Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Pharm Educ ; 88(2): 100641, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38185465

RESUMEN

OBJECTIVE: The objective of this review is to provide the conclusions from the American Association of Colleges of Pharmacy (AACP) Council of Deans (COD) Taskforce on Research and Scholarship. FINDINGS: The charges and the findings of the committee are: (1) Define the scholarship needs/opportunities to strengthen the outputs. The committee recommends that AACP update its definitions of research/scholarship to include discovery, integration, application/practice, and teaching/learning. A deployed survey demonstrated a high Special Interest Groups research/scholarship interest. (2) Assemble a toolkit of grant and scholarship resources to assist colleges/schools. The AACP should update the existing funding opportunity listing and combine it with additional resources. (3) Create a framework for effective research collaboration and mentorship. The AACP should identify key areas of pharmacy research and experts to serve as mentors and to meet with external stakeholders. (4) and (5) Consider the need for and purpose of a COD standing committee for research and scholarship. Explore the value of a formal research dean's subcommittee. It was recommended that AACP form a research/scholarship committee or Special Interest Groups and create the Pharmacy Scholarship, Research, and Graduate Education pre-meeting to the Interim Meeting. (6) Identify key statements/outputs of the COD that need to be prepared for publication/sharing. We recommended the key statement/outputs in the areas of discovery, integration, application/practice, and teaching and learning. SUMMARY: The taskforce reviewed the state of research and scholarship across the Academy and provided recommendations with the goal of advancing research across all areas of the pharmacy profession.


Asunto(s)
Educación de Postgrado en Farmacia , Educación en Farmacia , Investigación en Farmacia , Farmacia , Estados Unidos , Humanos , Becas , Facultades de Farmacia
2.
Adv Sci (Weinh) ; 10(14): e2206812, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36949364

RESUMEN

A critical barrier to effective cancer therapy is the improvement of drug selectivity, toxicity, and reduced recurrence of tumors expanded from tumor-initiating stem-like cells (TICs). The aim is to identify circulating tumor cell (CTC)-biomarkers and to identify an effective combination of TIC-specific, repurposed federal drug administration (FDA)-approved drugs. Three different types of high-throughput screens targeting the TIC population are employed: these include a CD133 (+) cell viability screen, a NANOG expression screen, and a drug combination screen. When combined in a refined secondary screening approach that targets Nanog expression with the same FDA-approved drug library, histone deacetylase (HDAC) inhibitor(s) combined with all-trans retinoic acid (ATRA) demonstrate the highest efficacy for inhibition of TIC growth in vitro and in vivo. Addition of immune checkpoint inhibitor further decreases recurrence and extends PDX mouse survival. RNA-seq analysis of TICs reveals that combined drug treatment reduces many Toll-like receptors (TLR) and stemness genes through repression of the lncRNA MIR22HG. This downregulation induces PTEN and TET2, leading to loss of the self-renewal property of TICs. Thus, CTC biomarker analysis would predict the prognosis and therapy response to this drug combination. In general, biomarker-guided stratification of HCC patients and TIC-targeted therapy should eradicate TICs to extend HCC patient survival.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Células Neoplásicas Circulantes , Ratones , Animales , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Línea Celular Tumoral , Tretinoina/uso terapéutico
3.
Mater Today Bio ; 19: 100567, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36747581

RESUMEN

Engineered mesenchymal stem cells (MSCs) have been investigated extensively for gene delivery and, more recently, for targeted small molecule delivery. While preclinical studies demonstrate the potential of MSCs for targeted delivery, clinical studies suggest that tumor homing of native MSCs may be inefficient. We report here a surprising finding that loading MSCs with the anticancer drug paclitaxel (PTX) by nanoengineering results in significantly improved tumor homing compared to naïve MSCs. Loading PTX in MSCs results in increased levels of mitochondrial reactive oxygen species (ROS). In response to this oxidative stress, MSCs upregulate two important set of proteins. First were critical antioxidant proteins, most importantly nuclear factor erythroid 2-like 2 (Nrf2), the master regulator of antioxidant responses; upregulation of antioxidant proteins may explain how MSCs protect themselves from drug-induced oxidative stress. The second was CXCR4, a direct target of Nrf2 and a key mediator of tumor homing; upregulation of CXCR4 suggested a mechanism that may underlie the improved tumor homing of nanoengineered MSCs. In addition to demonstrating the potential mechanism of improved tumor targeting of nanoengineered MSCs, our studies reveal that MSCs utilize a novel mechanism of resistance against drug-induced oxidative stress and cell death, explaining how MSCs can deliver therapeutic concentrations of cytotoxic payload while maintaining their viability.

4.
Cancers (Basel) ; 14(24)2022 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-36551577

RESUMEN

Immunostimulatory adjuvants that potently activate antigen-presenting cells and (in turn) prime cytotoxic T cells are a key component of anticancer vaccines. In this study, we investigated a multi-adjuvant approach combining a TLR 7/8 agonist (522) and a STING agonist (DMXAA) to promote enhanced antigen cross-presentation, stimulate specific antitumor T-cell responses, and provide improved anticancer efficacy. In vitro experiments using bone marrow-derived dendritic cells (BMDCs) confirmed enhanced activation with the 522-DMXAA combination based on both co-stimulatory molecule expression and pro-inflammatory cytokine secretion. The immunization of mice with vaccines comprising both 522 and DMXAA resulted in greater antitumor efficacy in B16F10 melanoma and MB49 bladder tumor models relative to mono-agonist vaccines. Flow cytometry-based analysis of immune cells from immunized mice revealed the significant activation of antigen-presenting cells, increased numbers of activated and Ag-specific CD8+ T cells in the spleen and lymph nodes, modest NK cell activation, and an overall reduction in CD206+ macrophages. These results were supported by an increase in the levels of IFN-γ and a reduction in IL-10 levels in the sera. Taken together, these findings demonstrate the potential of the TLR7/8 and STING agonist combination as vaccine adjuvants to activate both innate and adaptive immune responses.

5.
Front Bioeng Biotechnol ; 9: 639409, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33681172

RESUMEN

Andrographolide is a potential chemopreventive and chemotherapeutic agent that suffers from poor aqueous solubility. Encapsulation in poly(lactide-co-glycolide) (PLGA) nanoparticles can overcome solubility issues and enable sustained release of the drug, resulting in improved therapeutic efficacy. In this study, andrographolide was encapsulated in PLGA nanoparticles via emulsion solvent evaporation technique. Effect of various formulation parameters including polymer composition, polymer molecular weight, polymer to drug ratio, surfactant concentration and the organic solvent used on nanoparticle properties were investigated. A selected formulation was used to determine the effect of encapsulation in nanoparticles on andrographolide's in vitro anticancer efficacy. Nanoparticles formulated using a polymer with 85:15 lactide to glycolide ratio and ethyl acetate as the organic solvent were found to be optimal based on average hydrodynamic particle size (135 ± 4 nm) and drug loading (2.6 ± 0.6%w/w). This formulation demonstrated sustained release of andrographolide over 48 h and demonstrated significantly greater in vitro anticancer efficacy compared to free drug in a metastatic breast cancer cell line. These results suggest that additional, more in-depth efficacy studies are warranted for the nanoparticle formulation of andrographolide.

6.
Sci Rep ; 11(1): 3346, 2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33558639

RESUMEN

There is a significant interest in designing therapeutic agents that can enhance ADCC and thereby improve clinical responses with approved antibodies. We recently reported the combination of an imidazoquinoline-based TLR7/8 agonist (522) with a monoclonal antibody improved ADCC in vitro and in vivo. In the present study, we tested several new small molecule TLR7/8 agonists that induce significantly higher cytokines compared to both the FDA-approved TLR7 agonist, imiquimod, and 522. We evaluated these agonists in combination with monoclonal antibody therapy, with the main goal of enhancing ADCC. Our studies show these TLR7/8 agonists induce robust pro-inflammatory cytokine secretion and activate NK cells. Specifically, we found the agonists 574 and 558 significantly enhanced NK cell-mediated ADCC in vitro as well as enhanced the anti-cancer efficacy of monoclonal antibodies in two different in vivo mouse models. Additionally, we found the agonists were able to stimulate CD8 T cells, likely indicative of an early adaptive immune response.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Antineoplásicos Inmunológicos/farmacología , Linfocitos T CD8-positivos/inmunología , Imiquimod/farmacología , Células Asesinas Naturales/inmunología , Neoplasias Experimentales , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Células A549 , Animales , Humanos , Células Asesinas Naturales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 8/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Drug Deliv Transl Res ; 11(5): 1779-1805, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33095384

RESUMEN

Cancer stem cells (CSCs) are a small proportion of cancer cells with high tumorigenic activity, self-renewal ability, and multilineage differentiation potential. Standard anti-tumor therapies including conventional chemotherapy, radiation therapy, and molecularly targeted therapies are not effective against CSCs, and often lead to enrichment of CSCs that can result in tumor relapse. Therefore, it is hypothesized that targeting CSCs is key to increasing the efficacy of cancer therapies. In this review, CSC properties including CSC markers, their role in tumor growth, invasiveness, metastasis, and drug resistance, as well as CSC microenvironment are discussed. Further, CSC-targeted strategies including the use of targeted drug delivery systems are examined.


Asunto(s)
Antineoplásicos , Neoplasias , Sistemas de Liberación de Medicamentos , Humanos , Terapia Molecular Dirigida , Neoplasias/patología , Células Madre Neoplásicas/patología , Microambiente Tumoral
8.
Mol Pharm ; 17(6): 2109-2124, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32383885

RESUMEN

Activated natural killer (NK) cells can kill malignant tumor cells via granule exocytosis and secretion of IFN-γ, a key regulator of the TH1 response. Thus, mobilization of NK cells can augment cancer immunotherapy, particularly when mediated through antibody-dependent cellular cytotoxicity (ADCC). Stimulation of toll-like receptor (TLR)7/8 activity in dendritic cells promotes pro-inflammatory cytokine secretion and costimulatory molecule upregulation, both of which can potentiate NK cell activation. However, currently available TLR7/8 agonists exhibit unfavorable pharmacokinetics, limiting their in vivo efficacy. To enable efficient delivery to antigen-presenting cells, we encapsulated a novel imidazoquinoline-based TLR7/8 agonist in pH-responsive polymeric NPs. Enhanced costimulatory molecule expression on dendritic cells and a stronger pro-inflammatory cytokine response were observed with a NP-encapsulated agonist, compared to that with the soluble form. Treatment with NP-encapsulated agonists resulted in stronger in vivo cytotoxicity and prolonged activation of NK cells compared to that with a soluble agonist. In addition, TLR7/8 agonist-loaded NPs potentiated stronger NK cell degranulation, which resulted in enhanced in vitro and in vivo ADCC mediated by the epidermal growth factor receptor-targeting antibody cetuximab. TLR7/8 agonist-loaded NP treatment significantly enhanced the antitumor efficacy of cetuximab and an anti-HER2/neu antibody in mouse tumor models. Collectively, our data show that a pH-responsive NP-encapsulating TLR7/8 agonist could be used as a potent immunostimulatory adjuvant for antibody-based cancer immunotherapy by promoting NK cell activation.


Asunto(s)
Inmunoterapia/métodos , Células Asesinas Naturales/metabolismo , Nanopartículas/química , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Células A549 , Animales , Células Dendríticas/metabolismo , Citometría de Flujo , Humanos , Imiquimod/química , Células Asesinas Naturales/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nanomedicina/métodos
9.
Mol Carcinog ; 59(8): 886-896, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32291806

RESUMEN

Inhibiting the disease progression in KRAS-driven cancers after diagnosis has been a difficult task for clinicians to manage due to the lack of effective intervention/preventive therapies. KRAS-driven cancers depend on sustained KRAS signaling. Although developing inhibitors of KRAS signaling has proven difficult in the past, the quest for identifying newer agents has not stopped. Based on studies showing terpenoids as modulators of KRAS-regulated downstream molecular pathways, we asked if this chemical family has an affinity of inhibiting KRAS protein activity. Using crystal structure as a bait in silico, we identified 20 terpenoids for their KRAS protein-binding affinity. We next carried out biological validation of in silico data by employing in situ, in vitro, patient-derived explant ex vivo, and KPC transgenic mouse models. In this report, we provide a comprehensive analysis of a lup-20(29)-en-3b-ol (lupeol) as a KRAS inhibitor. Using nucleotide exchange, isothermal titration calorimetry, differential scanning fluorimetry, and immunoprecipitation assays, we show that lupeol has the potential to reduce the guanosine diphosphate/guanosine triphosphate exchange of KRAS protein including mutant KRASG12V . Lupeol treatment inhibited the KRAS activation in KRAS-activated cell models (NIH-panel, colorectal, lung, and pancreatic intraepithelial neoplasia) and patient tumor explants ex vivo. Lupeol reduced the three-dimensional growth of KRAS-activated cells. The pharmacokinetic analysis showed the bioavailability of lupeol after consumption via oral and intraperitoneal routes in animals. Tested under prevention settings, the lupeol consumption inhibited the development of pancreatic intraepithelial neoplasia in LSL-KRASG12D/Pdx-cre mice (pancreatic ductal adenocarcinoma progression model). These data suggest that the selected members of the triterpene family (such as lupeol) could be exploited as clinical agents for preventing the disease progression in KRAS-driven cancers which however warrants further investigation.


Asunto(s)
Antiinflamatorios/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Modelos Animales de Enfermedad , Neoplasias Pancreáticas/tratamiento farmacológico , Triterpenos Pentacíclicos/farmacología , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Animales , Apoptosis , Proliferación Celular , Transformación Celular Neoplásica/patología , Progresión de la Enfermedad , Femenino , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
10.
Immunotherapy ; 12(4): 255-267, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32208784

RESUMEN

Over the last decade, antibodies have become an important component in the arsenal of cancer therapeutics. High-specificity, low off-target effects, desirable pharmacokinetics and high success rate are a few of the many attributes that make antibodies amenable for development as drugs. To design antibodies for successful clinical applications, however, it is critical to have an understanding of their structure, functions, mechanisms of action and pharmacokinetic/pharmacodynamic properties. This review highlights some of these key aspects, as well as certain limitations encountered, with monoclonal antibody therapy. Further, we discuss rational combination therapies for clinical applications, some of which could help overcome the limitations.


Asunto(s)
Anticuerpos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Anticuerpos/química , Anticuerpos/clasificación , Anticuerpos/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos de Neoplasias/inmunología , Antineoplásicos Inmunológicos/inmunología , Quimioterapia Combinada , Humanos , Inhibidores de Puntos de Control Inmunológico , Neoplasias/inmunología
11.
Future Drug Discov ; 1(1): FDD8, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31448368

RESUMEN

AIM: We previously developed two antibodies that bind to a cell surface protein, perlecan, overexpressed in triple-negative breast cancer (TNBC). The goal of this study was to investigate these antibodies as targeting ligands for nanoparticle-mediated drug delivery. METHODS: Paclitaxel-loaded poly(D,L-lactide-co-glycolide) nanoparticles were functionalized with antibodies using thiol-maleimide chemistry. Effect of antibody functionalization on therapeutic efficacy of drug-loaded nanoparticles was investigated using in vitro and in vivo models of TNBC. RESULTS: The antibodies were covalently conjugated to nanoparticles without affecting antibody binding affinity or nanoparticle properties. Perlecan-targeted nanoparticles showed improved cell uptake, retention, cytotoxicity in vitro and enhanced tumor growth inhibition in vivo. CONCLUSION: The data presented here indicates that perlecan-targeted nanoparticles can improve tumor drug delivery to TNBC.

12.
Sci Rep ; 9(1): 12492, 2019 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-31462656

RESUMEN

In recent years, there have been significant advances in the treatment of breast cancer resulting in remarkably high survival rates. However, treatment options for metastatic triple negative breast cancer (TNBC) are quite limited due to a lack of identifiable, unique markers. Using a phage display-based whole cell biopanning procedure, we developed two human antibodies that bind to tumor cells with a metastatic TNBC phenotype. Our studies further identified domain 1 of HSPG2 (perlecan) protein as the cognate cell surface antigen bound by the antibody. Immunohistochemistry studies utilizing patient tissue samples revealed significant cell surface expression of HSPG2 in both primary tumors and metastatic lesions. Further, higher HSPG2 expression correlated with poor survival in TNBC. The affinity-matured antibody inhibited the growth of triple negative MDA-MB-231 tumors to a greater extent in nude mice than in NSG mice, pointing to the potential role of natural killer cell-mediated antibody-dependent cell cytotoxicity. This mechanism of action was confirmed through in vitro assays using mouse splenocytes and human peripheral blood mononuclear cells (PBMCs). These results suggest that HSPG2 is a promising target in metastatic TNBC and HSPG2-targeted antibodies could represent a potentially novel class of targeted therapeutics for TNBC.


Asunto(s)
Antineoplásicos Inmunológicos , Proteoglicanos de Heparán Sulfato , Proteínas de Neoplasias , Neoplasias de la Mama Triple Negativas , Adulto , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacología , Línea Celular Tumoral , Femenino , Proteoglicanos de Heparán Sulfato/genética , Proteoglicanos de Heparán Sulfato/inmunología , Humanos , Ratones Desnudos , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
FASEB J ; 33(8): 9453-9465, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31136203

RESUMEN

Oral consumption of curcumin, a natural polyphenol, is associated with reduced incidence of cancer. Yet, a significant amount of the orally dosed compound is eliminated in the feces, and a major fraction of the absorbed compound is metabolized to inactive glucuronides, resulting in poor bioavailability (<1%). It is not known how oral curcumin exhibits chemopreventive activity. We propose curcumin glucuronide is an inflammation-responsive natural prodrug that is converted back to curcumin on demand at the site of action. Our studies show elevated levels of ß-glucuronidase, an enzyme that hydrolyzes the glycosidic bond of glucuronides to generate the parent compound, in human breast cancer. Oral administration of curcumin in mouse tumor models generated significant tumor levels of the polyphenol. Intravenous administration of the glucuronide resulted in the formation of curcumin in the tumor tissue. Chronic daily oral curcumin dosing led to tumor accumulation of curcumin and inhibition of tumor growth in tumor models with high ß-glucuronidase activity. Overall, the study presented here provides preliminary evidence for a novel mechanism of action for orally administered curcumin.-Liu, G., Khanna, V., Kirtane, A., Grill, A., Panyam, J. Chemopreventive efficacy of oral curcumin: a prodrug hypothesis.


Asunto(s)
Curcumina/análogos & derivados , Curcumina/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Glucurónidos/farmacocinética , Administración Oral , Animales , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Curcumina/administración & dosificación , Curcumina/uso terapéutico , Femenino , Glucuronidasa/metabolismo , Glucurónidos/administración & dosificación , Glucurónidos/uso terapéutico , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Profármacos/administración & dosificación , Profármacos/farmacocinética , Profármacos/uso terapéutico
14.
Cancers (Basel) ; 11(4)2019 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-30959908

RESUMEN

Mesenchymal stem cells (MSCs) accumulate specifically in both primary tumors and metastases following systemic administration. However, the poor payload capacity of MSCs limits their use in small molecule drug delivery. To improve drug payload in MSCs, we explored polymeric nanoparticles that were functionalized with transactivator of transcription (TAT) peptide. Paclitaxel loaded poly(DL-lactide-co-glycolide) (PLGA) nanoparticles (15⁻16% w/w paclitaxel; diameter of 225 ± 7 nm; and zeta potential of -15 ± 4 mV) were fabricated by emulsion-solvent evaporation method, followed by TAT-conjugation to the surface of nanoparticles via maleimide-thiol chemistry. Our studies demonstrated that TAT functionalization improved the intracellular accumulation and retention of nanoparticles in MSCs. Further, nano-engineering of MSCs did not alter the migration and differentiation potential of MSCs. Treatment with nano-engineered MSCs resulted in significant (p < 0.05) inhibition of tumor growth and improved survival (p < 0.0001) in a mouse orthotopic model of lung cancer compared to that with free or nanoparticle encapsulated drug. In summary, our results demonstrated that MSCs engineered using TAT functionalized nanoparticles serve as an efficient carrier for tumor specific delivery of anticancer drugs, resulting in greatly improved therapeutic efficacy.

15.
Mol Pharm ; 16(3): 1200-1210, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30620878

RESUMEN

Cancer vaccines composed of tumor-associated antigens (TAAs) and toll-like receptor (TLR) agonists have shown promising antitumor efficacy in preclinical studies by generating antigen-specific CD8 T cells, but translation of cancer vaccines to the clinic has been limited due to variables responses and development of resistance. The tumor microenvironment deploys various immune escape mechanisms that neutralize CD8 T cell-mediated tumor rejection. Therefore, we hypothesized that modulation of the tumor microenvironment can augment CD8 T cell activation and enhance therapeutic efficacy of cancer vaccines. To accomplish this, we aimed to eliminate immune suppressive cells and block their inhibitory signaling. Combination of the tyrosine kinase inhibitor (TKI) sunitinib with a nanoparticle-based cancer vaccine (nanovaccine) resulted in the reduction of immune-suppressive myeloid-derived suppressive cells (MDSCs) and regulatory T cells (Tregs). Blockade of programmed death-ligand 1 (PD-L1) using anti-PD-L1 antibody was used to reduce CD8 T cell exhaustion. Combination of nanovaccine+sunitinib+PD-L1 antibody treatment reduced PD-L1high M2 macrophages and MDSCs and upregulated activation of CD8 T cells in the tumor. Nanovaccine+sunitinib+PD-L1 antibody treatment also stimulated antigen-specific CD8 T cell response, which led to improved therapeutic efficacy in MB49 and B16F10 murine tumor models. These results suggest that modulation of tumor microenvironment using sunitinib and PD-L1 blockade can significantly enhance the antitumor efficacy of cancer nanovaccine.


Asunto(s)
Anticuerpos/uso terapéutico , Antígeno B7-H1/inmunología , Vacunas contra el Cáncer/uso terapéutico , Glicoproteínas de Membrana/agonistas , Neoplasias/terapia , Sunitinib/uso terapéutico , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Animales , Antígenos de Neoplasias/inmunología , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Femenino , Interleucina-10/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , Linfocitos T Reguladores/inmunología , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos , Vacunación
16.
J Pharmacol Exp Ther ; 370(3): 715-724, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30610006

RESUMEN

Targeted drug delivery can significantly influence the efficacy of a drug. In the past decades, diverse drug-delivery technologies, including nano- and microparticles, co-crystals, and microneedles have been developed to maximize therapeutic efficacy and minimize undesired side effects of therapeutics. Nanoparticles-submicron-sized drug carriers-have been actively investigated for the delivery of antibiotics, nucleic acids, peptide/proteins, and chemotherapeutics. Recently, nanoparticles have gained attention as a vaccine delivery platform for tumor-associated antigens (TAAs) and/or vaccine adjuvants. Agonists of imidazoquinoline-based Toll-like receptor (TLR) 7/8 are potent cytokine inducers that are used as cancer vaccine adjuvants to elicit robust T-cell response by activating dendritic cells (DCs). Despite their in vitro potency, the translation of TLR7 agonists as cancer vaccine adjuvants in the clinic has been limited by their poor retention at the injection site. Therefore, a formulation that could improve the availability of TLR7/8 agonists to DCs via conventional vaccine administration routes (subcutaneous, intramuscular) can broaden the application of TLR7/8 agonists for cancer immunotherapy. Polymeric nanoparticles fabricated with poly(d,l-lactide-co-glycolide) (PLGA) can be an efficient TLR7/8 agonist delivery platform. PLGA is a biocompatible polymer, and nanoparticles prepared from this polymer are stable in saline and are small enough to be administered by subcutaneous or intramuscular injections. Furthermore, nanoparticulate TLR7/8 delivery can enhance DC uptake and facilitate lymphatic drainage, both of which can enhance the adjuvanticity of TLR7/8 agonists compared with soluble forms. In this review, we discuss the use of PLGA nanoparticles with TLR7/8 agonists for improving cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Animales , Vacunas contra el Cáncer/uso terapéutico , Humanos
17.
J Control Release ; 294: 268-278, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-30572036

RESUMEN

Hollow microneedles can help overcome the skin permeation barrier imposed by the stratum corneum and facilitate transcutaneous delivery of nanoparticle delivery systems. In the present study, we investigated the use of the hollow microneedle array for intradermal delivery of polymeric nanoparticles (NPs) in rats. Compared to intravenous and subcutaneous routes of administration, intradermal delivery of polymeric NPs via a hollow microneedle array resulted in a unique pharmacokinetic profile, characterized by an early burst transit through the draining lymph nodes and a relatively limited overall systemic exposure. Based on high local lymphatic concentrations achieved, we investigated the use of this modality for vaccine delivery. A model antigen ovalbumin (OVA) and TLR agonists imiquimod and monophosphoryl Lipid A encapsulated in poly(d,l-lactide-co-glycolide) (PLGA) NPs were used as the vaccine formulation. Compared to soluble OVA-based vaccine, OVA loaded NPs demonstrated faster antibody affinity maturation kinetics. Moreover, antigen loaded NPs delivered via a hollow microneedle array elicited a significantly higher IgG2a antibody response and higher number of interferon (IFN)-γ secreting lymphocytes, both markers of Th1 response, in comparison to antigen loaded NPs delivered by intramuscular injection and soluble antigen delivered through hollow microneedle array. Overall, our results show that hollow microneedle mediated intradermal delivery of polymeric NPs is a promising approach to improve the effectiveness of vaccine formulations.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antígenos/administración & dosificación , Nanopartículas/administración & dosificación , Ovalbúmina/administración & dosificación , Vacunas/administración & dosificación , Animales , Preparaciones de Acción Retardada/administración & dosificación , Células Dendríticas/inmunología , Imiquimod/administración & dosificación , Inmunoglobulina G/inmunología , Inyecciones Intradérmicas , Lípido A/administración & dosificación , Lípido A/análogos & derivados , Ganglios Linfáticos/metabolismo , Ratones Endogámicos C57BL , Microinyecciones , Agujas , Ratas , Receptor Toll-Like 7/agonistas
18.
Nanoscale ; 10(44): 20851-20862, 2018 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-30403212

RESUMEN

Synthetic imidazoquinoline-based toll-like receptor (TLR) 7/8 bi-specific agonists are promising vaccine adjuvants that can induce maturation of dendritic cells (DCs) and activate them to secrete pro-inflammatory cytokines. However, in vivo efficacy of these small molecule agonists is often hampered by their fast clearance from the injection site, limiting their use to topical treatments. In this study, we investigated the use of acidic pH-responsive poly(lactide-co-glycolide) (PLGA) nanoparticles for endo-lysosome specific release of 522, a novel TLR7/8 agonist. Bicarbonate salt was incorporated into the new formulation to generate carbon dioxide (CO2) gas at acidic pH, which can disrupt the polymer shell to rapidly release the payload. Compared to conventional PLGA nanoparticles, the pH responsive formulation resulted in 33-fold higher loading of 522. The new formulation demonstrated acid-responsive CO2 gas generation and drug release. The acid-responsive formulation increased the in vitro expression of co-stimulatory molecules on DCs and improved antigen-presentation via MHC I, both of which are essential for CD8 T cell priming. In vivo studies showed that the pH-responsive formulation elicited stronger antigen-specific CD8 T cell and natural killer (NK) cell responses than conventional PLGA nanoparticles, resulting in enhanced anticancer efficacy in a murine melanoma tumor model. Our results suggest that acidic-pH responsive, gas-generating nanoparticles are an efficient TLR7/8 agonist delivery platform for cancer immunotherapy.


Asunto(s)
Portadores de Fármacos/química , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Animales , Presentación de Antígeno , Células de la Médula Ósea/citología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Dióxido de Carbono/metabolismo , Línea Celular Tumoral , Microscopía por Crioelectrón , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Concentración de Iones de Hidrógeno , Inmunoterapia , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Nanopartículas/metabolismo , Nanopartículas/toxicidad , Neoplasias/terapia , Ovalbúmina/inmunología
19.
Mini Rev Med Chem ; 18(16): 1363-1372, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29692244

RESUMEN

Chlamydia trachomatis is responsible for the most frequent sexually transmitted bacterial infection in the world and for trachoma, the world's leading infectious cause of blindness. Genital chlamydial infection is very common among sexually active young people, and when untreated, leads to serious complications. No vaccine is yet available for this bacterial infection. Although Chlamydia resistance to antibiotics is rarely observed in vivo, studies showed that 10-20% of patients remain infected at the end of antibiotherapy, without being reinfected. The present review gives a global and comprehensive overview of the different targets and the related inhibitors proposed during the last decade, with a view to limiting the growth of this human pathogen. Metallic and polymeric nanoparticles in this field are also briefly presented.


Asunto(s)
Antibacterianos/farmacología , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/efectos de los fármacos , Enfermedades de Transmisión Sexual/microbiología , Antibacterianos/uso terapéutico , Infecciones por Chlamydia/tratamiento farmacológico , Chlamydia trachomatis/crecimiento & desarrollo , Farmacorresistencia Bacteriana , Humanos , Nanopartículas , Enfermedades de Transmisión Sexual/tratamiento farmacológico
20.
Mol Cancer Ther ; 17(6): 1196-1206, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29592881

RESUMEN

Tumor-targeted drug delivery has the potential to improve therapeutic efficacy and mitigate non-specific toxicity of anticancer drugs. However, current drug delivery approaches rely on inefficient passive accumulation of the drug carrier in the tumor. We have developed a unique, truly active tumor-targeting strategy that relies on engineering mesenchymal stem cells (MSC) with drug-loaded nanoparticles. Our studies using the A549 orthotopic lung tumor model show that nano-engineered MSCs carrying the anticancer drug paclitaxel (PTX) home to tumors and create cellular drug depots that release the drug payload over several days. Despite significantly lower doses of PTX, nano-engineered MSCs resulted in significant inhibition of tumor growth and superior survival. Anticancer efficacy of nano-engineered MSCs was confirmed in immunocompetent C57BL/6 albino female mice bearing orthotopic Lewis Lung Carcinoma (LL/2-luc) tumors. Furthermore, at doses that resulted in equivalent therapeutic efficacy, nano-engineered MSCs had no effect on white blood cell count, whereas PTX solution and PTX nanoparticle treatments caused leukopenia. Biodistribution studies showed that nano-engineered MSCs resulted in greater than 9-fold higher AUClung of PTX (1.5 µg.day/g) than PTX solution and nanoparticles (0.2 and 0.1 µg.day/g tissue, respectively) in the target lung tumors. Furthermore, the lung-to-liver and the lung-to-spleen ratios of PTX were several folds higher for nano-engineered MSCs relative to those for PTX solution and nanoparticle groups, suggesting that nano-engineered MSCs demonstrate significantly less off-target deposition. In summary, our results demonstrate that nano-engineered MSCs can serve as an efficient carrier for tumor-specific drug delivery and significantly improved anti-cancer efficacy of conventional chemotherapeutic drugs. Mol Cancer Ther; 17(6); 1196-206. ©2018 AACR.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Nanomedicina Teranóstica/métodos , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Biomarcadores de Tumor , Línea Celular Tumoral , Modelos Animales de Enfermedad , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Ratones , Nanopartículas/química , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Paclitaxel/administración & dosificación , Paclitaxel/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...