Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Prolif ; 57(7): e13622, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38509634

RESUMEN

Neoantigen delivery using extracellular vesicles (EVs) has gained extensive interest in recent years. EVs derived from tumour cells or immune cells have been used to deliver tumour antigens or antitumor stimulation signals. However, potential DNA contamination from the host cell and the cost of large-scale EV production hinder their therapeutic applications in clinical settings. Here, we develop an antigen delivery platform for cancer vaccines from red blood cell-derived EVs (RBCEVs) targeting splenic DEC-205+ dendritic cells (DCs) to boost the antitumor effect. By loading ovalbumin (OVA) protein onto RBCEVs and delivering the protein to DCs, we were able to stimulate and present antigenic OVA peptide onto major histocompatibility complex (MHC) class I, subsequently priming activated antigen-reactive T cells. Importantly, targeted delivery of OVA using RBCEVs engineered with anti-DEC-205 antibody robustly enhanced antigen presentation of DCs and T cell activation. This platform is potentially useful for producing personalised cancer vaccines in clinical settings.


Asunto(s)
Antígenos de Neoplasias , Vacunas contra el Cáncer , Células Dendríticas , Vesículas Extracelulares , Inmunoterapia , Ovalbúmina , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/metabolismo , Animales , Inmunoterapia/métodos , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/administración & dosificación , Ovalbúmina/inmunología , Ovalbúmina/administración & dosificación , Antígenos de Neoplasias/inmunología , Ratones , Presentación de Antígeno/inmunología , Ratones Endogámicos C57BL , Neoplasias/terapia , Neoplasias/inmunología , Humanos , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo
2.
Bioeng Transl Med ; 8(5): e10392, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37693065

RESUMEN

Induction of potent immune responses toward tumors remains challenging in cancer immunotherapy, in which it only showed benefits in a minority of patients with "hot" tumors, which possess pre-existing effector immune cells within the tumor. In this study, we proposed a nanoparticle-based strategy to fire up the "cold" tumor by upregulating the components associated with T and NK cell recruitment and activation and suppressing TGF-ß1 secretion by tumor cells. Specifically, LTX-315, a first-in-class oncolytic cationic peptide, and TGF-ß1 siRNA were co-entrapped in a polymer-lipid hybrid nanoparticle comprising PLGA, DSPE-mPEG, and DSPE-PEG-conjugated with cRGD peptide (LTX/siR-NPs). The LTX/siR-NPs showed significant inhibition of TGF-ß1 expression, induction of type I interferon release, and triggering immunogenic cell death (ICD) in treated tumor cells, indicated via the increased levels of danger molecules, an in vitro setting. The in vivo data showed that the LTX/siR-NPs could effectively protect the LTX-315 peptide from degradation in serum, which highly accumulated in tumor tissue. Consequently, the LTX/siR-NPs robustly suppressed TGF-ß1 production by tumor cells and created an immunologically active tumor with high infiltration of antitumor effector immune cells. As a result, the combination of LTX/siR-NP treatment with NKG2A checkpoint inhibitor therapy remarkably increased numbers of CD8+NKG2D+ and NK1.1+NKG2D+ within tumor masses, and importantly, inhibited the tumor growth and prolonged survival rate of treated mice. Taken together, this study suggests the potential of the LTX/siR-NPs for inflaming the "cold" tumor for potentiating the efficacy of cancer immunotherapy.

3.
J Control Release ; 361: 443-454, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37558053

RESUMEN

Triple-negative breast cancer (TNBC) is highly aggressive and has no standard treatment. Although being considered as an alternative to conventional treatments for TNBC, immunotherapy has to deal with many challenges that hinder its efficacy, particularly the poor immunogenic condition of the tumor microenvironment (TME). Herein, we designed a liposomal nanoparticle (LN) platform that delivers simultaneously toll-like receptor 7 (imiquimod, IQ) and toll-like receptor 3 (poly(I:C), IC) agonists to take advantage of the different toll-like receptor (TLR) signaling pathways, which enhances the condition of TME from a "cold" to a "hot" immunogenic state. The optimized IQ/IC-loaded LN (IQ/IC-LN) was effectively internalized by cancer cells, macrophages, and dendritic cells, followed by the release of the delivered drugs and subsequent stimulation of the TLR3 and TLR7 signaling pathways. This stimulation encouraged the secretion of type I interferon (IFN-α, IFN-ß) and CXCLl0, a T-cell and antigen-presenting cells (APCs) recruitment chemokine, from both cancer cells and macrophages and polarized macrophages to the M1 subtype in in vitro studies. Notably, systemic administration of IQ/IC-LN allowed for the high accumulation of drug content in the tumor, followed by the effective uptake by immune cells in the TME. IQ/IC-LN treatment comprehensively enhanced the immunogenic condition in the TME, which robustly inhibited tumor growth in tumor-bearing mice. Furthermore, synergistic antitumor efficacy was obtained when the IQ/IC-LN-induced immunogenic state in TME was combined with anti-PD1 antibody therapy. Thus, our results suggest the potential of combining 2 TLR agonists to reform the TME from a "cold" to a "hot" state, supporting the therapeutic efficacy of immune checkpoint inhibitors.


Asunto(s)
Receptor Toll-Like 3 , Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Adyuvantes Inmunológicos , Liposomas , Poli I-C/uso terapéutico , Inmunoterapia/métodos , Microambiente Tumoral
4.
Sci Adv ; 8(34): eabn8614, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-36001671

RESUMEN

Immunomodulation is an essential consideration for cell replacement procedures. Unfortunately, lifelong exposure to nonspecific systemic immunosuppression results in immunodeficiency and has toxic effects on nonimmune cells. Here, we engineered hybrid spheroids of mesenchymal stem cells (MSCs) with rapamycin-releasing poly(lactic-co-glycolic acid) microparticles (RAP-MPs) to prevent immune rejection of islet xenografts in diabetic C57BL/6 mice. Hybrid spheroids were rapidly formed by incubating cell-particle mixture in methylcellulose solution while maintaining high cell viability. RAP-MPs were uniformly distributed in hybrid spheroids and sustainably released RAP for ~3 weeks. Locoregional transplantation of hybrid spheroids containing low doses of RAP-MPs (200- to 4000-ng RAP per recipient) significantly prolonged islet survival times and promoted the generation of regional regulatory T cells. Enhanced programmed death-ligand 1 expression by MSCs was found to be responsible for the immunomodulatory performance of hybrid spheroids. Our results suggest that these hybrid spheroids offer a promising platform for the efficient use of MSCs in the transplantation field.


Asunto(s)
Células Madre Mesenquimatosas , Esferoides Celulares , Animales , Humanos , Inmunomodulación , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Trasplante Heterólogo
5.
J Extracell Biol ; 1(7): e49, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38938581

RESUMEN

Blood cell-derived extracellular vesicles (BCEVs) and lipoproteins are the major circulating nanoparticles in blood that play an important role in intercellular communication. They have attracted significant interest for clinical applications, given their endogenous characteristics which make them stable, biocompatible, well tolerated, and capable of permeating biological barriers efficiently. In this review, we describe the basic characteristics of BCEVs and lipoproteins and summarize their implications in both physiological and pathological processes. We also outline well accepted workflows for the isolation and characterization of these circulating nanoparticles. Importantly, we highlight the latest progress and challenges associated with the use of circulating nanoparticles as diagnostic biomarkers and therapeutic interventions in multiple diseases. We spotlight novel engineering approaches and designs to facilitate the development of these nanoparticles by enhancing their stability, targeting capability, and delivery efficiency. Therefore, the present work provides a comprehensive overview of composition, biogenesis, functions, and clinical translation of circulating nanoparticles from the bench to the bedside.

6.
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
7.
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
8.
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
9.
J Control Release ; 336: 274-284, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34144106

RESUMEN

Pancreatic islet replacement therapy is an advanced choice for severe cases of type I diabetes. Nevertheless, extensive host immune response toward islet grafts remains a huge challenge for long-term graft function, and a lack of islet donors further increases the difficulties associated with upscaling this therapy. Mounting evidence suggests local delivery of immunosuppressive agents provides a feasible means of enhancing graft-protection. Among many immunosuppressants, tacrolimus (FK506) is one of the most potent interleukin-2 (IL-2)-mediated T-cell proliferation blockers. Here, we reported the effect of locally-delivered FK506-releasing PLGA microspheres (FK506-M) combined with polyethylene glycol (PEG)-based islet surface modification on xenogeneic islet survival in C57BL/6 mouse model. FK506-M was prepared using an emulsion method to a particle size of 10-40 µm and released FK506 over 40 days in vitro. Around 80% of the initial dose of FK506-M stably localized near transplanted islets, as observed under a bioimaging instrument and by immunofluorescence staining of islet grafts. Interestingly, FK506-M at very low-doses (equivalent to 150 to 2400 ng FK506 per recipient) was found to inhibit the infiltration of immune cells into grafts and reduce serum IL-1ß levels, thereby improving graft survival times dose-dependently. The PEGylation of islets alone was not enough to protect islets from early rejection. However, combined treatment with FK506-M additively prolonged xenograft survival. In conclusion, this study describes a safe, effective approach for translating a systemic exposure-free local drug delivery into clinical trials of islet transplantation.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Animales , Rechazo de Injerto , Supervivencia de Injerto , Inmunosupresores , Ratones , Ratones Endogámicos C57BL , Microesferas , Polietilenglicoles , Tacrolimus
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.
ACS Appl Mater Interfaces ; 13(5): 5999-6010, 2021 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-33506682

RESUMEN

Cellular FLIP (cFLIP) is a crucial player of apoptosis-regulated pathways that is frequently overexpressed in solid cancers. To inhibit c-FLIP, pre- and post-transcriptionally, a multifunctional nanoparticle (NP) was created to deliver cFLIP-specific small interfering RNA (siRNA) into cancer cells. Specifically, Vorinostat (Vor)-loaded mesoporous silica nanoparticles (MSN) were conjugated with polyethylenimine-biotin (PB), followed by electrostatically binding with cFLIP siRNA (Vor/siR@MSN-PB). To stabilize and prolong the circulation time of nanoparticles, a bialdehyde-modified poly(ethylene glycol) (PEG) was cross-linked onto the polyethylenimine (PEI) backbone via the formation of the imine linkage (Schiff base) (Vor/siR@MSN-PB-PEG). The Schiff base is highly stable at physiological pH 7.4 but labile under slightly acidic pH conditions. In the acidic tumor microenvironment (TME), the PEG outer layer could be rapidly cleaved, resulting in the switching of the nanoparticle surface charge to positive, which specifically enhances internalization of the NPs to the biotin-positive tumor cells. Our results demonstrated the successful preparation of Vor/siR@MSN-PB-PEG NPs, in which the siRNA was effectively protected in serum and regulated the expression of cFlip, post-transcriptionally. The presence of the PEG layer resulted in high tumor accumulation and high efficacy in tumor inhibition, which was a result of the efficient cFLIP suppression. Furthermore, in the low-dose regimen of Vorinostat-the pre-transcriptional cFLIP suppressor, treatment with Vor/siR@MSN-PB-PEG NPs was found to be safe with the treated mice, indicating a promising combination regimen for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/antagonistas & inhibidores , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Nanopartículas/química , ARN Interferente Pequeño/farmacología , Vorinostat/farmacología , Animales , Antineoplásicos/química , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Tamaño de la Partícula , Polietilenglicoles/química , ARN Interferente Pequeño/química , Propiedades de Superficie , Vorinostat/química
12.
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
13.
ACS Appl Mater Interfaces ; 12(51): 56767-56781, 2020 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-33289550

RESUMEN

The consolidation of nanovectors with biological membranes has recently been a subject of interest owing to the prolonged systemic circulation time and delayed clearance by the reticuloendothelial system of such systems. Among the different biomembranes, the macrophage membrane has a similar systemic circulation time, with an additional chemotactic aptitude, targeting integrin proteins. In this study, we aimed to establish a laser-activated, disintegrable, and deeply tumor-penetrative nanoplatform. We used a highly tumor-ablative and laser-responsive disintegrable copper sulfide nanoparticle, loaded it with paclitaxel, and camouflaged it with the macrophage membrane for the fabrication of PTX@CuS@MMNPs. The in vitro paclitaxel release profile was favorable for release in the tumor microenvironment, and the release was accelerated after laser exposure. Cellular internalization was improved by membrane encapsulation. Cellular uptake, cytotoxicity, reactive oxygen species generation, and apoptosis induction of PTX@CuS@MMNPs were further improved upon laser exposure, and boosted permeation was achieved by co-administration of the tumor-penetrating peptide iRGD. In vivo tumor accumulation, tumor inhibition rate, and apoptotic marker expression induced by PTX@CuS@MMNPs were significantly improved by laser irradiation and iRGD co-administration. PTX@CuS@MMNPs induced downregulation of cellular proliferation and angiogenic markers but no significant changes in body weight, survival, or significant toxicities in vital organs after laser exposure, suggesting their biocompatibility. The disintegrability of the nanosystem, accredited to biodegradability, favored efficient elimination from the body. In conclusion, PTX@CuS@MMNPs showed promising traits in combination therapies for excellent tumor eradication.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Membrana Celular/química , Macrófagos/química , Nanopartículas del Metal/química , Neoplasias/tratamiento farmacológico , Paclitaxel/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cobre/química , Cobre/efectos de la radiación , Cobre/toxicidad , Portadores de Fármacos/química , Portadores de Fármacos/efectos de la radiación , Portadores de Fármacos/toxicidad , Rayos Infrarrojos , Nanopartículas del Metal/efectos de la radiación , Nanopartículas del Metal/toxicidad , Ratones , Ratones Endogámicos BALB C , Células RAW 264.7
14.
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
15.
Pharm Res ; 37(7): 129, 2020 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-32548664

RESUMEN

PURPOSE: Development of a nanoplatform constructed by the PEG-dual drug conjugation for co-delivery of paclitaxel (PTX) and Dihydroartemisinin (DHA) to the tumor. METHODS: PEG was conjugated with PTX and DHA to form PTX-PEG-DHA complex as a nanocarrier. The PTX and DHA were co-encapsulated in PTX-PEG-DHA nanoparticles (PD@PPD NPs) by the emulsion evaporation method. The physicochemical properties of PD@PPD Nps were characterized, including size, zeta potential, and morphology. The drug loading capacity and entrapment efficiency, in vitro drug release at different pH conditions were also evaluated. For in vitro assessment, the effects of the NPs on HT-29 colorectal cancer cells, including intracellular uptake, cytotoxicity, and Bcl-2 protein expression were assessed. The in vivo distribution of the NPs was investigated by labelling the NPs with Cyanine 5.5 fluorophore. Finally, the antitumor efficacy of the NPs was evaluated in HT-29 tumor-bearing mice. RESULTS: The nanoparticles were formed at small size (~114 nm) and narrow distribution. The combination of PTX and DHA in the DHA-PEG-PTX nanosystems (PD@PPD) showed remarkably increased apoptosis in colorectal adenocarcinoma HT-29 cells, as compared to free drug treatment. More importantly, the PD@PPD nanoparticles exhibited significantly higher accumulation in the tumor site owing to the enhanced permeability and retention (EPR) effect, effectively restrained the tumor growth in vivo at low-dose of PTX while reducing the systemic toxicity. CONCLUSIONS: The combination of PTX and DHA in a PEG-conjugated dual-drug co-delivery system can minimize the severe side effect associated with the high-dose of PTX while enhancing the antitumor efficacy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/química , Artemisininas/química , Neoplasias Colorrectales/tratamiento farmacológico , Nanocápsulas/química , Paclitaxel/química , Polietilenglicoles/química , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Artemisininas/farmacología , Permeabilidad de la Membrana Celular , Composición de Medicamentos , Liberación de Fármacos , Colorantes Fluorescentes/química , Regulación de la Expresión Génica/efectos de los fármacos , Células HT29 , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Imagen Óptica , Paclitaxel/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Distribución Tisular
16.
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
17.
Arch Pharm Res ; 43(1): 32-45, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31993969

RESUMEN

Natural killer (NK) cells have emerged as a potent alternative immunotherapeutic approach to T cell therapy for cancer. Despite promising results from preclinical and clinical studies, numerous challenges have limited the application of NK cell-based therapy, including poor expansion of NK cells in vitro, their short in vivo life span, time-intensiveness, treatment complexities, and the cost burden of the treatment. Recent advancements in the development of immune cell-delivering nanosystems have led to promising strategies to overcome these limitations and enhance NK cell toxicity towards cancer cells. This review first summarizes the biological roles of NK cells and their tumoricidal mechanisms. NK cells, in the context of the immune system and the tumor microenvironment, have reportedly provided novel insights into specific therapeutic targets. Eventually, various strategies targeting NK cells using nanoplatforms to modulate the NK cell responses for effective cancer immunotherapy are described herein. Altogether, this review discusses the potential of nanotechnology in advancements in NK cell-based onco-immunotherapy.


Asunto(s)
Inmunoterapia , Células Asesinas Naturales/inmunología , Nanopartículas/química , Neoplasias/inmunología , Neoplasias/terapia , Animales , Humanos
18.
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
19.
Pharmaceutics ; 11(11)2019 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-31671569

RESUMEN

In this study, we investigated the active targeted delivery of a hydrophobic drug, paclitaxel (PTX), via receptor-mediated endocytosis by folate receptors expressed on cancer cells using a protein-based nanoparticle system. PTX was loaded on zein nanoparticles and conjugated with folate (PTX/Zein-FA) to estimate its chemotherapeutic efficacy in folate receptor-expressing KB cancer cells. PTX/Zein-FA nanoparticles were successfully developed, with a nanoparticle size of ~180 nm and narrow polydispersity index (~0.22). Accelerated release of PTX in an acidic environment was observed for PTX/Zein-FA. An in vitro cellular study of PTX/Zein-FAs in KB cells suggested that PTX/Zein-FA improved the cytotoxic activity of PTX on folate receptors overexpressed in cancer cells by inducing proapoptotic proteins and inhibiting anti-apoptotic proteins. In addition, PTX/Zein-FA exhibited anti-migratory properties and could alter the cell cycle profile of KB cells. A549 cells, which are folate receptor-negative cancer cells, showed no significant enhancement in the in vitro cellular activities of PTX/Zein-FA. We describe the antitumor efficacy of PTX/Zein-FA in KB tumor-bearing mice with minimum toxicity in healthy organs, and the results were confirmed in comparison with free drug and non-targeted nanoparticles.

20.
ACS Appl Mater Interfaces ; 11(40): 36333-36346, 2019 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-31535550

RESUMEN

Cell-based delivery platforms have received great interest in recent years and have been indicated as a promising strategy for cancer immunotherapy. Despite their wide applications in the clinical and preclinical stages, their concomitant viability and efficacy remain major issues. Herein, a strategy for harnessing regulatory T (Treg) cells is developed as an actively targeting drug-delivery system to transport drug-loaded liposomes to the desired tumor sites via conjugating liposomes on the surface of Treg cells. Under the guidance of tumor-oriented chemokines, liposome-anchored Treg cells can be leveraged to migrate and infiltrate the acidic tumor microenvironment, where pH-sensitive liposomes release the loaded cargos [comprising interleukin-2, programmed cell death ligand 1 antibody (PD-L1), and imiquimod], provoke dramatic dendritic cell maturation, block the PD-1/PD-L1 immune-checkpoint, elevate the frequency of infiltrating CD8+ effector T cells, and collectively contribute to potent inhibition of in situ and metastatic tumors. Here, the findings suggest a potential approach that offers a simple, robust, and safe insight into the tuning of Treg cells as an encouraging vector for augmenting cancer immunotherapy.


Asunto(s)
Liposomas/química , Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/inmunología , Animales , Quimiotaxis , Citotoxicidad Inmunológica , Concentración de Iones de Hidrógeno , Inmunoterapia , Neoplasias Pulmonares/secundario , Ratones Endogámicos C57BL , Neoplasias/patología , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...