Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
2.
Mol Pharm ; 20(8): 3895-3913, 2023 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-37459272

RESUMEN

Overexpression of the antiapoptotic protein B-cell lymphoma 2 (Bcl-2) is correlated with poor survival outcomes in triple-negative breast cancer (TNBC), making Bcl-2 inhibition a promising strategy to treat this aggressive disease. Unfortunately, Bcl-2 inhibitors developed to date have limited clinical success against solid tumors, owing to poor bioavailability, insufficient tumor delivery, and off-target toxicity. To circumvent these problems, we loaded the Bcl-2 inhibitor ABT-737 in poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) that were wrapped with phospholipid membranes derived from 4T1 murine mammary cancer cells, which mimic the growth and metastasis of human TNBC. We show that the biomimetic cancer cell membrane coating enabled the NPs to preferentially target 4T1 TNBC cells over noncancerous mammary epithelial cells in vitro and significantly increased NP accumulation in orthotopic 4T1 tumors in mice after intravenous injection by over 2-fold compared to poly(ethylene glycol)-poly(lactide-co-glycolic) (PEG-PLGA) copolymer NPs. Congruently, the ABT-737 loaded, cancer cell membrane-wrapped PLGA NPs (ABT CCNPs) induced higher levels of apoptosis in TNBC cells in vitro than ABT-737 delivered freely or in PEG-PLGA NPs. When tested in a syngeneic spontaneous metastasis model, the ABT CCNPs significantly increased apoptosis (evidenced by elevated active caspase-3 and decreased Bcl-2 staining) and decreased proliferation (denoted by reduced Ki67 staining) throughout tumors compared with saline or ABT-loaded PEG-PLGA NP controls. Moreover, the ABT CCNPs did not alter animal weight or blood composition, suggesting that the specificity afforded by the TNBC cell membrane coating mitigated the off-target adverse effects typically associated with ABT-737. Despite these promising results, the low dose of ABT CCNPs administered only modestly reduced primary tumor growth and metastatic nodule formation in the lungs relative to controls. We posit that increasing the dose of ABT CCNPs, altering the treatment schedule, or encapsulating a more potent Bcl-2 inhibitor may yield more robust effects on tumor growth and metastasis. With further development, drug-loaded biomimetic NPs may safely treat solid tumors such as TNBC that are characterized by Bcl-2 overexpression.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Membrana Celular
3.
Int J Hyperthermia ; 40(1): 2272066, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37903544

RESUMEN

Cancer is a devastating health problem with inadequate treatment options. Many conventional treatments for solid-tumor cancers lack tumor specificity, which results in low efficacy and off-target damage to healthy tissues. Nanoparticle (NP)-mediated photothermal therapy (PTT) is a promising minimally invasive treatment for solid-tumor cancers that has entered clinical trials. Traditionally, NPs used for PTT are coated with passivating agents and/or targeting ligands, but alternative coatings are being explored to enhance tumor specific delivery. In particular, cell-derived membranes have emerged as promising coatings that improve the biointerfacing of photoactive NPs, which reduces their immune recognition, prolongs their systemic circulation and increases their tumor accumulation, allowing for more effective PTT. To maximize treatment success, membrane-wrapped nanoparticles (MWNPs) that enable dual tumor imaging and PTT are being explored. These multifunctional theranostic NPs can be used to enhance tumor detection and/or ensure a sufficient quantity of NPs that have arrived in the tumor prior to laser irradiation. This review summarizes the current state-of-the-art in engineering MWNPs for combination cancer imaging and PTT and discusses considerations for the path toward clinical translation.


Asunto(s)
Nanopartículas , Neoplasias , Línea Celular Tumoral , Fototerapia/métodos , Nanopartículas/uso terapéutico , Diagnóstico por Imagen , Neoplasias/diagnóstico por imagen , Neoplasias/terapia
4.
Genes Dev ; 29(7): 732-45, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25838542

RESUMEN

Glioblastoma multiforme (GBM) is a lethal, therapy-resistant brain cancer consisting of numerous tumor cell subpopulations, including stem-like glioma-initiating cells (GICs), which contribute to tumor recurrence following initial response to therapy. Here, we identified miR-182 as a regulator of apoptosis, growth, and differentiation programs whose expression level is correlated with GBM patient survival. Repression of Bcl2-like12 (Bcl2L12), c-Met, and hypoxia-inducible factor 2α (HIF2A) is of central importance to miR-182 anti-tumor activity, as it results in enhanced therapy susceptibility, decreased GIC sphere size, expansion, and stemness in vitro. To evaluate the tumor-suppressive function of miR-182 in vivo, we synthesized miR-182-based spherical nucleic acids (182-SNAs); i.e., gold nanoparticles covalently functionalized with mature miR-182 duplexes. Intravenously administered 182-SNAs penetrated the blood-brain/blood-tumor barriers (BBB/BTB) in orthotopic GBM xenografts and selectively disseminated throughout extravascular glioma parenchyma, causing reduced tumor burden and increased animal survival. Our results indicate that harnessing the anti-tumor activities of miR-182 via safe and robust delivery of 182-SNAs represents a novel strategy for therapeutic intervention in GBM.


Asunto(s)
Apoptosis/genética , Diferenciación Celular/genética , Glioblastoma/genética , MicroARNs/metabolismo , Animales , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/fisiopatología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/tratamiento farmacológico , Glioblastoma/fisiopatología , Humanos , Ratones , Ratones SCID , MicroARNs/administración & dosificación , MicroARNs/genética , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Análisis de Supervivencia
5.
Nano Lett ; 21(1): 68-76, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33306406

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive disease that requires new interventions. A promising approach to improve patient prognosis is to introduce tumor suppressive miR-34a into TNBC cells. Unfortunately, naked miR-34a is not effective therapeutically because it is degraded by nucleases and cannot passively enter cells. Nanocarriers designed to increase miR-34a stability and cellular entry have lacked specificity and potency. To overcome these limitations, we conjugated miR-34a to photoresponsive gold nanoshells (NS), which can release tethered miR-34a upon excitation with continuous wave (CW) or nanosecond (ns) pulsed near-infrared light to facilitate on-demand gene regulation. We demonstrate that miR-34a/NS can regulate downstream miR-34a targets following irradiation to reduce TNBC cell viability, proliferation, and migration. Further, we show ns pulsed light releases miRNA more effectively than CW light, and that released miR-34a is as potent as transfected miR-34a. These findings signify miR-34a/NS as promising tools for precisely controlled gene regulation of TNBC.


Asunto(s)
MicroARNs , Nanocáscaras , Neoplasias de la Mama Triple Negativas , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Pronóstico , Neoplasias de la Mama Triple Negativas/genética
6.
Bioconjug Chem ; 32(10): 2245-2256, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34543006

RESUMEN

The use of nucleic acids to regulate gene expression is a rapidly developing field with immense clinical potential. Nanomaterials are frequently used to deliver nucleic acids into cells as they can overcome the poor cellular uptake and endo/lysosomal degradation of bare nucleic acids. For these nanocarriers to be effective, they must escape endo/lysosomal compartments to deliver their nucleic acid cargo into the cytosol (for ribonucleic acid (RNA)) or nucleus (for deoxyribonucleic acid (DNA)). This process is poorly understood and remains an area of active research toward the goal of developing effective delivery strategies. Fluorescent endo/lysosomal markers are among the most widely employed tools used to evaluate the endosomal escape of nucleic acid nanocarriers. However, the endo/lysosomal labeling method may alter the extent of and route of nanocarrier uptake by cells. The impact of these markers on cellular function and cell-nanocarrier interactions has not been probed in a systematic manner. To investigate this, we compared the effects of several common lysosomal labeling methods, namely, LysoTracker Red (LT Red), transient lysosomal-associated membrane protein 1-mutant green fluorescent protein (LAMP1-mGFP) transfection (Transient GFP), and stable lentiviral LAMP1-mGFP transfection (Stable GFP), on cellular metabolic activity, nanocarrier uptake, nanocarrier/lysosomal label colocalization, and gene silencing potency in U87 glioblastoma and MDA-MB-231 breast cancer cells using polyethyleneimine (PEI)/ribonucleic acid (RNA) polyplexes as a model nanocarrier. In both U87s and MDA-MB-231s, Transient GFP and LT Red labeling reduced metabolic activity relative to untransfected (Parental) cells, while Stable GFP labeling increased metabolic activity. Congruently, flow cytometry indicates Stable GFP cells have greater polyplex uptake than LT Red-labeled cells in both cell lines. Despite these similar trends in uptake, polyplex intracellular trafficking differs in the two cell lines, as confocal imaging revealed greater polyplex/lysosome colocalization in Stable GFP U87 cells than LT Red-labeled U87 cells, while the trend was reversed in MBA-MB-231s. The level of RNA-mediated gene silencing achieved in Parental versus Stable GFP U87 and MDA-MB-231 cells agreed with the observed levels of polyplex/lysosome colocalization, supporting the established concept that endosomal escape is the rate-limiting step for RNA interference. These findings indicate that lysosomal labels can profoundly alter cellular function and cell-nanocarrier interactions, presenting critical new considerations for researchers investigating nanoparticle trafficking.


Asunto(s)
Polietileneimina , Transfección , Lisosomas
7.
Nanomedicine ; 36: 102412, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34147664

RESUMEN

The use of nanoparticles (NPs) to deliver therapeutics to reproductive organs is an emerging approach to safely and effectively treat mothers and babies facing pregnancy complications. This study investigates the biodistribution of two different sized gold-based NPs in pregnant mice following systemic delivery as a function of gestational age. Poly(ethylene glycol)-coated 15 nm gold nanoparticles or 150 nm diameter silica core/gold nanoshells were intravenously administered to pregnant mice at gestational days (E)9.5 or 14.5. NP distribution was analyzed twenty-four hours later by inductively coupled plasma-mass spectrometry and silver staining of histological specimens. More NPs accumulated in placentas than embryos and delivery to these tissues was greater at E9.5 than E14.5. Neither NP type affected fetal weight or placental weight, indicating minimal short-term toxicity in early to mid-stage pregnancy. These findings warrant continued development of NPs as tools to deliver therapeutics to reproductive tissues safely.


Asunto(s)
Materiales Biocompatibles Revestidos , Embrión de Mamíferos/metabolismo , Edad Gestacional , Oro , Nanopartículas del Metal , Placenta/metabolismo , Animales , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacocinética , Materiales Biocompatibles Revestidos/farmacología , Femenino , Oro/química , Oro/farmacocinética , Oro/farmacología , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Ratones , Embarazo
8.
Nano Lett ; 18(6): 3565-3570, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-29701993

RESUMEN

The ability to regulate intracellular gene expression with exogenous nucleic acids such as small interfering RNAs (siRNAs) has substantial potential to improve the study and treatment of disease. However, most transfection agents and nanoparticle-based carriers that are used for the intracellular delivery of nucleic acids cannot distinguish between diseased and healthy cells, which may cause them to yield unintended widespread gene regulation. An ideal delivery system would only silence targeted proteins in diseased tissue in response to an external stimulus. To enable spatiotemporal control over gene silencing, researchers have begun to develop nucleic acid-nanoparticle conjugates that keep their nucleic acid cargo inactive until it is released from the nanoparticle on-demand by externally applied near-infrared laser light. This strategy can overcome several limitations of other nucleic acid delivery systems, but the mechanisms by which these platforms operate remain ill understood. Here, we perform a detailed investigation of the mechanisms by which silica core/gold shell nanoshells (NSs) release conjugated siRNA upon excitation with either pulsed or continuous wave (CW) near-infrared (NIR) light, with the goal of providing insight into how these nanoconjugates can enable on-demand gene regulation. We demonstrate that siRNA release from NSs upon pulsed laser irradiation is a temperature-independent process that is substantially more efficient than siRNA release triggered by CW irradiation. Contrary to literature, which suggests that only pulsed irradiation releases siRNA duplexes, we found that both modes of irradiation release a mixture of siRNA duplexes and single-stranded oligonucleotides, but that pulsed irradiation results in a higher percentage of released duplexes. To demonstrate that the siRNA released from NSs upon pulsed irradiation remains functional, we evaluated the use of NSs coated with green fluorescent protein (GFP)-targeted siRNA (siGFP-NS) for on-demand knockdown of GFP in cells. We found that GFP-expressing cells treated with siGFP-NS and irradiated with a pulsed laser experienced a 33% decrease in GFP expression compared to cells treated with no laser. Further, we observed that light-triggered gene silencing mediated by siGFP-NS is more potent than using commercial transfection agents to deliver siRNA into cells. This work provides unprecedented insight into the mechanisms by which plasmonic NSs release siRNA upon light irradiation and demonstrates the importance of thoroughly characterizing photoresponsive nanosystems for applications in triggered gene regulation.


Asunto(s)
Preparaciones de Acción Retardada/química , Nanocáscaras/química , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , Transfección , Proteínas Fluorescentes Verdes/genética , Humanos , Luz , Imagen Óptica , ARN Interferente Pequeño/genética , Transfección/métodos
9.
Mol Pharm ; 15(11): 5135-5145, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30260647

RESUMEN

Glioblastoma (GBM) is the most common and lethal primary brain tumor in adults, with nearly 100% of patients ultimately succumbing to the disease. Median patient survival is 15 months, and no standard of care currently exists for recurrent cases. Glioma stem cells (GSCs), a rare and highly aggressive subpopulation of cells within these tumors, have recently emerged as drivers of tumor initiation and recurrence, and a growing body of evidence suggests that they must be completely eradicated to prevent relapse. Toward this goal, we have developed polyethylenimine-wrapped spherical nucleic acid nanoparticles (PEI-SNAs) targeting Gli1, a transcription factor within the Hedgehog signaling pathway that is crucial for the maintenance of GSCs. Here, we demonstrate that Gli1 PEI-SNAs bind scavenger receptors on GBM cells to undergo endocytosis in a caveolae/lipid raft/dynamin-dependent manner. They further achieve ∼30% silencing of tumor-promoting Hedgehog pathway genes and downstream target genes that promote the aggressive, chemoresistant phenotype of GBM. This produces a 30% decrease in proliferation that correlates with a robust onset of GBM cell senescence as well as an ∼60% decrease in metabolic activity with or without cotreatment with temozolomide (TMZ), the frontline chemotherapy for GBM. Most importantly, Gli1 PEI-SNAs impair the self-renewal capacity of GBM cells as indicated by a 30-40% reduction in the expression of stemness genes and further impair the formation of stem-like neurospheres. They also substantially improve neurosphere chemosensitivity as demonstrated by a 2-fold increase in the fraction of cells undergoing apoptosis in response to low doses of TMZ. These results underscore the potential for siRNA therapeutics targeting Gli1 to reduce GBM resistance to therapy and warrant further development of PEI-SNAs and Gli1-targeted therapies to alleviate drug resistance and recurrence for GBM patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Portadores de Fármacos/química , Glioblastoma/tratamiento farmacológico , ARN Interferente Pequeño/administración & dosificación , Proteína con Dedos de Zinc GLI1/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Glioblastoma/patología , Humanos , Nanopartículas/química , Células Madre Neoplásicas , Polietileneimina/química , ARN Interferente Pequeño/genética , Temozolomida/farmacología , Temozolomida/uso terapéutico , Proteína con Dedos de Zinc GLI1/genética
10.
Inorg Chem ; 57(17): 10608-10615, 2018 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-30132325

RESUMEN

Photodynamic therapy (PDT) represents a minimally invasive and highly localized treatment strategy to ablate tumors with few side effects. In PDT, photosensitizers embedded within tumors are activated by light and undergo intersystem crossing, followed by energy transfer to molecular oxygen, resulting in the production of toxic singlet oxygen (1O2). Previously, we reported a robust, linear tetrapyrrole palladium(II) complex, Pd[DMBil1], characterized by its facile and modular synthesis, broad absorption profile, and efficient 1O2 quantum yield of ΦΔ = 0.8 in organic media. However, the insolubility of this porphyrinoid derivative in aqueous solution prevents its use under biologically relevant conditions. In this work, we report the synthesis of Pd[DMBil1]-PEG750, a water-soluble dimethylbiladiene derivative that is appended with a poly(ethylene) glycol (PEG) functionality. Characterization of this complex shows that this PEGylated biladiene architecture maintains the attractive photophysical properties of the parent complex under biologically relevant conditions. More specifically, the absorption profile of Pd[DMBil1]-PEG750 closely matches that of Pd[DMBil1] and obeys the Beer-Lambert Law, suggesting that the complex does not aggregate under biologically relevant conditions. Additionally, the emission spectrum of Pd[DMBil1]-PEG750 retains the fluorescence and phosphorescence features characteristic of Pd[DMBil1]. Importantly, the PEGylated photosensitizer generates 1O2 with ΦΔ = 0.57, which is well within the range to warrant interrogation as a potential PDT agent for treatment of cancer cells. The Pd[DMBil1]-PEG750 is biologically compatible, as it is taken up by MDA-MB-231 triple negative breast cancer (TNBC) cells and has an effective dose (ED50) of only 0.354 µM when exposed to λex > 500 nm light for 30 min. Impressively, the lethal dose (LD50) of Pd[DMBil1]-PEG750 without light exposure was measured to be 1.87 mM, leading to a remarkably high phototoxicity index of ∼5300, which is vastly superior to existing photosensitizers that form the basis for clinical PDT treatments. Finally, through flow cytometry experiments, we show that PDT with Pd[DMBil1]-PEG750 induces primarily apoptotic cell death in MDA-MB-231 cells. Overall these results demonstrate that Pd[DMBil1]-PEG750 is an easily prepared, biologically compatible, and well-tolerated photochemotherapeutic agent that can efficiently drive the photoinduced apoptotic death of TNBC cells.

11.
Small ; 13(26)2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28544579

RESUMEN

Antibodies that antagonize cell signaling pathways specific to their targeted receptor are invaluable tools to study and treat malignancies, but their utility is limited by high production costs and treatment dosages. Researchers have shown that antibodies conjugated to nanoparticles display increased affinity for their target relative to freely delivered antibodies due to multivalency, and this study investigates how this multivalency can enable antibody-nanoparticle conjugates to inhibit oncogenic cell signaling more effectively than freely delivered antibodies. This effect was evaluated using triple negative breast cancer (TNBC) cells that are characterized by hyperactive Wnt signaling mediated through overexpressed Frizzled7 (FZD7) transmembrane receptors. Through analysis of the expression of ß-catenin and Axin2, two downstream targets in the Wnt pathway, the results demonstrate that FZD7 antibody-nanoshell conjugates (FZD7-NS) are drastically more effective at inhibiting Wnt signaling in TNBC cells than freely delivered FZD7 antibodies. Additionally, cells treated with FZD7-NS, but not cells treated with freely delivered FZD7 antibodies, have decreased viability, indicating the therapeutic potential of this technology. The results demonstrate that antibody-functionalized nanoparticles can exploit multivalency for improved signal cascade interference over free antibodies, and this may ultimately permit lower antibody dosages to be administered to study signaling pathways or to manage diseases.


Asunto(s)
Anticuerpos/farmacología , Receptores Frizzled/antagonistas & inhibidores , Nanopartículas/química , Nanocáscaras/química , Neoplasias de la Mama Triple Negativas/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Anticuerpos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos
12.
ACS Biomater Sci Eng ; 10(3): 1355-1363, 2024 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-38306303

RESUMEN

There is an outstanding need for targeted therapies for triple-negative breast cancer (TNBC), an aggressive breast cancer subtype. Since TNBC's rapid growth and metastasis are driven by hyperactive Wnt signaling, suppressing the key-pathway mediator ß-catenin through RNA interference may improve patient outcomes. However, small interfering ribonucleic acid (siRNA) molecules require a carrier to elicit targeted gene silencing. Here, we show that 4T1 cancer cell membrane wrapped poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) can deliver siRNA into TNBC cells, silence ß-catenin expression, and reduce the cells' tumorigenic qualities. Compared to unwrapped and nontargeted NPs, the cancer cell membrane wrapped nanoparticles (CCNPs) exhibit dramatically improved uptake by TNBC cells versus breast epithelial cells and greater gene silencing at mRNA and protein levels. Congruently, ß-catenin siRNA-loaded CCNPs significantly activate senescence in 2D cultured TNBC cells and reduce proliferation in 3D spheroids. This work advances the development of nucleic acid carriers for targeted RNA interference therapy.


Asunto(s)
Nanopartículas , Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Interferencia de ARN , beta Catenina/genética , beta Catenina/metabolismo , Línea Celular Tumoral , ARN Interferente Pequeño/genética , Nanopartículas/uso terapéutico
13.
ACS Omega ; 9(12): 14323-14335, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38559981

RESUMEN

Doxorubicin (DOX) is a chemotherapy agent commonly used to treat triple-negative breast cancer (TNBC), but it has insufficient efficacy against the disease and considerable toxicity due to its off-target delivery. To improve the specificity of DOX for TNBC, we encapsulated it in poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) coated with antibodies against Frizzled7 (FZD7), a receptor that is overexpressed on TNBC cells and which is a key activator of the Wnt signaling pathway. In vitro studies show that DOX encapsulation does not hinder its ability to localize to the nucleus in human TNBC cell cultures and that DOX delivered via NPs induces apoptosis and DNA damage via H2A.X phosphorylation to the same degree as freely delivered DOX. FZD7-targeted NPs delivering DOX caused significantly greater inhibition of metabolic activity and led to a smaller cell population following treatment when compared to freely delivered DOX or DOX-loaded NPs coated only with poly(ethylene glycol) (PEG). The FZD7 antibodies additionally provided significant levels of Wnt pathway inhibition, as demonstrated by an increase in ß-catenin phosphorylation, indicative of ß-catenin destruction and downregulation. These results show that FZD7-targeted platforms have great promise for improving the therapeutic window of otherwise toxic chemotherapies like DOX in TNBC and other cancers that display the overexpression of FZD7 receptors.

14.
Drug Deliv Transl Res ; 13(12): 3003-3013, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37365402

RESUMEN

At the onset of pregnancy, people with preexisting conditions face additional challenges in carrying their pregnancy to term, as the safety of the developing fetus and pregnant person is a significant factor of concern. Nanoparticle (NP)-based therapies have displayed success against various conditions and diseases in non-pregnant patients, but the use of NPs in maternal-fetal health applications needs to be better established. Local vaginal delivery of NPs is a promising administration route with the potential to yield high cargo retention in the vagina and improved therapeutic efficacy compared to systemic administration that results in rapid NP clearance by the hepatic first-pass effect. In this study, we investigated the biodistribution and short-term toxicity of poly(ethylene glycol)-poly(lactic-co-glycolic acid) (PEG-PLGA) NPs in pregnant mice following vaginal delivery. The NPs were either loaded with DiD fluorophores for tracking cargo distribution (termed DiD-PEG-PLGA NPs) or included Cy5-tagged PLGA in the formulation for tracking polymer distribution (termed Cy5-PEG-PLGA NPs). DiD-PEG-PLGA NPs were administered at gestational day (E)14.5 or 17.5, and cargo biodistribution was analyzed 24 h later by fluorescence imaging of whole excised tissues and histological sections. No gestational differences in DiD distribution were observed, so Cy5-PEG-PLGA NPs were administered at only E17.5 to evaluate polymer distribution in the reproductive organs of pregnant mice. Cy5-PEG-PLGA NPs distributed to the vagina, placentas, and embryos, whereas DiD cargo was only observed in the vagina. NPs did not impact maternal, fetal, or placental weight, suggesting they display no short-term effects on maternal or fetal growth. The results from this study encourage future investigation into the use of vaginally delivered NP therapies for conditions affecting the vagina during pregnancy.


Asunto(s)
Nanopartículas , Ácido Poliglicólico , Embarazo , Humanos , Femenino , Ratones , Animales , Ácido Láctico , Distribución Tisular , Placenta , Polietilenglicoles , Feto , Portadores de Fármacos
15.
Bioeng Transl Med ; 8(3): e10456, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37206243

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are desirable targets for gene therapy but are notoriously difficult to target and transfect. Existing viral vector-based delivery methods are not effective in HSPCs due to their cytotoxicity, limited HSPC uptake and lack of target specificity (tropism). Poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) are attractive, nontoxic carriers that can encapsulate various cargo and enable its controlled release. To engineer PLGA NP tropism for HSPCs, megakaryocyte (Mk) membranes, which possess HSPC-targeting moieties, were extracted and wrapped around PLGA NPs, producing MkNPs. In vitro, fluorophore-labeled MkNPs are internalized by HSPCs within 24 h and were selectively taken up by HSPCs versus other physiologically related cell types. Using membranes from megakaryoblastic CHRF-288 cells containing the same HSPC-targeting moieties as Mks, CHRF-wrapped NPs (CHNPs) loaded with small interfering RNA facilitated efficient RNA interference upon delivery to HSPCs in vitro. HSPC targeting was conserved in vivo, as poly(ethylene glycol)-PLGA NPs wrapped in CHRF membranes specifically targeted and were taken up by murine bone marrow HSPCs following intravenous administration. These findings suggest that MkNPs and CHNPs are effective and promising vehicles for targeted cargo delivery to HSPCs.

16.
Biomater Sci ; 10(16): 4378-4391, 2022 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-35796319

RESUMEN

There is an unmet need for carriers that can deliver nucleic acids (NAs) to cancer cells and tumors to perpetuate gene regulation and manage disease progression. Membrane-wrapped nanoparticles (NPs) can be loaded with exogenously designed nucleic acid cargoes, such as plasmid deoxyribonucleic acid (pDNA), messenger ribonucleic acid (mRNA), small interfering RNA (siRNA), microRNA (miRNA), and immunostimulatory CpG oligodeoxynucleotides (CpG ODNs), to mitigate challenges presented by NAs' undesirable negative charge, hydrophilicity, and relatively large size. By conjugating or encapsulating NAs within membrane-wrapped NPs, various physiological barriers can be overcome so that NAs experience increased blood circulation half-lives and enhanced accumulation in intended sites. This review discusses the status of membrane-wrapped NPs as NA delivery vehicles and their advancement in gene regulation for cancer management in vitro and in vivo. With continued development, membrane-wrapped NPs have great potential as future clinical tools to treat cancer and other diseases with a known genetic basis.


Asunto(s)
MicroARNs , Nanopartículas , Neoplasias , Ácidos Nucleicos , Humanos , MicroARNs/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico
17.
ACS Biomater Sci Eng ; 8(10): 4439-4448, 2022 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-36103274

RESUMEN

This work reports the development of a biomimetic membrane-wrapped nanoparticle (MWNP) platform for targeted chemotherapy of acute myeloid leukemia (AML). Doxorubicin (DOX), a chemotherapeutic used to treat leukemias, lymphomas, and other cancers, was encapsulated in polymeric NPs that were coated with cytoplasmic membranes derived from human AML cells. The release rate of DOX from the MWNPs was characterized under both storage and physiological conditions, with faster release observed at pH 5.5 than pH 7.4. The system was then introduced to AML cell cultures to test the functionality of the released DOX cargo as compared to DOX delivered freely or via NPs coated with poly(ethylene glycol) (PEG). The MWNPs delivered DOX in an efficient and targeted manner, inducing up to 80% apoptosis in treated cells at a dose of 5 µM, compared to 15% for free DOX and 17% for DOX-loaded PEG-coated NPs at the same drug concentration. The mechanism of cell death was confirmed as DNA double-strand breaks through a γH2A.X assay, indicating that the released DOX retained its expected mechanism of action. These findings designate MWNPs as a robust drug delivery system with great potential for future development in treatments of AML and other blood cancers.


Asunto(s)
Leucemia Mieloide Aguda , Nanopartículas , ADN , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Sistemas de Liberación de Medicamentos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Nanopartículas/química , Polietilenglicoles/química
18.
Nano Converg ; 9(1): 37, 2022 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-35960404

RESUMEN

Cancer is a global health problem that needs effective treatment strategies. Conventional treatments for solid-tumor cancers are unsatisfactory because they cause unintended harm to healthy tissues and are susceptible to cancer cell resistance. Nanoparticle-mediated photothermal therapy is a minimally invasive treatment for solid-tumor cancers that has immense promise as a standalone therapy or adjuvant to other treatments like chemotherapy, immunotherapy, or radiotherapy. To maximize the success of photothermal therapy, light-responsive nanoparticles can be camouflaged with cell membranes to endow them with unique biointerfacing capabilities that reduce opsonization, prolong systemic circulation, and improve tumor delivery through enhanced passive accumulation or homotypic targeting. This ensures a sufficient dose of photoresponsive nanoparticles arrives at tumor sites to enable their complete thermal ablation. This review summarizes the state-of-the-art in cell membrane camouflaged nanoparticles for photothermal cancer therapy and provides insights to the path forward for clinical translation.

19.
ACS Omega ; 7(41): 36653-36662, 2022 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-36278042

RESUMEN

Photothermal therapy (PTT) is a promising treatment option for diseases, including cancer, arthritis, and periodontitis. Typical photothermal agents (PTAs) absorb light in the near-infrared (NIR)-I region of 650-900 nm with a predominant focus around 800 nm, as these wavelengths are minimally absorbed by water and blood in the tissue. Recently, interest has grown in developing nanomaterials that offer more efficient photothermal conversion and that can be excited by light close to or within the NIR-II window of 1000-1700 nm, which offers less absorption by melanin. Herein, we report on the development of 5,5-diphenyl isocorrole (5-DPIC) complexes containing either Zn(II) or Pd(II) (Zn[5-DPIC] and Pd[5-DPIC], respectively) that absorb strongly across the 850-1000 nm window. We also show that poly(lactic-co-glycolic acid) (PLGA) nanoparticles loaded with these designer isocorroles exhibit low toxicity toward triple-negative breast cancer (TNBC) cells in the dark but enable efficient heat production and photothermal cell ablation upon excitation with 980 nm light. These materials represent an exciting new platform for 980 nm activated PTT and demonstrate the potential for designer isocorroles to serve as effective PTAs.

20.
Small ; 7(2): 169-83, 2011 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-21213377

RESUMEN

Nanotechnology-based cancer treatment approaches potentially provide localized, targeted therapies that aim to enhance efficacy, reduce side effects, and improve patient quality of life. Gold-nanoparticle-mediated hyperthermia shows particular promise in animal studies, and early clinical testing is currently underway. In this article, the rapidly evolving field of gold nanoparticle thermal therapy is reviewed, highlighting recent literature and describing current challenges to clinical translation of the technology.


Asunto(s)
Oro/química , Hipertermia Inducida/métodos , Nanopartículas del Metal/química , Nanotecnología/métodos , Neoplasias/terapia , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA