fed-state media. Drug crystallization in ASDs following exposure to dissolution media was evaluated by powder X-ray diffraction, scanning electron microscopy, and polarized light microscopy. In vivo oral pharmacokinetic evaluation was conducted in male cynomolgus monkeys (n = 4) given 30 mg PTM under both fasted and fed conditions in a crossover design. Three HPMCAS-based ASDs of PTM were selected for fasted-state animal studies based on their in vitro release performance. Enhanced bioavailability was observed for each of these formulations relative to the reference product that contained crystalline drug. The 20% drug loading PTM-HF ASD gave the best performance in the fasted state, with subsequent dosing in the fed state. Interestingly, while food improved drug absorption of the crystalline reference product, the exposure of the ASD formulation was negatively impacted. The failure of the HPMCAS-HF ASD to enhance absorption in the fed state was hypothesized to result from poor release in the reduced pH intestinal environment resulting from the fed state. In vitro experiments confirmed a reduced release rate under lower pH conditions, which was attributed to reduced polymer solubility and an enhanced crystallization tendency of the drug. These findings emphasize the limitations of in vitro assessment of ASD performance using standardized media conditions. Future studies are needed for improved understanding of food effects on ASD release and how this variability can be captured by in vitro testing methodologies for better prediction of in vivo outcomes, in particular for ASDs formulated with enteric polymers.


Asunto(s)
Polímeros , Animales , Masculino , Polímeros/química , Solubilidad , Cristalización , Liberación de Fármacos
3.
Mol Pharm ; 20(3): 1681-1695, 2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36730186

RESUMEN

Enteric polymers are widely used in amorphous solid dispersion (ASD) formulations. The aim of the current study was to explore ASD failure mechanisms across a wide range of pH conditions that mimic in vivo gastric compartment variations where enteric polymers such as hydroxypropyl methylcellulose phthalate (HPMCP) and hydroxypropyl methylcellulose acetate succinate (HPMCAS) are largely insoluble. Delamanid (DLM), a weakly basic drug used to treat tuberculosis, was selected as the model compound. Both DLM free base and the edisylate salt were formulated with HPMCP, while DLM edisylate ASDs were also prepared with different grades of HPMCAS. Two-stage release testing was conducted with the gastric stage pH varied between pH 1.6 and 5.0, prior to transfer to intestinal conditions of pH 6.5. ASD particles were collected following suspension in the gastric compartment and evaluated using X-ray powder diffraction and scanning electron microscopy. Additional samples were also evaluated with polarized light microscopy. In general, ASDs with HPMCP showed improved overall release for all testing conditions, relative to ASDs with HPMCAS. ASDs with the edisylate salt likewise outperformed those with DLM free base. Impaired release for certain formulations at intestinal pH conditions was attributed to surface drug crystallization that initiated during suspension in the gastric compartment where the polymer is insoluble; crystallization appeared more extensive for HPMCAS ASDs. These findings suggest that gastric pH variations should be evaluated for ASD formulations containing weakly basic drugs and enteric polymers.


Asunto(s)
Polímeros , Polímeros/química , Solubilidad , Composición de Medicamentos , Cristalización , Concentración de Iones de Hidrógeno
4.
ACS Appl Mater Interfaces ; 14(36): 40698-40710, 2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36054111

RESUMEN

Preventing crystallization is a primary concern when developing amorphous drug formulations. Recently, atomic layer coatings (ALCs) of aluminum oxide demonstrated crystallization inhibition of high drug loading amorphous solid dispersions (ASDs) for over 2 years. The goal of the current study was to probe the breadth and mechanisms of this exciting finding through multiple drug/polymer model systems, as well as particle and coating attributes. The model ASD systems selected provide for a range of hygroscopicity and chemical functional groups, which may contribute to the crystallization inhibition effect of the ALC coatings. Atomic layer coating was performed to apply a 5-25 nm layer of aluminum oxide or zinc oxide onto ASD particles, which imparted enhanced micromeritic properties, namely, reduced agglomeration and improved powder flowability. ASD particles were stored at 40 °C and a selected relative humidity level between 31 and 75%. Crystallization was monitored by X-ray powder diffraction and scanning electron microscopy (SEM) up to 48 weeks. Crystallization was observable by SEM within 1-2 weeks for all uncoated samples. After ALC, crystallization was effectively delayed or completely inhibited in some systems up to 48 weeks. The delay achieved was demonstrated regardless of polymer hygroscopicity, presence or absence of hydroxyl functional groups in drugs and/or polymers, particle size, or coating properties. The crystallization inhibition effect is attributed primarily to decreased surface molecular mobility. ALC has the potential to be a scalable strategy to enhance the physical stability of ASD systems to enable high drug loading and enhanced robustness to temperature or relative humidity excursions.


Asunto(s)
Óxido de Aluminio , Polímeros , Cristalización , Estabilidad de Medicamentos , Polímeros/química , Polvos/química , Solubilidad
5.
Eur J Pharm Biopharm ; 174: 131-143, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35413402

RESUMEN

Poor solubility is a major challenge that can limit the oral bioavailability of many drugs, including delamanid, a weakly basic nitro-dihydro-imidazooxazole derivative used to treat tuberculosis. Amorphous solid dispersion (ASD) can improve the bioavailability of poorly water-soluble compounds, yet drug crystallization is a potential failure mechanism, particularly as the drug loading increases. The goal of the current study was two-fold: to enhance the stability of amorphous delamanid against crystallization and to improve drug release by developing ASDs containing the salt form of the drug. Various sulfonate salts of delamanid were prepared in amorphous form and evaluated for their tendency to crystallize and undergo chemical degradation following storage at 40 °C/75% relative humidity. Drug release was evaluated by a two-stage dissolution test consisting of an initial low pH stage, followed by transfer to a higher pH medium. For ASDs of the free base, small amounts of crystallinity during preparation were found to limit the drug release. Delamanid salts with sulfonic acids showed considerably improved amorphous stability. Tosylate, besylate, edisylate, and mesylate salts had high glass transition temperatures as well as good chemical and physical stability. In addition, a remarkable improvement in the drug release was observed when ASDs were prepared with these salts in comparison to the free base form. Specifically, ASDs with hydroxypropyl methylcellulose phthalate (HPMCP) at 25% drug loading exhibited near-complete drug release for all four sulfonate salts. These findings suggest that the dual strategy combining salt formation with ASD formation is a promising approach to alter the crystallization tendency and to improve drug release of problematic poorly water-soluble compounds.


Asunto(s)
Sales (Química) , Agua , Liberación de Fármacos , Estabilidad de Medicamentos , Nitroimidazoles , Oxazoles , Solubilidad , Agua/química
6.
Int J Pharm ; 620: 121747, 2022 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-35427750

RESUMEN

Physical instability remains a major concern with amorphous solid dispersions (ASDs). In addition to bulk crystallization inhibition, another potential strategy to improve the physical stability of ASDs is surface engineering. However, coating processes are extremely challenging for ASD microparticles. Herein, we describe for the first time the application of atomic layer coating (ALC), a solvent-free technique, to deposit a pinhole-free, ultra-thin film of aluminum oxide onto the surface of spray-dried ASD particles containing high drug loadings of ezetimibe with hydroxypropyl methylcellulose acetate succinate. ALC affords excellent control over the thickness, uniformity and conformality of the coating at the atomic scale. The freshly prepared coated ASD powders exhibited less agglomeration, a lower hygroscopicity, as well as improved wettability, flowability and compressibility compared to the uncoated samples. Under accelerated storage conditions, crystallization was detected in the uncoated 50% and 70% drug loading ASDs after only a few days, whereas the coated samples showed no evidence of physical instability for two years. Consequently, there was a dramatic decrease in the drug release from the uncoated ASDs during storage, while little change was observed for the coated samples. Using ALC for surface nanocoating of ASD paves the way for the development of higher drug loading ASD without compromising physical stability, thereby reducing the pill burden.


Asunto(s)
Solubilidad , Cristalización/métodos , Composición de Medicamentos/métodos , Liberación de Fármacos , Estabilidad de Medicamentos , Polvos/química , Humectabilidad
7.
Colloids Surf B Biointerfaces ; 208: 112093, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34482192

RESUMEN

The transmembrane proteins, CD47 and signal-regulatory protein α are overexpressed in cancer cells and macrophages, respectively, and facilitate the escape of cancer cells from macrophage-mediated phagocytosis. The immunomodulatory and targeting properties of CD47, the chemotherapeutic effects of dabrafenib (D), and the anti-programmed death-1 antibodies (PD-1) pave the way for effective chemoimmunomodulation-mediated anticancer combination therapy. In this study, CD47-conjugated, D-loaded human serum albumin (HSA) nanosystems were fabricated by modified nanoparticle albumin-bound technology. Cis-aconityl-PEG-maleimide (CA), an acid-labile linker, was used to conjugate D@HSA and CD47; the resultant CD47-CA@D@HSA exhibited tumor-specificity through receptor targeting, as well as preferential cleavage and drug release in the acidic tumor microenvironment (pH 5) compared to normal physiological pH conditions (pH 6.5, 7.4). The successful preparation of nanosized (∼220 nm), narrowly dispersed (∼0.13) CD47-CA@D@HSA was proven by physicochemical characterization. In vitro and in vivo internalization, accumulation, cytotoxicity, and apoptosis were observed to be higher with CD47-conjugated nanoconstructs, than with free D or non-targeted nanoconstructs. CD47-CA@D@HSA was found to promote the infiltration of cytotoxic T cells and tumor-associated macrophages into tumors and improve in vivo tumor inhibition. Administration in combination with PD-1 further improved antitumor efficacy by promoting immune responses that blocked the immune checkpoint. No signs of toxicity were seen in mice treated with the nanoconstructs; the formulation was, therefore, thought to be biocompatible and as having potential for clinical use. The targeted chemoimmunomodulation achieved by this combination therapy was found to combat major immunosuppressive facets, making it a viable candidate for use in the treatment of cancer.


Asunto(s)
Antígeno CD47 , Albúmina Sérica Humana , Animales , Imidazoles/farmacología , Ratones , Oximas , Fagocitosis
8.
J Control Release ; 338: 211-223, 2021 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-34419495

RESUMEN

Despite the significant efforts in developing cancer vaccines, there are still numerous challenges that need to be addressed to ensure their clinical efficacy. Herein, a lymphatic dendritic cell (DC)-targeted artificial nanovaccine mimicking tumor cell membrane (ATM-NV) is developed to boost effector immune response and control immunosuppression simultaneously. The NVs are formulated with lipids, tumor cell membrane proteins, imiquimod (IMQ), and IL-10 siRNA. IL-10 siRNA is incorporated to inhibit the secretion of IL-10, an immunosuppressive cytokine, of maturated DCs upon IMQ. To enhance the DC targeting ability, the nanovaccine surface was non-covalently conjugated with the anti-CD205 antibody. The IMQ and IL-10 siRNA co-loaded, CD205 receptor-targeted artificial tumor membrane NVs (IMQ/siR@ATM-NVs) efficiently migrate to the tumor-draining lymph node and target DCs. Furthermore, immunization with IMQ/siR@ATM-NVs reduces the production of IL-10 and increases Th1-driven antitumor immunity resulted in a great tumor inhibition efficacy. Our results suggest a potential strategy to promote the vaccination's antitumor efficacy by blocking the intrinsic negative regulators in DCs.


Asunto(s)
Vacunas contra el Cáncer , Melanoma , Animales , Células Dendríticas , Humanos , Inmunidad , Interleucina-10 , Melanoma/terapia , Ratones , Ratones Endogámicos C57BL
9.
Int J Pharm ; 605: 120816, 2021 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-34161810

RESUMEN

Anticancer regimens have been substantially enriched through monoclonal antibodies targeting immune checkpoints, programmed cell death-1/programmed cell death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte antigen-4. Inconsistent clinical efficacy after solo immunotherapy may be compensated by nanotechnology-driven combination therapy. We loaded human serum albumin (HSA) nanoparticles with paclitaxel (PTX) via nanoparticle albumin-bound technology and pooled them with anti-PD-L1 monoclonal antibody through a pH-sensitive linker for targeting and immune response activation. Our tests demonstrated satisfactory preparation of paclitaxel-loaded, PD-L1-targeted albumin nanoparticles (PD-L1/PTX@HSA). They had small particle size (~200 nm) and polydispersity index (~0.12) and successfully incorporated each constituent. Relative to normal physiological pH, the formulation exhibited higher drug-release profiles favoring cancer cell-targeted release at low pH. Modifying nanoparticles with programmed cell death-ligand 1 increased cancer cell internalization in vitro and tumor accumulation in vivo in comparison with non-PD-L1-modified nanoparticles. PD-L1/PTX@HSA constructed by nanoparticle albumin-bound technology displayed successful tumor inhibition efficacy both in vitro and in vivo. There was successful effector T-cell infiltration, immunosuppressive programmed cell death-ligand 1, and regulatory T-cell suppression because of cytotoxic T-lymphocyte antigen-4 synergy. Moreover, PD-L1/PTX@HSA had low organ toxicity. Hence, the anti-tumor immune responses of PD-L1/PTX@HSA combined with chemotherapy and cytotoxic T-lymphocyte antigen-4 is a potential anti-tumor strategy for improving quantitative and qualitative clinical efficacy.


Asunto(s)
Nanopartículas , Albúminas , Línea Celular Tumoral , Liberación de Fármacos , Humanos , Inmunoterapia
10.
Biomaterials ; 269: 120677, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33503557

RESUMEN

Senescent cells drive atherosclerosis at all stages and contribute to cardiovascular disease. However, the markers in these senescent aortic plaques have not been well studied, creating a huge obstacle in the exploration of a precise and efficient system for atherosclerosis treatment. Recently, CD9 has been found to induce cellular senescence and aggravated atherosclerotic plaque formation in apolipoprotein E knockout (ApoE-/-) mice. In the present study, this result has been leveraged to develop CD9 antibody-modified, hyaluronic acid-coated mesoporous silica nanoparticles with a hyaluronidase-responsive drug release profile. In invitro models of senescent foamy macrophages and senescent endothelial cells stimulated with oxidized high-density-lipoprotein, the CD9 antibody-modified mesoporous silica nanoparticles exhibit high cellular uptake; reduce the reactive oxygen species level, high-density lipoprotein oxidation, and production of TNF-α and IL-6; and attenuate the senescence process, contributing to improved cell viability. In vivo experiment demonstrated that these nanoparticles can successfully target the senescent lesion areas, deliver the anti-senescence drug rosuvastatin to the senescent atherosclerotic plaques (mainly endothelial cells and macrophages), and alleviate the progression of atherosclerosis in ApoE-/- mice. By providing deep insight regarding the markers in senescent atherosclerotic plaque and developing a nano-system targeting this lesion area, the study proposes a novel and an accurate therapeutic approach for mitigating atherosclerosis through senescent cell clearance.


Asunto(s)
Aterosclerosis , Células Endoteliales , Macrófagos , Nanopartículas , Placa Aterosclerótica , Animales , Aorta , Aterosclerosis/tratamiento farmacológico , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados para ApoE , Placa Aterosclerótica/tratamiento farmacológico , Dióxido de Silicio
11.
J Control Release ; 329: 524-537, 2021 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-32971203

RESUMEN

Accumulating clinical data shows that less than half of patients are beneficial from PD-1/PD-L1 blockage therapy owing to the limited infiltration of effector immune cells into the tumor and abundant of the immunosuppressive factors in the tumor microenvironment. In this study, PD-L1 inhibition therapy and BRAF-targeted therapy, which showed clinical benefit, were combined in a CXCR4-targeted nanoparticle co-delivering dabrafenib (Dab), a BRAF inhibitor, and miR-200c which can down-regulate PD-L1 expression. The cationic PCL-PEI core containing Dab- and miR-200c- were coated with poly-L-glutamic acid conjugated with LY2510924, a CXCR-4 antagonist peptide, (PGA-pep) to obtain miR@PCL-PEI/Dab@PGA-pep nanoformulation. The stimulus pH- and redox- reactive of PGA-pep was ascribed to exhibit an enhanced release of drug in the tumor microenvironment as well as improve the stability of miR-200c during the blood circulation. In addition, the presence of LY2510924 peptide would enhance the binding affinity of miR@PCL-PEI/Dab@PGA-pep NPs to cancer cells, leading to improved cellular uptake, cytotoxicity, and in vivo accumulation into tumor area. The in vivo results indicated that both, the immunogenic cell death (ICD) and the inhibition of PD-L1 expression, induced by treatment with CXCR-4 targeted nanoparticles, enables to improve the DC maturation in lymph node and CD8+ T cell activation in the spleen. More importantly, effector T cells were increasingly infiltrated into the tumor, whereas the immunosuppressive factors like PD-L1 expression and regulatory T cells were significantly reduced. They, all together, promote the immune responses against the tumor, indicating the therapeutic efficiency of the current strategy in cancer treatment.


Asunto(s)
MicroARNs , Nanopartículas , Neoplasias , Línea Celular Tumoral , Humanos , Sistema Inmunológico , Microambiente Tumoral
12.
Arch Womens Ment Health ; 24(2): 271-279, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32728774

RESUMEN

Intimate partner violence (IPV) perpetrators are often husbands. Understanding factors pertaining to women's male partners is essential for programming interventions against IPV. The objective of the study was to describe husband-related social and behavioural risk factors and assess how they are associated with IPV during pregnancy. Cross-sectional data were collected among 1309 pregnant women with husbands in Dong Anh district, Vietnam. Information on sociodemographic characteristics of husbands, the husband's behaviour and the husband's involvement in pregnancy care was indirectly collected via women's report at first antenatal care visit. Data on exposure to intimate partner violence during pregnancy were collected when the women returned for antenatal care in 30-34 gestational weeks. Logistic regression analyses were used to measure the relationships between IPV during pregnancy and risk factors from the husband. Pregnant women who had husbands who were younger or blue-collar worker/farmer/unemployed had more likelihood to be exposed to IPV. Women with husbands who drank alcohol before sexual intercourse and gambled were more likely to be exposed to IPV repeated times. Those with husbands who had intentions of having a child had over three times increased OR to be exposed to IPV once (AOR = 3.2, 95% CI 1.1-9.7). If the husband had a preference for sons, the woman had 1.5 times increased OR (AOR = 1.5; 95% CI 1.1-1.9) to be exposed to IPV repeated times during pregnancy. This study highlights significant associations between IPV and maternal perceptions of husbands' behaviours and involvement in pregnancy. Findings may help to identify at-risk pregnant women to IPV and guide the development of targeted interventions to prevent IPV from husbands.


Asunto(s)
Violencia de Pareja , Esposos , Niño , Estudios Transversales , Femenino , Humanos , Masculino , Embarazo , Mujeres Embarazadas , Factores de Riesgo , Vietnam/epidemiología
13.
Acta Biomater ; 115: 371-382, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32798721

RESUMEN

The therapeutic efficacy of current cancer vaccines is far from optimal, mainly because of insufficient induction of antigen-specific T cells and because tumor cells can hijack immunosuppressive mechanisms to evade the immune responses. Generating specific, robust, and long-term immune responses against cancer cells and the attenuating of immunosuppressive factors are critical for effective cancer vaccination. Recently, the engineering of exosomes specifically bind to T cells, and then stimulating tumor-specific T-cell immune responses has emerged as a potential alternative strategy for cancer vaccination. In this study, we generated a bifunctional exosome combining the strategy of vaccination and checkpoint blockade. Exosomes prepared from Ovalbumin (OVA)-pulsed, activated dendritic cells were modified with anti-CTLA-4 antibody (EXO-OVA-mAb) to block this inhibitory molecule and to enhance the specificity of the exosomes toward T cells. Our study provides a unique strategy for functionalizing exosome membrane with anti-CTLA-4 antibody via lipid-anchoring method to synergize efficacy of cancer vaccination and immune checkpoint blockade against the tumor. STATEMENT OF SIGNIFICANCE: We designed T-cell-targeting exosomes (EXO-OVA-mAb) decorated with costimulatory molecules, MHCs, antigenic OVA peptide, and anti-CTLA-4 antibody, combining the strategies of vaccines and checkpoint blockade. The exosomes showed enhanced binding to T cells in tumor-draining lymph nodes, effectively induced T-cell activation, and improved the tumor homing of effector T cells, ultimately significantly restraining tumor growth. Thus, EXO-OVA-mAb greatly facilitates T-cell targeting, induces a strong tumor-specific T-cell response, and increased the ratio of effector T cells/regulatory T cells within tumors, resulting in appreciable tumor growth inhibition.


Asunto(s)
Vacunas contra el Cáncer , Exosomas , Animales , Línea Celular Tumoral , Células Dendríticas , Ganglios Linfáticos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL
14.
Lab Anim Res ; 36: 18, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32551299

RESUMEN

[This retracts the article DOI: 10.5625/lar.2018.34.2.49.].

15.
J Control Release ; 324: 413-429, 2020 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-32461115

RESUMEN

Hypoxia is a common feature of the tumor microenvironment, which is characterized by tissue oxygen deficiency due to an aggressive proliferation of cancer cells. Hypoxia activates hypoxia-inducible factor-dependent signaling, which in turn regulates metabolic reprogramming, immune suppression, resistance to apoptosis, angiogenesis, metastasis, and invasion to secondary sites. In this review, we provide an overview of the use of nanotechnology to harmonize intra-tumoral oxygen or suppress hypoxia-related signaling for an improved efficacy of cancer treatment. The biological background was followed by conducting a literature review on the (1) nanoparticles responsible for enhancing oxygen levels within the tumor, (2) nanoparticles sensitizing hypoxia, (3) nanoparticles suppressing hypoxia-inducing factor, (4) nanoparticles that relieve tumor hypoxia for enhancement of chemotherapy, photodynamic therapy, and immunotherapy, either individually or in combination. Lastly, the heterogeneity of cancer and limitations of nanotechnology are discussed to facilitate translational therapeutic treatment.


Asunto(s)
Neoplasias , Fotoquimioterapia , Humanos , Hipoxia/terapia , Nanotecnología , Neoplasias/tratamiento farmacológico , Hipoxia Tumoral , Microambiente Tumoral
16.
J Control Release ; 321: 509-518, 2020 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-32087300

RESUMEN

Clinical intraportal pancreatic islet infusion is popular for treating type I diabetes. However, multiple doses of islets and anti-rejection protocols are needed to compensate for early large cell losses post-infusion due to the harsh hepatic environment. Thus, extrahepatic sites are utilized to enable efficient islet engraftment and reduce islet mass. Here, we reported an effective islet revascularization protocol that was based on the co-implantation of islet/fibrin gel construct with poly(lactic-co-glycolic) acid sheet releasing NECA (5'-(N-ethylcarboxamido) adenosine; a potent agonist of adenosine) into mouse epididymal fat pad. Thin, flexible sheets (d = 4 mm) prepared by simple casting exhibited sustained NECA release for up to 21 days, which effectively improved early islet engraftment with a median diabetic reversal time of 18.5 days. Western blotting revealed the facilitative effect of NECA on VEGF expression from islets in vitro and from grafts in vivo. In addition, NECA directly promoted the angiogenic activities of islet-derived endothelial cells by enhancing their proliferation and vessel-like tube formation. As a result, neovasculatures were effectively formed in the engrafted islet vicinity, as evidenced by vasculature imaging and immunofluorescence. Taken together, we suggest NECA-releasing PLGA sheets offer a safe and effective drug delivery system that enhances islet engraftment while reducing islet mass at extrahepatic sites for clinical relevance.


Asunto(s)
Adenosina-5'-(N-etilcarboxamida) , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Prótesis e Implantes , Animales , Células Endoteliales , Ratones , Trasplante de Órganos , Polímeros
17.
J Control Release ; 315: 126-138, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31672625

RESUMEN

In this study, dual drug-loaded nanoparticles were constructed to co-deliver low-dose doxorubicin (DOX) and miR-200c (DOX/miR-NPs) to inhibit programmed death-1 receptor (PD-L1) expression and trigger immunogenic cell death (ICD) in cancer cells. Two block copolymers, folic acid (FA)-conjugated PLGA-PEG (PLGA-PEG-FA) and PLGA-PEI, were formulated as folate-targeted NPs and loaded with DOX and miR-200c. The NPs, which were formed as nanosize objects (110.4 ± 2.1) with narrow size distribution (0.19 ± 0.02), effectively protected the miR-200c from degradation in serum. Modifying the NPs with FA increased not only their uptake by cancer cells in vitro but also their accumulation in tumor microenvironments in vivo, as compared with those properties of non-FA-modified NPs. The DOX/miR-NPs also exhibited efficacious inhibition of PD-L1 expression and robust induction of ICD in cancer cells in vitro and in vivo, resulting in increased dendritic cell maturation and CD8+ T cell response towards cancer cells. Furthermore, tumor growth was significantly inhibited by folate-targeted NPs loaded with the low-dose DOX/miR-200c combination, but not by treatments with free DOX, miR-NPs or DOX-NPs. Thus, our results suggest that simultaneous PD-L1 inhibition via microRNAs and the induction of an immunogenic tumor microenvironment via low-dose cytotoxic drugs may improve cancer therapy efficacy.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , MicroARNs/administración & dosificación , Nanopartículas , Animales , Antibióticos Antineoplásicos/inmunología , Antígeno B7-H1/antagonistas & inhibidores , Línea Celular Tumoral , Doxorrubicina/inmunología , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Femenino , Ácido Fólico/química , Humanos , Muerte Celular Inmunogénica/inmunología , Ratones Endogámicos C57BL , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Poliésteres/química , Polietilenglicoles/química , Linfocitos T/inmunología , Microambiente Tumoral/inmunología
18.
Biomaterials ; 221: 119415, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31419652

RESUMEN

Host immune response remains an obstacle in cell-replacement therapy for treating type I diabetes. Long-term systemic immunosuppression results in suboptimal efficacy and adverse reactions. Thus, "cell-particle hybrids" of pancreatic islets and tissue-adhesive, polydopamine-coated, FK506-loaded biodegradable microspheres (PD-FK506-MS) were developed to locally modulate the immune response at the transplantation site. Coating of FK506-MS with PD enabled the rapid formation of stable cell-particle hybrids without significant changes in islet viability and functionality. Extremely low quantities of FK506 (approximately 600 ng per recipient) sustainably released from cell-particle hybrids effectively prolonged survival of xenogeneic islet graft. Interestingly, FK506 exhibited extended bioavailability in the grafts but was undetectable in systemic circulation and other tissues. Moreover, mRNA expression of inflammatory cytokines was significantly inhibited in the PD-FK506-MS-containing grafts but not in lymphoid organs. This study presents a promising platform that facilitates the translation of local immunomodulation towards an effective strategy with improved safety profiles for treating type I diabetes.


Asunto(s)
Terapia de Inmunosupresión/métodos , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/metabolismo , Microesferas , Trasplante Heterólogo/métodos , Animales , Citometría de Flujo , Prueba de Tolerancia a la Glucosa , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Polímeros/química , Tacrolimus
19.
ACS Appl Mater Interfaces ; 11(28): 24959-24970, 2019 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-31265222

RESUMEN

Inorganic nanomaterial (INM)-based combination cancer therapies have been extensively employed over the past two decades because of their benefits over traditional chemo- and radiotherapies. However, issues regarding the toxicity and accumulation of INMs in the body have arisen. This problem may be improved through the use of biodegradable or disintegrable nanosystems such as black phosphorus (BP). Challenges to the manufacture of fully nanodimensional BP remain. In addition, improvements in recently developed cancer immunotherapies require their incorporation with drugs, targeting agents, and delivery vehicles. With these needs in mind, this study develops a method for instant in-flight manufacture of nanodimensional BP using plug-and-play devices for subsequent assembly of photoimmunotherapeutic core@shell composites containing mutated B-raf inhibitors (dabrafenib), immune checkpoint inhibitors (PD-L1), and cancer-targeting antibodies (CXCR4). The resulting nanocomposites exhibited cancer targetability and regulatability of PD-L1 expression both in vitro and in vivo. These activities were further increased upon near-infrared irradiation due to the incorporation of a phototherapeutic component. These results suggest that these nanocomposites are promising as a new class of advanced cancer therapeutic agents.


Asunto(s)
Antineoplásicos , Imidazoles , Inmunoterapia , Melanoma Experimental/terapia , Nanocompuestos , Oximas , Fototerapia , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Imidazoles/química , Imidazoles/farmacología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Nanocompuestos/química , Nanocompuestos/uso terapéutico , Oximas/química , Oximas/farmacología
20.
Acta Biomater ; 94: 82-96, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31129358

RESUMEN

The expression of Toll-like receptors (TLRs) on antigen presenting cells, especially dendritic cells, offers several sensitive mediators to trigger an adaptive immune response, which potentially can be exploited to detect and eliminate pathogenic objects. Consequently, numerous agonists that target TLRs are being used clinically either alone or in combination with other therapies to strengthen the immune system in the battle against cancer. This review summarizes the roles of TLRs in tumor biology, and focuses on relevant TLR-dependent antitumor pathways and the conjugation of TLR agonists as adjuvants to nano- and micro-particles for boosting responses leading to cancer suppression and eradication. STATEMENT OF SIGNIFICANCE: Toll-like receptors (TLRs), which express on antigen presenting cells, such as dendritic cells and macrophages, play an important role in sensing pathogenic agents and inducing adaptive immunity. As a result, several TLR agonists have been investigating as therapeutic agents individually or in combination with other treatment modalities for cancer treatment through boosting the immune system. This review aims to focus on the roles of TLRs in cancer and TLR-dependent antitumor pathways as well as the use of different nano- or micro-particles bearing TLR agonists for tumor inhibition and elimination.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Receptores Toll-Like/metabolismo , Microambiente Tumoral , Inmunidad Adaptativa , Adyuvantes Inmunológicos , Animales , Células Presentadoras de Antígenos/inmunología , Células Dendríticas/metabolismo , Humanos , Sistema Inmunológico , Nanopartículas/química , ARN Mensajero/metabolismo , Transducción de Señal , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo , Receptor Toll-Like 9/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...