Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Anal Chem ; 96(6): 2286-2291, 2024 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-38289025

RESUMEN

The link between inflammation and the evolution of cancer is well established. Visualizing and tracking both tumor proliferation and the associated inflammatory response within a living organism are vital for dissecting the nexus between these two processes and for crafting precise treatment modalities. We report the creation and synthesis of an advanced NIR chemiluminescence probe that stands out for its exceptional selectivity, extraordinary sensitivity at nanomolar concentrations, swift detection capabilities, and broad application prospects. Crucially, this probe has been successfully utilized to image endogenous ONOO- across different inflammation models, including abdominal inflammation triggered by LPS, subcutaneous inflammatory conditions, and tumors grafted onto mice. These findings highlight the significant promise of chemiluminescence imaging in enhancing our grasp of the intricate interplay between cancer and inflammation and in steering the development of potent, targeted therapeutic strategies.


Asunto(s)
Inflamación , Neoplasias , Animales , Ratones , Inflamación/diagnóstico por imagen , Luminiscencia , Neoplasias/diagnóstico por imagen , Colorantes Fluorescentes , Ácido Peroxinitroso
2.
Liver Int ; 44(8): 1808-1831, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38700443

RESUMEN

Hepatocellular carcinoma (HCC), one of the most prevalent and destructive causes of cancer-related deaths worldwide, approximately 70% of patients with HCC exhibit advanced disease at diagnosis, limiting the potential for radical treatment. For such patients, lenvatinib, a long-awaited alternative to sorafenib for first-line targeted therapy, has become a key treatment. Unfortunately, despite some progress, the prognosis for advanced HCC remains poor because of drug resistance development. However, the molecular mechanisms underlying lenvatinib resistance and ways to relief drug resistance in HCC are largely unknown and lack of systematic summary; thus, this review not only aims to explore factors contributing to lenvatinib resistance in HCC, but more importantly, summary potential methods to conquer or mitigate the resistance. The results suggest that abnormal activation of pathways, drug transport, epigenetics, tumour microenvironment, cancer stem cells, regulated cell death, epithelial-mesenchymal transition, and other mechanisms are involved in the development of lenvatinib resistance in HCC and subsequent HCC progression. To improve the therapeutic outcomes of lenvatinib, inhibiting acquired resistance, combined therapies, and nano-delivery carriers may be possible approaches.


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Resistencia a Antineoplásicos , Neoplasias Hepáticas , Compuestos de Fenilurea , Quinolinas , Carcinoma Hepatocelular/tratamiento farmacológico , Humanos , Quinolinas/uso terapéutico , Quinolinas/farmacología , Compuestos de Fenilurea/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Antineoplásicos/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico
3.
Small ; 19(30): e2300280, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37060227

RESUMEN

Anticancer immunotherapy is hampered by the poor tumor immunogenicity and immunosuppressive tumor microenvironment (TME). Herein, a liposome nanodrug co-encapsulating doxycycline hydrochloride (Doxy) and chlorin e6 (Ce6) to simultaneously induce autophagy inhibition and mitochondria dysfunction for potentiating tumor photo-immunotherapy is developed. Under near infrared laser irradiation, Ce6 generates cytotoxic reactive oxygen species (ROS) and elicits robust photodynamic therapy (PDT)-induced immunogenic cell death (ICD) for immunosuppressive TME remodeling. In addition, Doxy induced mitochondria dysfunction, which increases ROS generation and enhances PDT to exert more potent killing effect and more powerful ICD. Meanwhile, Doxy increases MHC-I expression on tumor cells surface by efficient autophagy inhibition, leading to more efficient antigen presentation and CTLs recognition to increase tumor immunogenicity. The nanodrugs elicit remarkable antitumor therapy by combining Ce6-mediated PDT and Doxy-induced autophagy inhibition and mitochondria dysfunction. The developed nanodrugs represent a highly efficient strategy for improving cancer immunotherapy.


Asunto(s)
Nanopartículas , Fotoquimioterapia , Porfirinas , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Inmunoterapia , Nanopartículas/uso terapéutico , Autofagia , Línea Celular Tumoral , Porfirinas/farmacología , Porfirinas/uso terapéutico , Microambiente Tumoral
4.
Small ; 18(15): e2107732, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35218310

RESUMEN

Immunotherapy brings great benefits for tumor therapy in clinical treatments but encounters the severe challenge of low response rate mainly because of the immunosuppressive tumor microenvironment. Multifunctional nanoplatforms integrating effective drug delivery and medical imaging offer tremendous potential for cancer treatment, which may play a critical role in combinational immunotherapy to overcome the immunosuppressive microenvironment for efficient tumor therapy. Here, a nanodrug (BMS-SNAP-MOF) is prepared using glutathione (GSH)-sensitive metal-organic framework (MOF) to encapsulate an immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO) inhibitor BMS-986205, and the nitric oxide (NO) donor s-nitrosothiol groups. The high T1 relaxivity allows magnetic resonance imaging to monitor nanodrug distribution in vivo. After the nanodrug accumulation in tumor tissue via the EPR effect and subsequent internalization into tumor cells, the enriched GSH therein triggers cascade reactions with MOF, which disassembles the nanodrug to rapidly release the IDO-inhibitory BMS-986205 and produces abundant NO. Consequently, the IDO inhibitor and NO synergistically modulate the immunosuppressive tumor microenvironment with increase CD8+ T cells and reduce Treg cells to result in highly effective immunotherapy. In an animal study, treatment using this theranostic nanodrug achieves obvious regressions of both primary and distant 4T1 tumors, highlighting its application potential in advanced tumor immunotherapy.


Asunto(s)
Estructuras Metalorgánicas , Animales , Linfocitos T CD8-positivos , Inhibidores Enzimáticos , Glutatión , Inmunoterapia/métodos , Indolamina-Pirrol 2,3,-Dioxigenasa , Óxido Nítrico , Microambiente Tumoral
5.
Nano Lett ; 21(7): 3225-3236, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33764789

RESUMEN

Developing strategies for efficient expansion of cancer stem-like cells (CSCs) in vitro will help investigate the mechanism underlying tumorigenesis and cancer recurrence. Herein, we report a dynamic culture substrate tethered with integrin ligand-bearing magnetic nanoparticles via a flexible polymeric linker to enable magnetic manipulation of the nanoscale ligand tether mobility. The cancer cells cultured on the substrate with high ligand tether mobility develop into large semispherical colonies with CSCs features, which can be abrogated by magnetically restricting the ligand tether mobility. Mechanistically, the substrate with high ligand tether mobility suppresses integrin-mediated mechanotransduction and histone-related methylation, thereby enhancing cancer cell stemness. The culture-derived high-stemness cells can generate tumors both locally and at the distant lung and uterus much more efficiently than the low-stemness cells. We believe that this magnetic nanoplatform provides a promising strategy for investigating the dynamic interaction between CSCs and the microenvironment and establishing a cost-effective tumor spheroid model.


Asunto(s)
Mecanotransducción Celular , Neoplasias , Línea Celular Tumoral , Femenino , Humanos , Integrinas , Ligandos , Células Madre Neoplásicas , Microambiente Tumoral
6.
Small ; 11(21): 2543-54, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-25626995

RESUMEN

A novel dual-pH sensitive charge-reversal strategy is designed to deliver antitumor drugs targeting to tumor cells and to further promote the nuclei internalization by a stepwise response to the mildly acidic extracellular pH (≈6.5) of a tumor and endo/lysosome pH (≈5.0). Poly(L-lysine)-block-poly(L-leucine) diblock copolymer is synthesized and the lysine amino residues are amidated by 2,3-dimethylmaleic anhydride to form ß-carboxylic amide, making the polypeptides self-assemble into negatively charged micelles. The amide can be hydrolyzed when exposed to the mildly acidic tumor extracellular environment, which makes the micelles switch to positively charged and they are then readily internalized by tumor cells. A nuclear targeting Tat peptide is further conjugated to the polypeptide via a click reaction. The Tat is amidated by succinyl chloride to mask its positive charge and cell-penetrating function and thus to inhibit nonspecific cellular uptake. After the nanoparticles are internalized into the more acidic intracellular endo/lysosomes, the Tat succinyl amide is hydrolyzed to reactivate the Tat nuclear targeting function, promoting nanoparticle delivery into cell nuclei. This polypeptide nanocarrier facilitates tumor targeting and nuclear delivery simultaneously by simply modifying the lysine amino residues of polylysine and Tat into two different pH-sensitive ß-carboxylic amides.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Sistemas de Liberación de Medicamentos , Nanopartículas/química , Péptidos/química , Transporte Activo de Núcleo Celular , Núcleo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacocinética , Portadores de Fármacos/química , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Micelas , Péptidos/síntesis química , Polilisina/química , Multimerización de Proteína , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
7.
ACS Nano ; 18(17): 11070-11083, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38639726

RESUMEN

Effective antitumor immunotherapy depends on evoking a cascade of cancer-immune cycles with lymph nodes (LNs) as the initial sites for activating antitumor immunity, making drug administration through the lymphatic system highly attractive. Here, we describe a nanomedicine with dual responsiveness to pH and enzyme for a programmed activation of antitumor immune through the lymphatic system. The proposed nanomedicine can release the STING agonist diABZI-C2-NH2 in the LNs' acidic environment to activate dendritic cells (DCs) and T cells. Then, the remaining nanomedicine hitchhikes on the activated T cells (PD-1+ T cells) through binding to PD-1, resulting in an effective delivery into tumor tissues owing to the tumor-homing capacity of PD-1+ T cells. The enzyme matrix metalloproteinase-2 (MMP-2) being enriched in tumor tissue triggers the release of PD-1 antibody (aPD-1) which exerts immune checkpoint blockade (ICB) therapy. Eventually, the nanomedicine delivers a DNA methylation inhibitor GSK-3484862 (GSK) into tumor cells, and then the latter combines with granzyme B (GZMB) to trigger tumor cell pyroptosis. Consequently, the pyroptotic tumor cells induce robust immunogenic cell death (ICD) enhancing the DCs maturation and initiating the cascading antitumor immune response. Study on a 4T1 breast tumor mouse model demonstrates the prominent antitumor therapeutic outcome of this nanomedicine through creating a positive feedback loop of cancer-immunity cycles including immune activation in LNs, T cell-mediated drug delivery, ICB therapy, and tumor cell pyroptosis-featured ICD.


Asunto(s)
Nanomedicina , Animales , Ratones , Humanos , Inmunoterapia , Femenino , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Antineoplásicos/farmacología , Antineoplásicos/química , Línea Celular Tumoral
8.
Biomed Mater ; 18(5)2023 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-37567189

RESUMEN

Chemodynamic therapy (CDT) is a rising technology for cancer therapy by converting intracellular hydrogen peroxide (H2O2) into hydroxyl radical (•OH) via transition-metal-containing nanoparticles (NPs) catalysis reaction (i.e. Fenton reaction) to kill tumor cells. Highly efficient Fenton reaction and favorable delivery of the catalytic NPs 'nanoenzyme' are the key for successful treatment of cancer. In this work, we developed a novel nanoenzyme MnFe2O4@GFP forin vitroandin vivoantitumor therapy. A new MnFe2O4nanoparticle containing two transition-metal-element Fe and Mn was synthesized for enhanced Fenton reaction and used to co-deliver protein with high biocompatibility through post-modification with dopamine polymerization, green fluorescent protein adsorption, and PEG coating. The enrichment of H2O2and glutathione (GSH) in tumor tissue provided a favorable microenvironment forin situgeneration of toxic free radicals. Fe3+and GSH triggered a redox reaction to produce Fe2+, which in turn catalyzed H2O2into •OH, with the consumption of antioxidant GSH. By combining Fe3+with another catalyzer, the catalytic efficiency of the nanoenzyme were greatly improved. Consequently, the nanoenzyme showed efficient antitumor ability bothin vitroandin vivo. Thus, the multifunctional CDT nanoenzyme platform shows great promising for antitumor therapy through the combination of catalyzers Fe3+and Mn2+and codelivery of protein cargo.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Neoplasias , Humanos , Peróxido de Hidrógeno , Adsorción , Antioxidantes , Dopamina , Glutatión , Proteínas Fluorescentes Verdes , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
9.
Biomater Sci ; 11(23): 7489-7511, 2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-37873617

RESUMEN

Sonodynamic therapy (SDT) is an emerging treatment method. In comparison with photodynamic therapy (PDT), SDT exhibits deep penetration, high cell membrane permeability, and free exposure to light capacity. Unfortunately, owing to inappropriate ultrasound parameter selection, poor targeting of sonosensitizers, and the complex tumor environment, SDT is frequently ineffective. In this review, we describe the approaches for selecting ultrasound parameters and how to develop sonosensitizers to increase targeting and improve adverse tumor microenvironments. Furthermore, the potential of combining SDT with other treatment methods, such as chemotherapy, chemodynamic therapy, photodynamic therapy, photothermal therapy, and immunotherapy, is discussed to further increase the treatment efficiency of SDT.


Asunto(s)
Neoplasias , Fotoquimioterapia , Terapia por Ultrasonido , Humanos , Terapia por Ultrasonido/métodos , Neoplasias/tratamiento farmacológico , Fotoquimioterapia/métodos , Terapia Combinada , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Microambiente Tumoral
10.
J Control Release ; 355: 68-84, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36682726

RESUMEN

Treatments for osteosarcoma (OS) with pulmonary metastases reach a bottleneck with a survival rate of 10-20%. The suppressive tumor associated macrophages(TAMs) and CD47 over-expression greatly lead to the treatment failure. Sonodynamic therapy (SDT) can generate ROS with deep tumor penetration to induce tumor cell apoptosis, which is reported to further induce M1 macrophage polarization. CD47 inhibition combined with SDT to synergistically modulate TAMs may induce superior effects for OS treatment. In this work, for the first time, a biomimetic nanodrug named MPIRx was deveploped by loading IR780 (a sonosensitizer) and RRx-001 (a CD47 inhibitor) in PEG-PCL nanomicelles and then coating with OS cell membranes. After ultrasound activation, the nanodrug significantly inhibited OS proliferation and migration, induced apoptosis and immunogenic cell death in OS cells. Furthermore, MPIRx could guide macrophage migrating towards tumor cells and promote M1-type polarization while increasing the phagocytosis activity of macrophages on OS cells. Ultimately, MPIRx showed good tumor accumulation in vivo and successfully inhibited subcutaneous OS and orthotopic tumor with deterioration of pulmonary metastasis. Overall, by creating a local oxidative microenvironment and modulating the TAMs/CD47 in tumor tissue, the MPIRx nanodrug presents a novel strategy for macrophage-related immunotherapy to successfully eliminate OS and inhibit the intractable pulmonary metastasis.


Asunto(s)
Neoplasias Óseas , Nanopartículas , Osteosarcoma , Humanos , Antígeno CD47 , Fagocitosis , Osteosarcoma/tratamiento farmacológico , Neoplasias Óseas/tratamiento farmacológico , Nanopartículas/uso terapéutico , Línea Celular Tumoral , Microambiente Tumoral
11.
Biomaterials ; 299: 122134, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37167895

RESUMEN

Activated hepatic stellate cell (aHSC) is mainly responsible for deposition of extracellular collagen matrix that causes liver fibrosis. Although several siRNAs adequately inhibited HSC activation in vitro, they were demonstrated poor RNAi efficiency in vivo. Developing HSC-targeting and cytoplasmic delivery nanocarrier is highly essential to acquire a desirable siRNA therapeutic index for anti-liver fibrosis. Here, we developed a unique crosslinking nanopolyplex (called T-C-siRNA) modified by vitamin A (VA) with the well-designed natures, including the negative charge, retinol-binding protein (RBP) hijacking, and cytoplasmic siRNA release in response to ROS and cis diol molecules. The nanopolyplex was given a yolk-shell-like shape, camouflage ability in blood, and HSC-targeting capability by hijacking the endogenous ligand RBP via surface VA. PDGFR-ß siRNA (siPDGFR-ß) supplied via T-C-siPDGFR-ß nanopolyplex dramatically reduced HSC activation and its production of pro-fibrogenic proteins in vitro and in vivo. Furthermore, T-C-siPDGFR-ß nanopolyplex effectively alleviated CCl4-induced liver injury, decreased hepatic collagen sediment, and recovered liver function in mice. This study provides a sophisticated method for HSC-targeting cytoplasmic RNA delivery using endogenous ligand hijacking and dual sensitivity of ROS and cis diol compounds.


Asunto(s)
Células Estrelladas Hepáticas , Proteínas de Unión al Retinol , Animales , Ratones , Colágeno/metabolismo , Citoplasma/metabolismo , Ligandos , Cirrosis Hepática/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Proteínas de Unión al Retinol/genética , Proteínas de Unión al Retinol/metabolismo , Proteínas de Unión al Retinol/farmacología , ARN Bicatenario , ARN Interferente Pequeño/metabolismo
12.
Acta Biomater ; 161: 265-274, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36893956

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) exhibits difficult penetration of most drugs, leading to a very poor therapeutic outcome with a quite low five-year survival rate. The foremost reason is the highly-dense extracellular matrix (ECM) with abundant collagen and fibronectin secreted by the activated pancreatic stellate cells (PSCs). Here, we constructed a sono-responsive polymeric perfluorohexane (PFH) nanodroplet to elicit a deep drug penetration in PDAC via the combination of exogenous ultrasonic (US) exposure and endogenous ECM modulation for potent sonodynamic therapy (SDT) of PDAC. Under US exposure, the rapid drug release and deep penetration in PDAC tissues were realized. The released and well penetrated all-trans retinoic acid (ATRA) as an inhibitor of activated PSCs successfully reduced the secretion of ECM components to form a non-dense matrix conducive to drug diffusion. Meanwhile, the sonosensitizer, manganese porphyrin (MnPpIX), was triggered to produce robust reactive oxygen species (ROS) to exert the SDT effect under US exposure. Furthermore, oxygen (O2) delivered by PFH nanodroplets alleviated tumor hypoxia and enhanced the eradication of cancer cells. Overall, the sono-responsive polymeric PFH nanodroplets were successfully developed as an efficient strategy for PDAC therapy. STATEMENT OF SIGNIFICANCE: Pancreatic ductal adenocarcinoma (PDAC) is a representative refractory cancer with a highly dense extracellular matrix (ECM), making it difficult for most drugs to penetrate the nearly impenetrable desmoplastic stroma. Seeking methods for deep drug penetration is an extremely pressing matter for the treatment of PDAC and many other solid tumors. Herein, we designed a fluoroalkane-modified polymer to prepare a sono-responsive polymeric perfluorohexane (PFH) nanodroplet for loading sonosensitizers, and inhibitors of activated PSCs and O2. Under ultrasonic exposure, the nanodroplet elicited deep drug penetration in PDAC via ultrasonic disturbance and stromal remodeling, inducing potent sonodynamic therapy (SDT) of PDAC. By combining exogenous ultrasonic exposure and endogenous ECM modulation, this work successfully alleviated the severe physiological barrier of PDAC and achieved a favourable treatment effect.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Matriz Extracelular/patología , Línea Celular Tumoral , Neoplasias Pancreáticas
13.
Int J Biol Macromol ; 251: 126539, 2023 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-37634787

RESUMEN

Cancer immunotherapy is the most promising method for tumor therapy in recent years, among which the macrophages play a critical role in the antitumor immune response. However, tumor-associated macrophages (TAMs) usually display the tumor-promoting M2 phenotype rather than the tumor-killing M1 phenotype. Moreover, the over-expressed CD47 on tumor cells severely hinders the function of macrophages by blocking the CD47/SIRPα pathway. Herein, a nano-assembly system of CHTR/siRNA was constructed through the host-guest interaction of a hyperbranched amino-functionalized ß-cyclodextrin and immune agonist imiquimod (R848), while CD47 siRNA was loaded inside through electrostatic interaction. The Toll-like receptor (TLR) 7/8 agonist R848 can "re-educate" macrophages from the protumoral M2 phenotype to antitumoral M1 phenotype, while CD47 siRNA can down-regulate the "don't eat me" CD47 signal on the surface of cancer cells and enhance the phagocytosis of cancer cells by macrophages. Through the dual regulation of TAMs, the immunosuppressive tumor microenvironment was relieved, and the host-guest drug-carrying system resulted in synergistic immunotherapy effect on tumors and inhibited tumor growth. The facile self-assembly of nanodrug offers a new strategy in co-delivery of multiple therapeutic agents for cascade cancer immunotherapy.

14.
Biomater Sci ; 10(15): 4170-4183, 2022 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-35726827

RESUMEN

Although CuO-deposited bovine serum albumin (CuO-BSA) and glucose oxidase (GOx) were combined to achieve H2O2 self-supplied chemo-dynamic therapy (CDT) and glucose consumption-based starvation therapy, the uses of copper and GOx have not been optimized to enhance tumour-selective reactive oxygen species (ROS) generation and minimize toxicity to normal cells as well. Here, chemo-dynamic nanoparticles (CBGP NPs) were prepared through a facile biomineralization process and subsequent coatings with GOx and the cationic polymer PEG2k-PEI1.8k. Through optimizing the use of copper, GOx, and PEG2k-PEI1.8k, the CBGP NPs showed high cellular uptake efficiency, enhanced tumour-selective ROS generation, and minimal side effects toward normal cells. The CBGP NP-mediated glucose consumption, GSH-depletion, and ˙OH generation synergistically induced tumour cell apoptosis both in vitro and in vivo. It is believed that the optimized CBGP NPs can be a promising nanoplatform for effective tumour therapy with minimal side effects.


Asunto(s)
Glioma , Nanopartículas , Neoplasias , Animales , Línea Celular Tumoral , Cobre , Glioma/tratamiento farmacológico , Glucosa , Glucosa Oxidasa/farmacología , Peróxido de Hidrógeno , Ratones , Neoplasias/tratamiento farmacológico , Especies Reactivas de Oxígeno
15.
Biomaterials ; 269: 120636, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33453632

RESUMEN

Although a combination with photodynamic therapy (PDT) is a potential means to improve the immune checkpoint blockade (ICB)-based anticancer immunotherapy, this strategy is subjected to the extremely poor light penetration in melanoma. Herein, we develop a lipid (LP)-based micellar nanocarrier encapsulating sonosensitizer chlorin e6 (Ce6) in the core, conjugating anti-PD-L1 antibody (aPD-L1) to the interlayer through MMP-2-cleavable peptide, and bearing a PEG coating sheddable at low pH value (≈6.5) of tumor microenvironment. The unique nanocarrier design allows a tumor-targeting delivery to activate the anti-tumor immunity and meanwhile to reduce immune-related adverse effects (irAEs). Moreover, a sonodynamic therapy (SDT) is triggerable by using ultrasonic insonation to produce tumor-killing reactive oxygen species (ROS), thereby bypassing the poor light penetration which restricts PDT in melanoma. A combination of SDT with aPD-L1 immunotherapy effectively promotes tumor infiltration and activation of cytotoxic T cells, which resulted in robust anti-cancer immunity and long-term immune memory to effectively suppress melanoma growth and postoperative recurrence. This strategy for tumor-targeting codelivery of immune checkpoint inhibitors and SDT agents could be readily extended to other tumor types for better immunotherapeutic outcome and reduced irAEs.


Asunto(s)
Fotoquimioterapia , Microambiente Tumoral , Línea Celular Tumoral , Humanos , Inmunoterapia , Recurrencia Local de Neoplasia
16.
Free Radic Biol Med ; 175: 193-205, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34492311

RESUMEN

Hepatic stellate cells (HSCs) play key roles in liver fibrosis (LF) and hepatocellular carcinoma (HCC). We previously reported that spleen tyrosine kinase (SYK) is critical for HSCs activation, however, the mechanisms are insufficiently understood. In the present study, we found that SYK facilitated autophagy to promote HSCs activation by enhancing reactive oxygen species (ROS) generation. However, SYK inhibitor GS-9973 could efficiently reduce HSCs ROS generation in vitro but not in vivo. Mechanistically, hepatocytes (HCs) would release ROS outside and then diffuse into HSCs to promote autophagy and activation in vitro in the context of inflammation. We then further examined the ROS contents in liver sections and primary liver cells of carbon tetrachloride (CCl4) induced mice treated with or without different doses of Silybin, a natural compound characterized by a well-established antioxidant and hepatoprotective properties, and found that ROS intensities in both liver sections and their deprived primary cells were efficiently inhibited in a dose-dependent fashion. Lastly, we evaluated the rational combination of Silybin and GS-9973 in the treatment of CCl4 induced mice and found that this combination is well tolerated and acts synergistically against HSCs activity, LF and HCC. The combinational use of Silybin and GS-9973 could be a promising therapeutic strategy in patients suffering from LF and even HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Células Estrelladas Hepáticas/patología , Humanos , Hígado/patología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/patología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Ratones , Especies Reactivas de Oxígeno , Quinasa Syk
17.
Biomater Sci ; 9(15): 5218-5226, 2021 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-34169939

RESUMEN

Combination therapy provides significantly better outcomes than a single drug treatment and becomes an efficient strategy for cancer therapy at present. Owing to the advantages of improved drug bioavailability, decreased side effects, and drug codelivery properties, polymeric carrier-based nanodrugs show great application potential in combination therapy. In this study, a pH-responsive block polymer consisting of polyethylene glycol (mPEG) and poly(asparagyl diisopropylethylenediamine-co-phenylalanine) (P(Asp(DIP)-co-Phe)) is synthesized for drug delivery. The polymer self-assembles into nanovesicles and simultaneously encapsulates the hydrophilic hypoxia-activated prodrug tirapazamine (TPZ) and the hydrophobic photosensitizer dihydrogen porphin (chlorin e6, Ce6). The formed nanodrug can be triggered by near infrared irradiation to induce photodynamic therapy (PDT), resulting in a hypoxic tumor environment to activate the prodrug TPZ to achieve efficient chemotherapy. The cascade synergistic therapeutic effect is evaluated both in vitro and in vivo in a breast cancer mice model. This study reveals a potential strategy for efficient cancer therapy by using Ce6 and TPZ co-encapsulated nanovesicles.


Asunto(s)
Nanopartículas , Neoplasias , Fotoquimioterapia , Porfirinas , Animales , Línea Celular Tumoral , Hipoxia , Ratones , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes , Tirapazamina
18.
Biomater Sci ; 9(20): 6966-6969, 2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34546259

RESUMEN

Correction for 'Recent development of gene therapy for pancreatic cancer using non-viral nanovectors' by Yu Liu et al., Biomater. Sci., 2021, DOI: 10.1039/d1bm00748c.

19.
Biomater Sci ; 9(20): 6673-6690, 2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34378568

RESUMEN

Pancreatic cancer (PC), characterized by its dense desmoplastic stroma and hypovascularity, is one of the most lethal cancers with a poor prognosis in the world. Traditional treatments such as chemotherapy, radiotherapy, and targeted therapy show little benefit in the survival rate in patients with advanced PC due to the poor penetration and resistance of drugs, low radiosensitivity, or severe side effects. Gene therapy can modify the morbific and drug-resistant genes as well as insert the tumor-suppressing genes, which has been shown to have great potential in PC treatment. The development of safe non-viral vectors for the highly efficient delivery of nucleic acids is essential for effective gene therapy, and has been attracting much attention. In this review, we first summarized the PC-promoting genes and gene therapies using plasmid DNA, mRNA, miRNA/siRNA-based RNA interference technology, and genome editing technology. Second, the commonly used non-viral nanovector and theranostic gene delivery nanosystem, especially the tumor microenvironment-sensitive delivery nanosystem and the cell/tumor-penetrating delivery nanosystem, were introduced. Third, a combination of non-viral nanovector-based gene therapy and other therapies, such as immunotherapy, chemotherapy, photothermal therapy (PTT), and photodynamic therapy (PDT), for PDAC treatment was discussed. Finally, a number of clinical trials have demonstrated the proof-of-principle that gene therapy or the combination of gene therapy and chemotherapy using non-viral vectors can inhibit the progression of PC. Although most of the non-viral vector-based gene therapies and their combination therapy are still under preclinical research, the development of genetics, molecular biology, and novel vectors would promote the clinical transformation of gene therapy.


Asunto(s)
MicroARNs , Neoplasias Pancreáticas , Terapia Genética , Humanos , Inmunoterapia , MicroARNs/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Microambiente Tumoral
20.
ACS Appl Mater Interfaces ; 13(11): 12845-12856, 2021 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-33709684

RESUMEN

Tumor immunotherapy like immune checkpoint blockade (ICB) shows great success nowadays but is severely limited by low response rates and immune-related adverse events (IRAEs). While photodynamic therapy (PDT) could efficiently eradicate tumor cells and further induce immune responses to promote activating of T lymphocytes. Herein a nanodrug hierarchically incorporating photosensitizer and PD-L1 antibody was developed for synergistic tumor immuno-photodynamic therapy. A pH/enzyme dual-sensitive polymeric micelle with sheddable PEG coating was designed for codelivery of PD-L1 antibody and zinc phthalocyanine (ZnPc) in the tumor. The tumor microenvironment featuring low pH and high matrix metallopeptidase 2 (MMP-2) sequentially triggered the shedding of PEG and the release of PD-L1 antibody to exert local ICB in tumor tissue, after which the remaining nanodrug with ZnPc undergoing charge reversal was readily delivered into tumor cells. With light irradiation, the photodynamic therapy effect of sAMPc induced immunogenic cell death of tumor cells and further promoted intratumor recruitment of CD8+ T cells, thus resulting in a synergistic immuno-photodynamic therapy with ICB. Moreover, the PEG-sheddable strategy endowed the nanodrug with stealth properties in blood circulation, making the IRAEs of PD-L1 antibody significantly reduced. This pH/MMP-2 dual-sensitive PEG sheddable nanodrug provids a promising strategy for well-combined ICB therapy and PDT to achieve improved anticancer immuno-photodynamic therapy with reduced adverse effects.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Antígeno B7-H1/antagonistas & inhibidores , Preparaciones de Acción Retardada/química , Indoles/administración & dosificación , Compuestos Organometálicos/administración & dosificación , Fármacos Fotosensibilizantes/administración & dosificación , Polietilenglicoles/química , Antineoplásicos Inmunológicos/uso terapéutico , Línea Celular Tumoral , Humanos , Concentración de Iones de Hidrógeno , Indoles/uso terapéutico , Isoindoles , Masculino , Neoplasias/tratamiento farmacológico , Compuestos Organometálicos/uso terapéutico , Fotoquimioterapia , Fármacos Fotosensibilizantes/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Compuestos de Zinc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA