Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.837
Filtrar
Más filtros











Intervalo de año de publicación
1.
Biomaterials ; 312: 122712, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39098305

RESUMEN

Immunosuppressive tumor microenvironment (ITM) severely limited the efficacy of immunotherapy against triple-negative breast cancer (TNBC). Herein, Apt-LPR, a light-activatable photodynamic therapy (PDT)/RNAi immune synergy-enhancer was constructed by co-loading miR-34a and photosensitizers in cationic liposomes (in phase III clinical trial). Interestingly, the introduction of tumor-specific aptamers creates a special "Liposome-Aptamer-Target" interface, where the aptamers are initially in a "lying down" state but transform to "standing up" after target binding. The interfacing mechanism was elaborately revealed by computational and practical experiments. This unique interface endowed Apt-LPR with neutralized surface potential of cationic liposomes to reduce non-specific cytotoxicity, enhanced DNase resistance to protect aptamers, and preserved target-binding ability for selective drug delivery. Upon near-infrared irradiation, the generated reactive oxygen species would oxidize unsaturated phospholipids to destabilize both liposomes and lysosomes, realizing stepwise lysosomal escape of miR-34a for tumor cell apoptosis and downregulation of PD-L1 to suppress immune escape. Together, tumor-associated antigens released from PDT-damaged mitochondria and endoplasmic reticulum could activate the suppressive immune cells to establish an "immune hot" milieu. The collaborative immune-enhancing strategy effectively aroused systemic antitumor immunity and inhibited primary and distal tumor progression as well as lung metastasis in 4T1 xenografted mouse models. The photo-controlled drug release and specific tumor-targeting capabilities of Apt-LPR were also visualized in MDA-MB-231 xenografted zebrafish models. Therefore, this photoswitchable PDT/RNAi immune stimulator offered a powerful approach to reprogramming ITM and reinforcing cancer immunotherapy efficacy.


Asunto(s)
Liposomas , MicroARNs , Fotoquimioterapia , Fármacos Fotosensibilizantes , Neoplasias de la Mama Triple Negativas , Microambiente Tumoral , Animales , Humanos , Liposomas/química , MicroARNs/genética , MicroARNs/metabolismo , Fotoquimioterapia/métodos , Microambiente Tumoral/efectos de los fármacos , Línea Celular Tumoral , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Femenino , Neoplasias de la Mama Triple Negativas/terapia , Neoplasias de la Mama Triple Negativas/patología , Ratones , Aptámeros de Nucleótidos/química , Preparaciones de Acción Retardada/química , Interferencia de ARN , Pez Cebra
2.
Biomaterials ; 313: 122801, 2025 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-39236630

RESUMEN

Chemoimmunotherapy is an emerging paradigm in the clinic for treating several malignant diseases, such as non-small cell lung cancer, breast cancer, and large B-cell lymphoma. However, the efficacy of this strategy is still restricted by serious adverse events and a high therapeutic termination rate, presumably due to the lack of tumor-targeted distribution of both chemotherapeutic and immunotherapeutic agents. Targeted drug delivery has the potential to address this issue. Among the most promising nanocarriers in clinical translation, liposomes have drawn great attention in cancer chemoimmunotherapy in recent years. Liposomes-enabled cancer chemoimmunotherapy has made significant progress in clinics, with impressive therapeutic outcomes. This review summarizes the latest preclinical and clinical progress in liposome-enabled cancer chemoimmunotherapy and discusses the challenges and future directions of this field.


Asunto(s)
Inmunoterapia , Liposomas , Neoplasias , Liposomas/química , Humanos , Inmunoterapia/métodos , Animales , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Antineoplásicos/uso terapéutico , Antineoplásicos/administración & dosificación
3.
J Colloid Interface Sci ; 677(Pt B): 79-90, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39137565

RESUMEN

Photothermal therapy combined with chemotherapy has shown great promise in the treatment of cancer. In this synergistic system, a safe, stable, and efficient photothermal agent is desired. Herein, an effective photothermal agent, carbon quantum dots (CQDs), was initially synthesized and then rationally constructed a folic acid (FA)-targeted photothermal multifunctional nanoplatform by encapsulating CQDs and the anticancer drug doxorubicin (DOX) in the liposomes. Indocyanine green (ICG), a near infrared (NIR) photothermal agent, approved by the U.S. Food and Drug Administration, was embedded in the bilayer membrane to further enhance the photothermal effects and facilitate the rapid cleavage of liposomes for drug release. Triggered by the NIR laser, this engineered photothermal multifunctional nanoplatform, not only exhibited an excellent performance with the photothermal conversion efficiency of up to 47.14%, but also achieved controlled release of the payloads. In vitro, and in vivo experiments demonstrated that the photothermal multifunctional nanoplatform had excellent biocompatibility, enhanced tumor-specific targeting, stimuli-responsive drug release, effective cancer cell killing and tumor suppression through multi-modal synergistic therapy. The successful construction of this NIR light-triggered targeted photothermal multifunctional nanoplatform will provide a promising strategy for the design and development of synergistic chemo-photothermal combination therapy and improve the therapeutic efficacy of cancer treatment.


Asunto(s)
Carbono , Doxorrubicina , Terapia Fototérmica , Puntos Cuánticos , Doxorrubicina/farmacología , Doxorrubicina/química , Humanos , Puntos Cuánticos/química , Animales , Ratones , Carbono/química , Carbono/farmacología , Verde de Indocianina/química , Verde de Indocianina/farmacología , Ácido Fólico/química , Ácido Fólico/farmacología , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Liposomas/química , Tamaño de la Partícula , Ensayos de Selección de Medicamentos Antitumorales , Antineoplásicos/farmacología , Antineoplásicos/química , Rayos Infrarrojos , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/administración & dosificación , Propiedades de Superficie , Proliferación Celular/efectos de los fármacos , Ratones Endogámicos BALB C , Neoplasias Experimentales/patología , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/terapia , Nanopartículas/química
4.
J Nanobiotechnology ; 22(1): 603, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-39367395

RESUMEN

Surgical resection remains the primary treatment modality for glioblastoma (GBM); however, the infiltrative nature of GBM margins complicates achieving complete tumor removal. Additionally, the blood-brain barrier (BBB) poses a formidable challenge to effective probe delivery, thereby hindering precise imaging-guided surgery. Here, we introduce hybrid cell membrane-coated indocyanine green (ICG) liposomes (HM-Lipo-ICG) as biomimetic near-infrared (NIR) fluorescent probes for targeted BBB penetration and accurate delineation of infiltrative GBM margins. HM-Lipo-ICG encapsulates clinically approved ICG within its core and utilizes a hybrid cell membrane exterior, enabling specific targeting and enhanced BBB permeation. Quantitative assessments demonstrate that HM-Lipo-ICG achieves BBB penetration efficiency 2.8 times higher than conventional ICG liposomes. Mechanistically, CD44 receptor-mediated endocytosis facilitates BBB translocation of HM-Lipo-ICG. Furthermore, HM-Lipo-ICG enables high-contrast NIR imaging, achieving a signal-to-background ratio of 6.5 in GBM regions of an orthotopic glioma mouse model, thereby improving tumor margin detection accuracy fourfold (84.4% vs. 22.7%) compared to conventional ICG liposomes. Application of HM-Lipo-ICG facilitates fluorescence-guided precision surgery, resulting in complete resection of GBM cells. This study underscores the potential of hybrid cell membrane-coated liposomal probes in precisely visualizing and treating infiltrative GBM margins.


Asunto(s)
Barrera Hematoencefálica , Neoplasias Encefálicas , Membrana Celular , Glioblastoma , Verde de Indocianina , Liposomas , Verde de Indocianina/química , Liposomas/química , Barrera Hematoencefálica/metabolismo , Glioblastoma/diagnóstico por imagen , Glioblastoma/patología , Animales , Ratones , Humanos , Neoplasias Encefálicas/diagnóstico por imagen , Línea Celular Tumoral , Membrana Celular/metabolismo , Colorantes Fluorescentes/química , Ratones Desnudos , Imagen Óptica/métodos
5.
Mol Pharm ; 21(10): 4912-4923, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39370820

RESUMEN

The disadvantage of a traditional dosage regimen is the inability to deliver a sufficient drug concentration to the lesion site, which can result in adverse side effects due to nonspecific drug delivery. Actively targeting hepatic cells is a promising therapeutic strategy for liver disease. In this study, l-carnitine and a targeting peptide derived from the hepatitis B virus large envelope protein were used to modify liposomes for drug delivery to the liver through the sodium taurocholate cotransporting polypeptide (NTCP) and the organic cation/carnitine transporter 2 (OCTN2) receptors. Silybin was selected as the model drug. The solubility of silybin can reach 0.3 mg/mL after encapsulation in liposomes. The NTCP-specific and OCTN2-accelerated Myrcludex B and l-carnitine dual-modified liposomes were validated in vitro. The uptake of coumarin-6 in dual ligand-modified liposomes by hepatocytes was up to 2.36 µg/mg compared with unmodified liposomes (1.05 µg/mg). The pharmacokinetics and targeting abilities of various liposome formulations were evaluated in Kunming mice. Targeted liposomes increased the concentration of silybin and prolonged the drug's retention time in the liver. The area under the liver's pharmacokinetic curve of targeted liposomes was twice that of silybin injection, suggesting the promising application potential of silybin-loaded hepatotropic nanovesicles.


Asunto(s)
Liposomas , Hígado , Transportadores de Anión Orgánico Sodio-Dependiente , Silibina , Simportadores , Silibina/farmacocinética , Silibina/administración & dosificación , Liposomas/química , Animales , Ratones , Simportadores/metabolismo , Hígado/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Humanos , Masculino , Miembro 5 de la Familia 22 de Transportadores de Solutos , Carnitina/farmacocinética , Carnitina/administración & dosificación , Carnitina/química , Hepatocitos/metabolismo , Hepatocitos/efectos de los fármacos , Silimarina/farmacocinética , Silimarina/administración & dosificación , Silimarina/química , Cumarinas/química , Cumarinas/farmacocinética , Cumarinas/administración & dosificación , Lipopéptidos
6.
Int J Nanomedicine ; 19: 8709-8727, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39220191

RESUMEN

Background: The formation of adhesion after tendon injury represents a major obstacle to tendon repair, and currently there is no effective anti-adhesion method in clinical practice. Oxidative stress, inflammation, and fibrosis can occur in tendon injury and these factors can lead to tendon adhesion. Antioxidant carbon dots and ursolic acid (UA) both possess antioxidant and anti-inflammatory properties. In this experiment, we have for the first time created RCDs/UA@Lipo-HAMA using red fluorescent carbon dots and UA co-encapsulated liposomes composite hyaluronic acid methacryloyl hydrogel. We found that RCDs/UA@Lipo-HAMA could better attenuate adhesion formation and enhance tendon healing in tendon injury. Materials and Methods: RCDs/UA@Lipo-HAMA were prepared and characterized. In vitro experiments on cellular oxidative stress and fibrosis were performed. Reactive oxygen species (ROS), and immunofluorescent staining of collagens type I (COL I), collagens type III (COL III), and α-smooth muscle actin (α-SMA) were used to evaluate anti-oxidative and anti-fibrotic abilities. In vivo models of Achilles tendon injury repair (ATI) and flexor digitorum profundus tendon injury repair (FDPI) were established. The major organs and blood biochemical indicators of rats were tested to determine the toxicity of RCDs/UA@Lipo-HAMA. Biomechanical testing, motor function analysis, immunofluorescence, and immunohistochemical staining were performed to assess the tendon adhesion and repair after tendon injury. Results: In vitro, the RCDs/UA@Lipo group scavenged excessive ROS, stabilized the mitochondrial membrane potential (ΔΨm), and reduced the expression of COL I, COL III, and α-SMA. In vivo, assessment results showed that the RCDs/UA@Lipo-HAMA group improved collagen arrangement and biomechanical properties, reduced tendon adhesion, and promoted motor function after tendon injury. Additionally, the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1) in the RCDs/UA@Lipo-HAMA group increased; the levels of cluster of differentiation 68 (CD68), inducible Nitric Oxide Synthase (iNOS), COL III, α-SMA, Vimentin, and matrix metallopeptidase 2 (MMP2) decreased. Conclusion: In this study, the RCDs/UA@Lipo-HAMA alleviated tendon adhesion formation and enhanced tendon healing by attenuating oxidative stress, inflammation, and fibrosis. This study provided a novel therapeutic approach for the clinical treatment of tendon injury.


Asunto(s)
Antioxidantes , Carbono , Hidrogeles , Liposomas , Ratas Sprague-Dawley , Traumatismos de los Tendones , Triterpenos , Ácido Ursólico , Animales , Triterpenos/farmacología , Triterpenos/química , Antioxidantes/farmacología , Antioxidantes/química , Liposomas/química , Traumatismos de los Tendones/tratamiento farmacológico , Adherencias Tisulares/tratamiento farmacológico , Carbono/química , Carbono/farmacología , Hidrogeles/química , Hidrogeles/farmacología , Ratas , Estrés Oxidativo/efectos de los fármacos , Masculino , Cicatrización de Heridas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Puntos Cuánticos/química , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Tendón Calcáneo/efectos de los fármacos , Tendón Calcáneo/lesiones
7.
AAPS PharmSciTech ; 25(7): 213, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39266895

RESUMEN

To overcome the challenges associated with the co-delivery of AuNPs (gold nanoparticles) and miRNA as an anti-breast cancer combination therapy, niosomal systems were developed using Span 60, cholesterol, and a cationic lipid (CTAB), and the formulations were optimized using Box-Behnken experimental design. The niosomal formulations with the smallest size were selected for further optimization of size, surface charge, entrapment efficiency, and stability. To achieve this, AuNPs and DSPE-PEG2000 (2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000)were added to the formulation. The optimized niosomal formulation could effectively encapsulate AuNPs with an entrapment efficiency of 34.49% ± 0.84 and a spherical particle size of 153.6 ± 4.62 nm. The incorporation of PEG and CTAB led to notable enhancements in the overall characteristics of the delivery system. To evaluate the effectiveness of the combination therapy, various assessments such as cytotoxicity, apoptosis, and gene expression properties were conducted. The results demonstrated that the combination delivery using the new C-PEG-Nio-AuNPs (cationic pegylated niosomal gold nanoparticles) system and miRNA had the lowest IC50, the highest apoptosis rate, and the most significant upregulation of miRNA and BAX/BCL2 expression in MCF-7 cell growth. In conclusion, this innovative co-delivery approach represents a promising breakthrough in the development of therapeutic agents for breast cancer treatment. By combining multiple therapeutic agents within a single delivery system, this method has the potential to enhance treatment efficacy, reduce side effects, and improve patient outcomes.


Asunto(s)
Neoplasias de la Mama , Oro , Liposomas , Nanopartículas del Metal , MicroARNs , Tamaño de la Partícula , Polietilenglicoles , Oro/química , Humanos , MicroARNs/administración & dosificación , Células MCF-7 , Polietilenglicoles/química , Nanopartículas del Metal/química , Liposomas/química , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Cationes/química , Apoptosis/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Supervivencia Celular/efectos de los fármacos , Fosfatidiletanolaminas/química
8.
ACS Appl Bio Mater ; 7(9): 6025-6033, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39231306

RESUMEN

Liposomes are applied to various anticancer treatments as representative drug delivery carriers. However, liposomes do not have their own targeting properties; therefore, there are limitations in drug delivery to specific tissues or cells. High targetability in drug delivery is an important factor in improving bioavailability and drug efficacy and reducing side effects; recent research has been actively investigated to modify the surface of liposomes to give them specific functions. In this study, we studied a drug delivery system for anticancer treatment that enhances targeting ability through fusion with exosomes on the surface of liposomes. We designed exosome-liposome hybrid nanoparticles loaded with a gemcitabine prodrug as a treatment for pancreatic ductal adenocarcinoma (PDAC). Membrane fusion with exosomes shows excellent targeting ability to pancreatic cancer cells due to intrinsic targeting ability and expansion of the macropinocytosis pathway.


Asunto(s)
Carcinoma Ductal Pancreático , Desoxicitidina , Ensayos de Selección de Medicamentos Antitumorales , Vesículas Extracelulares , Gemcitabina , Liposomas , Nanopartículas , Neoplasias Pancreáticas , Tamaño de la Partícula , Profármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/química , Desoxicitidina/farmacología , Profármacos/química , Profármacos/farmacología , Humanos , Liposomas/química , Nanopartículas/química , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/metabolismo , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Proliferación Celular/efectos de los fármacos , Ensayo de Materiales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Antineoplásicos/química , Antineoplásicos/farmacología
9.
Int J Mol Sci ; 25(17)2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39273099

RESUMEN

Cholesterol homeostasis is pivotal for cellular function. Acyl-coenzyme A:cholesterol acyltransferase 1 (ACAT1), also abbreviated as SOAT1, is an enzyme responsible for catalyzing the storage of excess cholesterol to cholesteryl esters. ACAT1 is an emerging target to treat diverse diseases including atherosclerosis, cancer, and neurodegenerative diseases. F12511 is a high-affinity ACAT1 inhibitor. Previously, we developed a stealth liposome-based nanoparticle to encapsulate F12511 to enhance its delivery to the brain and showed its efficacy in treating a mouse model for Alzheimer's disease (AD). In this study, we introduce F26, a close derivative of F12511 metabolite in rats. F26 was encapsulated in the same DSPE-PEG2000/phosphatidylcholine (PC) liposome-based nanoparticle system. We employed various in vitro and in vivo methodologies to assess F26's efficacy and toxicity compared to F12511. The results demonstrate that F26 is more effective and durable than F12511 in inhibiting ACAT1, in both mouse embryonic fibroblasts (MEFs), and in multiple mouse tissues including the brain tissues, without exhibiting any overt systemic or neurotoxic effects. This study demonstrates the superior pharmacokinetic and safety profile of F26 in wild-type mice, and suggests its therapeutic potential against various neurodegenerative diseases including AD.


Asunto(s)
Liposomas , Nanopartículas , Esterol O-Aciltransferasa , Animales , Liposomas/química , Ratones , Nanopartículas/química , Esterol O-Aciltransferasa/antagonistas & inhibidores , Esterol O-Aciltransferasa/metabolismo , Acetil-CoA C-Acetiltransferasa/antagonistas & inhibidores , Acetil-CoA C-Acetiltransferasa/metabolismo , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacocinética , Ratas , Masculino , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo
10.
Molecules ; 29(17)2024 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-39274849

RESUMEN

Stimulus-responsive materials hold significant promise for antitumor applications due to their variable structures and physical properties. In this paper, a series of peptides with a responsive viologen derivative, Pep-CnV (n = 1, 2, 3) were designed and synthesized. The process and mechanism of the interaction were studied and discussed. An ultraviolet-visible (UV) spectrophotometer and fluorescence spectrophotometer were used to study their redox responsiveness. Additionally, their secondary structures were measured by Circular Dichroism (CD) in the presence or absence of the reductant, Na2SO3. DPPC and DPPG liposomes were prepared to mimic normal and tumor cell membranes. The interaction between Pep-CnV and biomembranes was investigated by the measurements of surface tension and cargo leakage. Results proved Pep-CnV was more likely to interact with the DPPG liposome and destroy its biomembrane under the stimulus of the reductant. And the destruction increased with the length of the hydrophobic tail chain. Pep-CnV showed its potential as an intelligent antitumor agent.


Asunto(s)
Interacciones Hidrofóbicas e Hidrofílicas , Liposomas , Liposomas/química , Sustancias Reductoras/química , Oxidación-Reducción , Péptidos/química , Membrana Celular/química , Membrana Celular/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacología , Fosfatidilgliceroles/química , Dicroismo Circular
11.
Molecules ; 29(17)2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39274927

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is one of the most common metabolic diseases encountered in clinical practice, which is characterized by the excessive accumulation of triglycerides (steatosis), and a variety of metabolic abnormalities including lipid metabolism and bile acid metabolism are closely related to NAFLD. In China, Gynostemma pentaphyllum is used as functional food and Chinese medicine to treat various diseases, especially NAFLD, for a long time. However, the active components that exert the main therapeutic effects and their mechanisms remain unclear. In this study, Gypensapogenin A was isolated from the total saponins of G. pentaphyllum and prepared as a liposomal delivery system. Gypensapogenin A liposomes could activate FXR, inhibit the expression of CYP7A1 and CYP8B1, increase the expression of CYP27A1, modulate the ratio of CA and CDCA, decrease the content of CA, and increase the content of CDCA, thus forming a virtuous cycle of activating FXR to play a role in lowering blood lipid levels.


Asunto(s)
Gynostemma , Metabolismo de los Lípidos , Liposomas , Receptores Citoplasmáticos y Nucleares , Receptores Citoplasmáticos y Nucleares/metabolismo , Liposomas/química , Metabolismo de los Lípidos/efectos de los fármacos , Humanos , Animales , Gynostemma/química , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Saponinas/farmacología , Saponinas/química , Células Hep G2 , Ratones , Ácidos y Sales Biliares/metabolismo , Hepatocitos/metabolismo , Hepatocitos/efectos de los fármacos
12.
J Nanobiotechnology ; 22(1): 561, 2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39272205

RESUMEN

BACKGROUND: T cell-based immunotherapies are facing great challenges in the recruitment and activation of tumor-specific T cells against solid tumors. Among which, utilizing nanobody (Nb) or nanobodies (Nbs) to construct T cell engager has emerged as a more practical potential for enhancing the anti-tumor effectiveness of T cells. Here, we designed a new Nb-guided multifunctional T cell engager (Nb-MuTE) that not only recruited effector T cells into the tumor tissues, but also efficiently activated T cells anti-tumor immunity when synergies with photothermal effect. RESULTS: The Nb-MuTE, which was constructed based on an indocyanine green (ICG)-containing liposome with surface conjugation of CD105 and CD3 Nbs, and showed excellent targetability to both tumor and T cells, following enhancement of activation, proliferation and cytokine secretion of tumor-specific T cells. Notably, the immunological anti-tumor functions of Nb-MuTE-mediated T cells were further enhanced by the ICG-induced photothermal effect in vitro and in vivo. CONCLUSIONS: Such a new platform Nb-MuTE provides a practical and "all-in-one" strategy to potentiate T cell responses for the treatment of solid tumor in clinic.


Asunto(s)
Inmunoterapia , Verde de Indocianina , Anticuerpos de Dominio Único , Linfocitos T , Animales , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/inmunología , Ratones , Linfocitos T/inmunología , Verde de Indocianina/química , Inmunoterapia/métodos , Línea Celular Tumoral , Humanos , Neoplasias/terapia , Neoplasias/inmunología , Femenino , Ratones Endogámicos BALB C , Terapia Fototérmica/métodos , Liposomas/química , Activación de Linfocitos , Ratones Endogámicos C57BL , Complejo CD3/inmunología
13.
Int J Mol Sci ; 25(17)2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39273286

RESUMEN

The search for new antineoplastic agents is imperative, as cancer remains one of the most preeminent causes of death worldwide. Since the discovery of the therapeutic potential of cisplatin, the study of metallodrugs in cancer chemotherapy acquired increasing interest. Starting from cisplatin derivatives, such as oxaliplatin and carboplatin, in the last years, different compounds were explored, employing different metal centers such as iron, ruthenium, gold, and palladium. Nonetheless, metallodrugs face several drawbacks, such as low water solubility, rapid clearance, and possible side toxicity. Encapsulation has emerged as a promising strategy to overcome these issues, providing both improved biocompatibility and protection of the payload from possible degradation in the biological environment. In this respect, liposomes, which are spherical vesicles characterized by an aqueous core surrounded by lipid bilayers, have proven to be ideal candidates due to their versatility. In fact, they can encapsulate both hydrophilic and hydrophobic drugs, are biocompatible, and their properties can be tuned to improve the selective delivery to tumour sites exploiting both passive and active targeting. In this review, we report the most recent findings on liposomal formulations of metallodrugs, with a focus on encapsulation techniques and the obtained biological results.


Asunto(s)
Antineoplásicos , Liposomas , Neoplasias , Liposomas/química , Humanos , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Antineoplásicos/administración & dosificación , Neoplasias/tratamiento farmacológico , Animales , Composición de Medicamentos , Sistemas de Liberación de Medicamentos/métodos
14.
J Nanobiotechnology ; 22(1): 558, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39267061

RESUMEN

Breast cancer therapy has significantly advanced by targeting the programmed cell death-ligand 1/programmed cell death-1 (PD-L1/PD-1) pathway. BMS-202 (a smallmolecule PD-L1 inhibitor) induces PD-L1 dimerization to block PD-1/PD-L1 interactions, allowing the T-cell-mediated immune response to kill tumor cells. However, immunotherapy alone has limited effects. Clinically approved photodynamic therapy (PDT) activates immunity and selectively targets malignant cells. However, PDT aggravates hypoxia, which may compromise its therapeutic efficacy and promote tumor metastasis. We designed a tumor-specific delivery nanoplatform of liposomes that encapsulate the hypoxia-sensitive antitumor drug tirapazamine (TPZ) and the small-molecule immunosuppressant BMS. New indocyanine green (IR820)-loaded polyethylenimine-folic acid (PEI-FA) was complexed with TPZ and BMS-loaded liposomes via electrostatic interactions to form lipid nanocomposites. This nanoplatform can be triggered by near-infrared irradiation to induce PDT, resulting in a hypoxic tumor environment and activation of the prodrug TPZ to achieve efficient chemotherapy. The in vitro and in vivo studies demonstrated excellent combined PDT, chemotherapy, and immunotherapy effects on the regression of distant tumors and lung metastases, providing a reference method for the preparation of targeted agents for treating breast cancer.


Asunto(s)
Neoplasias de la Mama , Inmunoterapia , Liposomas , Liposomas/química , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/terapia , Inmunoterapia/métodos , Animales , Ratones , Humanos , Línea Celular Tumoral , Fotoquimioterapia/métodos , Verde de Indocianina/química , Verde de Indocianina/uso terapéutico , Verde de Indocianina/análogos & derivados , Ratones Endogámicos BALB C , Tirapazamina/química , Tirapazamina/farmacología , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Fototerapia/métodos
15.
J Nanobiotechnology ; 22(1): 551, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39252079

RESUMEN

Chemodynamic therapy represents a novel tumor therapeutic modality via triggering catalytic reactions in tumors to yield highly toxic reactive oxygen species (ROS). Nevertheless, low efficiency catalytic ability, potential systemic toxicity and inefficient tumor targeting, have hindered the efficacy of chemodynamic therapy. Herein, a rationally designed catalytic nanoplatform, composed of folate acid conjugated liposomes loaded with copper peroxide (CP) and chloroquine (CQ; a clinical drug) (denoted as CC@LPF), could power maximal tumor cytotoxicity, mechanistically via maneuvering endogenous and exogenous copper for a highly efficient catalytic reaction. Despite a massive autophagosome accumulation elicited by CP-powered autophagic initiation and CQ-induced autolysosomal blockage, the robust ROS, but not aberrant autophagy, underlies the synergistic tumor inhibition. Otherwise, this combined mode also elicits an early onset, above all, long-term high-level existence of immunogenic cell death markers, associated with ROS and aberrant autophagy -triggered endoplasmic reticulum stress. Besides, CC@LPF, with tumor targeting capability and selective tumor cytotoxicity, could elicit intratumor dendritic cells (mainly attributed to CQ) and tumor infiltrating CD8+ T cells, upon combining with PD-L1 therapeutic antibody, further induce significant anti-tumor effect. Collectively, the rationally designed nanoplatform, CC@LPF, could enhance tumor chemoimmunotherapy via deploying endogenous plus exogenous copper and remodeling tumor microenvironment.


Asunto(s)
Cobre , Inmunoterapia , Especies Reactivas de Oxígeno , Microambiente Tumoral , Microambiente Tumoral/efectos de los fármacos , Animales , Cobre/química , Cobre/farmacología , Ratones , Inmunoterapia/métodos , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Liposomas/química , Catálisis , Autofagia/efectos de los fármacos , Ácido Fólico/química , Ácido Fólico/farmacología , Humanos , Cloroquina/farmacología , Femenino , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
16.
Int J Nanomedicine ; 19: 8949-8970, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39246424

RESUMEN

Introduction: RNA interference (RNAi) stands as a widely employed gene interference technology, with small interfering RNA (siRNA) emerging as a promising tool for cancer treatment. However, the inherent limitations of siRNA, such as easy degradation and low bioavailability, hamper its efficacy in cancer therapy. To address these challenges, this study focused on the development of a nanocarrier system (HLM-N@DOX/R) capable of delivering both siRNA and doxorubicin for the treatment of breast cancer. Methods: The study involved a comprehensive investigation into various characteristics of the nanocarrier, including shape, diameter, Fourier transform infrared (FT-IR) spectroscopy, X-ray photoelectron spectroscopy (XPS), encapsulation efficiency, and drug loading. Subsequently, in vitro and in vivo studies were conducted on cytotoxicity, cellular uptake, cellular immunofluorescence, lysosome escape, and mouse tumor models to evaluate the efficacy of the nanocarrier in reversing tumor multidrug resistance and anti-tumor effects. Results: The results showed that HLM-N@DOX/R had a high encapsulation efficiency and drug loading capacity, and exhibited pH/redox dual responsive drug release characteristics. In vitro and in vivo studies showed that HLM-N@DOX/R inhibited the expression of P-gp by 80%, inhibited MDR tumor growth by 71% and eliminated P protein mediated multidrug resistance. Conclusion: In summary, HLM-N holds tremendous potential as an effective and targeted co-delivery system for DOX and P-gp siRNA, offering a promising strategy for overcoming MDR in breast cancer.


Asunto(s)
Neoplasias de la Mama , Doxorrubicina , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Liposomas , ARN Interferente Pequeño , Animales , Doxorrubicina/farmacología , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/administración & dosificación , Femenino , Liposomas/química , Ratones , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/química , ARN Interferente Pequeño/farmacocinética , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Células MCF-7 , Ratones Endogámicos BALB C , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Nanopartículas/química , Liberación de Fármacos , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Int J Nanomedicine ; 19: 8929-8947, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39246429

RESUMEN

Introduction: Cell death regulation holds a unique value in the field of cancer therapy. Recently, disulfidptosis has garnered substantial scientific attention. Previous studies have reported that sonodynamic therapy (SDT) based on reactive oxygen species (ROS) can regulate cancer cell death, achieving an limited anti-cancer effect. However, the integration of SDT with disulfidptosis as an anti-cancer strategy has not been extensively developed. In this study, we constructed an artificial membrane disulfidptosis sonosensitizer, specifically, a nanoliposome (SC@lip) coated with a combination of the chemotherapy medicine Sorafenib (Sora) and sonosensitizer Chlorin e6 (Ce6), to realize a one-stop enhanced SDT effect that induces disulfidptosis-like cancer cell death. Methods: Sorafenib and Ce6 were co-encapsulated into PEG-modified liposomes, and SC@Lip was constructed using a simple rotary evaporation phacoemulsification method. The cell phagocytosis, ROS generation ability, glutathione (GSH) depletion ability, lipid peroxidation (LPO), and disulfidptosis-like death mediated by SC@Lip under ultrasound (US) irradiation were evaluated. Based on a 4T1 subcutaneous tumor model, both the in vivo biological safety assessment and the efficacy of SDT were assessed. Results: SC@Lip exhibits high efficiency in cellular phagocytosis. After being endocytosed by 4T1 cells, abundant ROS were produced under SDT activation, and the cell survival rates were below 5%. When applied to a 4T1 subcutaneous tumor model, the enhanced SDT mediated by SC@Lip inhibited tumor growth and prolonged the survival time of mice. In vitro and in vivo experiments show that SC@Lip can enhance the SDT effect and trigger disulfidptosis-like cancer cell death, thus achieving anti-tumor efficacy both in vitro and in vivo. Conclusion: SC@Lip is a multifunctional nanoplatform with an artificial membrane, which can integrate the functions of sonosensitization and GSH depletion into a biocompatible nanoplatform, and can be used to enhance the SDT effect and promote disulfidptosis-like cancer cell death.


Asunto(s)
Clorofilidas , Peroxidación de Lípido , Liposomas , Porfirinas , Especies Reactivas de Oxígeno , Sorafenib , Terapia por Ultrasonido , Animales , Liposomas/química , Peroxidación de Lípido/efectos de los fármacos , Sorafenib/farmacología , Sorafenib/química , Terapia por Ultrasonido/métodos , Ratones , Línea Celular Tumoral , Especies Reactivas de Oxígeno/metabolismo , Porfirinas/química , Porfirinas/farmacología , Porfirinas/administración & dosificación , Femenino , Ratones Endogámicos BALB C , Nanopartículas/química , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Glutatión/metabolismo , Muerte Celular/efectos de los fármacos
18.
ACS Appl Bio Mater ; 7(9): 5784-5794, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39226406

RESUMEN

Hydrated dispersions containing equimolar mixtures of cationic and anionic amphiphiles, referred to as catanionic systems, exhibit synergistic physicochemical properties, and mixing single-chain cationic and anionic lipids can lead to the spontaneous formation of vesicles as well as other phase structures. In the present work, we have characterized two catanionic systems prepared by mixing N-acyltaurines (NATs) and sarcosine alkyl esters (SAEs) bearing 11 and 12 C atoms in the acyl/alkyl chains. Turbidimetric and isothermal titration calorimetric studies revealed that both NATs form equimolar complexes with SAEs having matching acyl/alkyl chains. The three-dimensional structure of the sarcosine lauryl ester (lauryl sarcosinate, LS)-N-lauroyltaurine (NLT) equimolar complex has been determined by single-crystal X-ray diffraction. The LS-NLT equimolar complex is stabilized by electrostatic attraction and multiple hydrogen bonds, including classical, strong N-H···O hydrogen bonds as well as several C-H···O hydrogen bonds between the two amphiphiles. DSC studies showed that both equimolar complexes show single sharp phase transitions. Transmission electron microscopy and dynamic light scattering studies have demonstrated that the LS-NLT catanionic complex assemblies yield stable medium-sized vesicles (diameter 280-350 nm). These liposomes were disrupted at high pH, suggesting that the designed catanionic complexes can be used to develop base-labile drug delivery systems. In vitro studies with these catanionic liposomes showed efficient entrapment (73% loading) and release of the anticancer drug 5-fluorouracil in the physiologically relevant pH range of 6.0-8.0. The release rate was highest at pH 8.0, reaching about 78%, 90%, and 100% drug release at 2, 6, and 12 h, respectively. These observations indicate that LS-NLT catanionic vesicles will be useful for designing drug delivery systems, particularly for targeting organs such as the colon, which are inherently at basic pH.


Asunto(s)
Materiales Biocompatibles , Fluorouracilo , Tamaño de la Partícula , Fluorouracilo/química , Estructura Molecular , Materiales Biocompatibles/química , Materiales Biocompatibles/síntesis química , Ensayo de Materiales , Cationes/química , Sarcosina/química , Sarcosina/análogos & derivados , Ésteres/química , Humanos , Liposomas/química
19.
Photochem Photobiol Sci ; 23(9): 1673-1685, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39237687

RESUMEN

Photodynamic therapy (PDT) is a promising alternative treatment for localized lesions and infections, utilizing reactive oxygen species (ROS) generated by photosensitizers (PS) upon light activation. Singlet oxygen (1O2) is a key ROS responsible for photodynamic damage. However, the effectiveness of PS in biological systems may not correlate with the efficiency of singlet oxygen generation in homogeneous solutions. This study investigated singlet oxygen generation and its decay in various cellular microenvironments using liposome and ARPE-19 cell models. Rose Bengal (RB), methylene blue (MB), and protoporphyrin IX (PpIX) were employed as selected PS. Lifetimes of singlet oxygen generated by the selected photosensitizers in different cellular compartments varied, indicating different quenching rates with singlet oxygen. RB, located near cell membranes, exhibited the highest phototoxicity and lipid/protein peroxidation, followed by PpIX, while MB showed minimal cytotoxicity in similar conditions. Singlet oxygen decay lifetimes provide insights into PS localization and potential phototoxicity, highlighting the importance of the lipid microenvironment in PDT efficacy, providing useful screening method prior to in vivo applications.


Asunto(s)
Liposomas , Azul de Metileno , Fotoquimioterapia , Fármacos Fotosensibilizantes , Protoporfirinas , Rosa Bengala , Oxígeno Singlete , Oxígeno Singlete/metabolismo , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Humanos , Rosa Bengala/farmacología , Rosa Bengala/química , Protoporfirinas/química , Protoporfirinas/farmacología , Protoporfirinas/metabolismo , Liposomas/química , Azul de Metileno/química , Azul de Metileno/farmacología , Supervivencia Celular/efectos de los fármacos , Línea Celular , Luz
20.
Photochem Photobiol Sci ; 23(9): 1757-1769, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39242437

RESUMEN

Porphyrazines (Pzs) are porphyrin derivatives that show potential application as photosensitizers for photodynamic therapy (PDT), but are still far less explored in the literature. In this work, we evaluate how the photophysics and phototoxicity of the octakis(trifluoromethylphenyl)porphyrazine (H2Pz) against tumor cells can be modulated by coordination with Mg(II), Zn(II), Cu(II) and Co(II) ions. Fluorescence and singlet oxygen quantum yields for the Pzs were measured in organic solvents and in soy phosphatidylcholine (PC) liposomes suspended in water. While H2Pz and the respective complexes with Cu(II) and Co(II) showed very low efficiency to fluoresce and to produce 1O2, the Mg(II) and Zn(II) complexes showed significantly higher quantum yields in organic solvents. The fluorescence of these two Pzs in the liposomes was sensitive to the fluidity of the membrane, showing potential use as viscosity markers. The cytotoxicity of the compounds was tested in HaCaT (normal) and A431 (tumor) cells using soy PC liposomes as drug carriers. Despite the low 1O2 quantum yields in water, the Mg(II) and Zn(II) complexes showed IC50 values against A431 cells in the nanomolar range when activated with low doses of red LED light. Their phototoxicity was ca. three times higher for the tumor cells compared to the normal ones, showing promising application as photosensitizers for PDT protocols. Considering that H2Pz and the respective Co(II) and Cu(II) complexes were practically non-phototoxic to the cells, we demonstrate the importance of the central metal ion in the modulation of the photodynamic activity of porphyrazines.


Asunto(s)
Liposomas , Fármacos Fotosensibilizantes , Porfirinas , Humanos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Porfirinas/química , Porfirinas/farmacología , Liposomas/química , Fotoquimioterapia , Oxígeno Singlete/metabolismo , Oxígeno Singlete/química , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/síntesis química , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Zinc/química , Zinc/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Iones/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA