Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.329
Filtrar
1.
Int J Nanomedicine ; 19: 6757-6776, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38983132

RESUMEN

Glioma is a primary malignant tumor in the central nervous system. In recent years, the treatment of glioma has developed rapidly, but the overall survival of glioma patients has not significantly improved. Due to the presence of the blood-brain barrier and intracranial tumor barrier, many drugs with good effects to cure glioma in vitro cannot be accurately transported to the corresponding lesions. In order to enable anti-tumor drugs to overcome the barriers and target glioma, nanodrug delivery systems have emerged recently. It is gratifying that liposomes, as a multifunctional nanodrug delivery carrier, which can be compatible with hydrophilic and hydrophobic drugs, easily functionalized by various targeted ligands, biodegradable, and hypoimmunogenic in vivo, has become a quality choice to solve the intractable problem of glioma medication. Therefore, we focused on the liposome nanodrug delivery system, and summarized its current research progress in glioma. Hopefully, this review may provide new ideas for the research and development of liposome-based nanomaterials for the clinical treatment of glioma.


Asunto(s)
Antineoplásicos , Barrera Hematoencefálica , Neoplasias Encefálicas , Glioma , Liposomas , Nanoestructuras , Glioma/tratamiento farmacológico , Liposomas/química , Humanos , Neoplasias Encefálicas/tratamiento farmacológico , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Antineoplásicos/química , Antineoplásicos/administración & dosificación , Animales , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Nanomedicina/métodos , Portadores de Fármacos/química
2.
Int J Nanomedicine ; 19: 6857-6893, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39005956

RESUMEN

Periodontitis is a disease of inflammation that affects the tissues supporting the periodontium. It is triggered by an immunological reaction of the gums to plaque, which leads to the destruction of periodontal attachment structures. Periodontitis is one of the most commonly recognized dental disorders in the world and a major factor in the loss of adult teeth. Scaling and root planing remain crucial for managing patients with persistent periodontitis. Nevertheless, exclusive reliance on mechanical interventions like periodontal surgery, extractions, and root planning is insufficient to halt the progression of periodontitis. In response to the problem of bacterial resistance, some researchers are committed to finding alternative therapies to antibiotics. In addition, some scholars focus on finding new materials to provide a powerful microenvironment for periodontal tissue regeneration and promote osteogenic repair. Nanoparticles possess distinct therapeutic qualities, including exceptional antibacterial, anti-inflammatory, and antioxidant properties, immunomodulatory capacities, and the promotion of bone regeneration ability, which made them can be used for the treatment of periodontitis. However, there are many problems that limit the clinical translation of nanoparticles, such as toxic accumulation in cells, poor correlation between in vitro and in vivo, and poor animal-to-human transmissibility. In this paper, we review the present researches on nanoparticles in periodontitis treatment from the perspective of three main categories: inorganic nanoparticles, organic nanoparticles, and nanocomposites (including nanofibers, hydrogels, and membranes). The aim of this review is to provide a comprehensive and recent update on nanoparticles-based therapies for periodontitis. The conclusion section summarizes the opportunities and challenges in the design and clinical translation of nanoparticles for the treatment of periodontitis.


Asunto(s)
Nanopartículas , Periodontitis , Humanos , Periodontitis/terapia , Periodontitis/tratamiento farmacológico , Nanopartículas/química , Animales , Antibacterianos/química , Antibacterianos/farmacología , Antiinflamatorios/química , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Nanocompuestos/química , Nanocompuestos/uso terapéutico , Nanomedicina/métodos
3.
Theranostics ; 14(9): 3486-3508, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38948064

RESUMEN

Rationale: Device implantation frequently triggers cardiac remodeling and fibrosis, with monocyte-driven inflammatory responses precipitating arrhythmias. This study investigates the role of m6A modification enzymes METTL3 and METTL14 in these responses and explores a novel therapeutic strategy targeting these modifications to mitigate cardiac remodeling and fibrosis. Methods: Peripheral blood mononuclear cells (PBMCs) were collected from patients with ventricular septal defects (VSD) who developed conduction blocks post-occluder implantation. The expression of METTL3 and METTL14 in PBMCs was measured. METTL3 and METTL14 deficiencies were induced to evaluate their effect on angiotensin II (Ang II)-induced myocardial inflammation and fibrosis. m6A modifications were analyzed using methylated RNA immunoprecipitation followed by quantitative PCR. NF-κB pathway activity and levels of monocyte migration and fibrogenesis markers (CXCR2 and TGF-ß1) were assessed. An erythrocyte microvesicle-based nanomedicine delivery system was developed to target activated monocytes, utilizing the METTL3 inhibitor STM2457. Cardiac function was evaluated via echocardiography. Results: Significant upregulation of METTL3 and METTL14 was observed in PBMCs from patients with VSD occluder implantation-associated persistent conduction block. Deficiencies in METTL3 and METTL14 significantly reduced Ang II-induced myocardial inflammation and fibrosis by decreasing m6A modification on MyD88 and TGF-ß1 mRNAs. This disruption reduced NF-κB pathway activation, lowered CXCR2 and TGF-ß1 levels, attenuated monocyte migration and fibrogenesis, and alleviated cardiac remodeling. The erythrocyte microvesicle-based nanomedicine delivery system effectively targeted inflamed cardiac tissue, reducing inflammation and fibrosis and improving cardiac function. Conclusion: Inhibiting METTL3 and METTL14 in monocytes disrupts the NF-κB feedback loop, decreases monocyte migration and fibrogenesis, and improves cardiac function. Targeting m6A modifications of monocytes with STM2457, delivered via erythrocyte microvesicles, reduces inflammation and fibrosis, offering a promising therapeutic strategy for cardiac remodeling associated with device implantation.


Asunto(s)
Fibrosis , Metiltransferasas , Monocitos , FN-kappa B , Humanos , Metiltransferasas/metabolismo , Metiltransferasas/genética , Monocitos/metabolismo , Masculino , Animales , FN-kappa B/metabolismo , Eritrocitos/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Femenino , Metilación , Ratones , Factor de Crecimiento Transformador beta1/metabolismo , Micropartículas Derivadas de Células/metabolismo , Leucocitos Mononucleares/metabolismo , Angiotensina II/metabolismo , Receptores de Interleucina-8B/metabolismo , Receptores de Interleucina-8B/genética , Remodelación Ventricular , Miocardio/metabolismo , Miocardio/patología , Nanomedicina/métodos
4.
J Transl Med ; 22(1): 648, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38987805

RESUMEN

Glioma is the most common malignant tumor in central nervous system, with significant health burdens to patients. Due to the intrinsic characteristics of glioma and the lack of breakthroughs in treatment modalities, the prognosis for most patients remains poor. This results in a heavy psychological and financial load worldwide. In recent years, cannabidiol (CBD) has garnered widespread attention and research due to its anti-tumoral, anti-inflammatory, and neuroprotective properties. This review comprehensively summarizes the preclinical and clinical research on the use of CBD in glioma therapy, as well as the current status of nanomedicine formulations of CBD, and discusses the potential and challenges of CBD in glioma therapy in the future.


Asunto(s)
Cannabidiol , Glioma , Cannabidiol/uso terapéutico , Cannabidiol/farmacología , Humanos , Glioma/tratamiento farmacológico , Glioma/patología , Animales , Investigación Biomédica Traslacional , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Nanomedicina/métodos
5.
Int J Mol Sci ; 25(13)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-39000136

RESUMEN

Nanomedicine could improve the treatment of diabetes by exploiting various therapeutic mechanisms through the use of suitable nanoformulations. For example, glucose-sensitive nanoparticles can release insulin in response to high glucose levels, mimicking the physiological release of insulin. Oral nanoformulations for insulin uptake via the gut represent a long-sought alternative to subcutaneous injections, which cause pain, discomfort, and possible local infection. Nanoparticles containing oligonucleotides can be used in gene therapy and cell therapy to stimulate insulin production in ß-cells or ß-like cells and modulate the responses of T1DM-associated immune cells. In contrast, viral vectors do not induce immunogenicity. Finally, in diabetic wound healing, local delivery of nanoformulations containing regenerative molecules can stimulate tissue repair and thus provide a valuable tool to treat this diabetic complication. Here, we describe these different approaches to diabetes treatment with nanoformulations and their potential for clinical application.


Asunto(s)
Diabetes Mellitus , Nanomedicina , Nanopartículas , Humanos , Nanomedicina/métodos , Animales , Diabetes Mellitus/tratamiento farmacológico , Nanopartículas/química , Terapia Genética/métodos , Insulina/metabolismo , Hipoglucemiantes/uso terapéutico , Hipoglucemiantes/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos
6.
AAPS J ; 26(4): 74, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38955936

RESUMEN

The paper highlights the necessity for a robust regulatory framework for assessing nanomedicines and their off-patent counterparts, termed as nanosimilar, which could be considered as 'similar' to the prototype nanomedicine,based on essential criteria describing the 'similarity'. The term 'similarity' should be focused on criteria that describe nanocarriers, encompassing their physicochemical, thermodynamic, morphological, and biological properties, including surface interactions and pharmacokinetics. Nanocarriers can be regarded as advanced self-assembled excipients (ASAEs) due to their complexity and chaotic behavior and should be evaluated by using essential criteria in order for off-patent nanomedicines be termed as nanosimilars, from a regulatory perspective. Collaboration between the pharmaceutical industry, regulatory bodies, and artificial intelligence (AI) startups is pivotal for the precise characterization and approval processes for nanomedicines and nanosimilars and embracing innovative tools and terminology facilitates the development of a sustainable regulatory framework, ensuring safety and efficacy. This crucial shift toward precision R&D practices addresses the complexity inherent in nanocarriers, paving the way for therapeutic advancements with economic benefits.


Asunto(s)
Nanomedicina , Nanomedicina/legislación & jurisprudencia , Nanomedicina/métodos , Humanos , Biosimilares Farmacéuticos/administración & dosificación , Biosimilares Farmacéuticos/farmacocinética , Inteligencia Artificial , Nanopartículas , Industria Farmacéutica/legislación & jurisprudencia , Aprobación de Drogas/legislación & jurisprudencia , Portadores de Fármacos/química
7.
Int J Nanomedicine ; 19: 6619-6641, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38975321

RESUMEN

The high malignant degree and poor prognosis of pancreatic cancer (PC) pose severe challenges to the basic research and clinical translation of next-generation therapies. The rise of immunotherapy has improved the treatment of a variety of solid tumors, while the application in PC is highly restricted by the challenge of immunosuppressive tumor microenvironment. The latest progress of nanotechnology as drug delivery platform and immune adjuvant has improved drug delivery in a variety of disease backgrounds and enhanced tumor therapy based on immunotherapy. Based on the immune loop of PC and the status quo of clinical immunotherapy of tumors, this article discussed and critically analyzed the key transformation difficulties of immunotherapy adaptation to the treatment of PC, and then proposed the rational design strategies of new nanocarriers for drug delivery and immune regulation, especially the design of combined immunotherapy. This review also put forward prospective views on future research directions, so as to provide information for the new means of clinical treatment of PC combined with the next generation of nanotechnology and immunotherapy.


Asunto(s)
Inmunoterapia , Neoplasias Pancreáticas , Microambiente Tumoral , Humanos , Inmunoterapia/métodos , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Animales , Nanotecnología/métodos , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química , Nanopartículas/uso terapéutico , Nanomedicina/métodos
8.
Int J Nanomedicine ; 19: 6731-6756, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38979531

RESUMEN

Osteoarthritis (OA) is the most common degenerative joint disease, affecting more than 595 million people worldwide. Nanomaterials possess superior physicochemical properties and can influence pathological processes due to their unique structural features, such as size, surface interface, and photoelectromagnetic thermal effects. Unlike traditional OA treatments, which suffer from short half-life, low stability, poor bioavailability, and high systemic toxicity, nanotherapeutic strategies for OA offer longer half-life, enhanced targeting, improved bioavailability, and reduced systemic toxicity. These advantages effectively address the limitations of traditional therapies. This review aims to inspire researchers to develop more multifunctional nanomaterials and promote their practical application in OA treatment.


Asunto(s)
Nanoestructuras , Osteoartritis , Osteoartritis/tratamiento farmacológico , Osteoartritis/terapia , Humanos , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Animales , Nanomedicina/métodos , Disponibilidad Biológica
9.
Nanotheranostics ; 8(4): 473-496, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38961885

RESUMEN

Cardiotoxicity, the often-overlooked second leading cause of death in cancer patients, has been associated with certain anticancer drugs. These drugs can induce cardiac damage through various pathways, and their adverse effects on the heart are not fully understood. Cardiotoxicity is a major issue in cancer treatment, particularly with chemotherapeutics, because it can cause cardiac dysfunction such as hypotension, heart failure, and even death. Doxorubicin, 5-fluorouracil, and trastuzumab, all of which are very potent anticancer drugs, are known to cause cardiotoxicity. When it comes to lowering cardiotoxicity and alleviating the harmful effects of chemotherapy medications, nanomedicine has the potential to transport therapeutic molecules. Nanotheranostics offers novel options for identifying and treating cardiotoxicity resulting from a wide range of substances, including anticancer medications. Additionally, theranostics platforms such as micellar systems, carbon-based nanomedicine, solid lipid nanoparticles, polymeric nanoparticles, and liposomes can transport chemotherapeutic medications while minimising their cardiotoxicity. The present level of understanding of the molecular and cellular processes that lead to cardiotoxicity in reaction to both traditional chemotherapy and targeted drug delivery systems is summarised in this article. This review delves into nanomedicine and nanotheranostics, with an emphasis on reducing anticancer medication-induced cardiac toxicity. Nanotheranostics provide potential solutions for early diagnosis and tailored therapy of heart injury by combining diagnostic and therapeutic capabilities into nanomedicine.


Asunto(s)
Antineoplásicos , Cardiotoxicidad , Nanomedicina , Nanomedicina Teranóstica , Humanos , Antineoplásicos/efectos adversos , Antineoplásicos/química , Cardiotoxicidad/etiología , Nanomedicina/métodos , Nanomedicina Teranóstica/métodos , Animales , Cardiopatías/inducido químicamente , Neoplasias/tratamiento farmacológico , Nanopartículas/química
11.
Molecules ; 29(11)2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38893373

RESUMEN

Developing clinically meaningful nanomedicines for cancer therapy requires the drugs to be effective, safe, simple, cheap, and easy to store. In the present work, we report that a simple cationic Fe(III)-rich salt of [FeIIICl(TMPPH2)][FeIIICl4]2 (Fe-TMPP) exhibits a superior anticancer performance on a broad spectrum of cancer cell lines, including breast, colorectal cancer, liver, pancreatic, prostate, and gastric cancers, with half maximal inhibitory concentration (IC50) values in the range of 0.098-3.97 µM (0.066-2.68 µg mL-1), comparable to the best-reported medicines. Fe-TMPP can form stand-alone nanoparticles in water without the need for extra surface modification or organic-solvent-assisted antisolvent precipitation. Critically, Fe-TMPP is TME-responsive (TME = tumor microenvironment), and can only elicit its function in the TME with overexpressed H2O2, converting H2O2 to the cytotoxic •OH to oxidize the phospholipid of the cancer cell membrane, causing ferroptosis, a programmed cell death process of cancer cells.


Asunto(s)
Antineoplásicos , Ferroptosis , Nanomedicina , Humanos , Ferroptosis/efectos de los fármacos , Línea Celular Tumoral , Nanomedicina/métodos , Antineoplásicos/farmacología , Antineoplásicos/química , Nanopartículas/química , Compuestos Férricos/química , Microambiente Tumoral/efectos de los fármacos , Peróxido de Hidrógeno/química , Peróxido de Hidrógeno/farmacología , Supervivencia Celular/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/patología
12.
Molecules ; 29(11)2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38893460

RESUMEN

There is a myriad of diseases that plague the world ranging from infectious, cancer and other chronic diseases with varying interventions. However, the dynamism of causative agents of infectious diseases and incessant mutations accompanying other forms of chronic diseases like cancer, have worsened the treatment outcomes. These factors often lead to treatment failure via different drug resistance mechanisms. More so, the cost of developing newer drugs is huge. This underscores the need for a paradigm shift in the drug delivery approach in order to achieve desired treatment outcomes. There is intensified research in nanomedicine, which has shown promises in improving the therapeutic outcome of drugs at preclinical stages with increased efficacy and reduced toxicity. Regardless of the huge benefits of nanotechnology in drug delivery, challenges such as regulatory approval, scalability, cost implication and potential toxicity must be addressed via streamlining of regulatory hurdles and increased research funding. In conclusion, the idea of nanotechnology in drug delivery holds immense promise for optimizing therapeutic outcomes. This work presents opportunities to revolutionize treatment strategies, providing expert opinions on translating the huge amount of research in nanomedicine into clinical benefits for patients with resistant infections and cancer.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanomedicina , Nanoestructuras , Humanos , Nanoestructuras/química , Nanomedicina/métodos , Neoplasias/tratamiento farmacológico , Animales , Nanotecnología/métodos
13.
Int J Mol Sci ; 25(11)2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38892023

RESUMEN

We launched this Special Issue amidst the COVID-19 pandemic, spurred by the growing interest in nanotherapeutic formulations for delivering SARS-CoV-2 viral messenger Ribonucleic Acid (mRNA) vaccines [...].


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Nanomedicina , SARS-CoV-2 , Humanos , Nanomedicina/métodos , COVID-19/virología , COVID-19/prevención & control
14.
ACS Appl Mater Interfaces ; 16(26): 33070-33080, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38904394

RESUMEN

Nanomaterials have been extensively exploited in tumor treatment, leading to numerous innovative strategies for cancer therapy. While nanomedicines present immense potential, their application in cancer therapy is characterized by significant complexity and unpredictability, especially regarding biocompatibility and anticancer efficiency. These considerations underscore the essential need for the development of ex vivo research models, which provide invaluable insights and understanding into the biosafety and efficacy of nanomedicines in oncology. Fortunately, the emergence of organoid technology offers a novel approach to the preclinical evaluation of the anticancer efficacy of nanomedicines in vitro. Hence, in this study, we constructed intestine and hepatocyte organoid models (Intestine-orgs and Hep-orgs) for assessing intestinal and hepatic toxicity at the microtissue level. We utilized three typical metal-organic frameworks (MOFs), ZIF-8, ZIF-67, and MIL-125, as nanomedicines to further detect their interactions with organoids. Subsequently, the MIL-125 with biocompatibility loaded methotrexate (MTX), forming the nanomedicine (MIL-125-PEG-MTX), indicated a high loading efficiency (82%) and a well-release capability in an acid microenvironment. More importantly, the anticancer effect of the nanomedicine was investigated using an in vitro patient-derived organoids (PDOs) model, achieving inhibition rates of 48% and 78% for PDO-1 and PDO-2, respectively, demonstrating that PDOs could predict clinical response and facilitate prospective therapeutic selection. These achievements presented great potential for organoid-based ex vivo models for nano theragnostic evaluation in biosafety and function.


Asunto(s)
Estructuras Metalorgánicas , Nanomedicina , Organoides , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Humanos , Organoides/efectos de los fármacos , Organoides/metabolismo , Nanomedicina/métodos , Metotrexato/farmacología , Metotrexato/química , Metotrexato/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Intestinos/efectos de los fármacos , Intestinos/patología , Animales
15.
J Nanobiotechnology ; 22(1): 354, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38902775

RESUMEN

Fundus neovascularization diseases are a series of blinding eye diseases that seriously impair vision worldwide. Currently, the means of treating these diseases in clinical practice are continuously evolving and have rapidly revolutionized treatment opinions. However, key issues such as inadequate treatment effectiveness, high rates of recurrence, and poor patient compliance still need to be urgently addressed. Multifunctional nanomedicine can specifically respond to both endogenous and exogenous microenvironments, effectively deliver drugs to specific targets and participate in activities such as biological imaging and the detection of small molecules. Nano-in-micro (NIM) delivery systems such as metal, metal oxide and up-conversion nanoparticles (NPs), quantum dots, and carbon materials, have shown certain advantages in overcoming the presence of physiological barriers within the eyeball and are widely used in the treatment of ophthalmic diseases. Few studies, however, have evaluated the efficacy of NIM delivery systems in treating fundus neovascular diseases (FNDs). The present study describes the main clinical treatment strategies and the adverse events associated with the treatment of FNDs with NIM delivery systems and summarizes the anatomical obstacles that must be overcome. In this review, we wish to highlight the principle of intraocular microenvironment normalization, aiming to provide a more rational approach for designing new NIM delivery systems to treat specific FNDs.


Asunto(s)
Sistemas de Liberación de Medicamentos , Humanos , Animales , Sistemas de Liberación de Medicamentos/métodos , Neovascularización Patológica/tratamiento farmacológico , Fondo de Ojo , Puntos Cuánticos/química , Nanopartículas Multifuncionales/química , Neovascularización Retiniana/tratamiento farmacológico , Nanomedicina/métodos , Nanopartículas/química
16.
Biomed Pharmacother ; 176: 116904, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38878686

RESUMEN

Globally, cancer is a serious health problem. It is unfortunate that current anti-cancer strategies are insufficiently specific and damage the normal tissues. There's urgent need for development of new anti-cancer strategies. More recently, increasing attention has been paid to the new application of ferroptosis and nano materials in cancer research. Ferroptosis, a condition characterized by excessive reactive oxygen species-induced lipid peroxidation, as a new programmed cell death mode, exists in the process of a number of diseases, including cancers, neurodegenerative disease, cerebral hemorrhage, liver disease, and renal failure. There is growing evidence that inducing ferroptosis has proven to be an effective strategy against a variety of chemo-resistant cancer cells. Nano-drug delivery system based on nanotechnology provides a highly promising platform with the benefits of precise control of drug release and reduced toxicity and side effects. This paper reviews the latest advances of combination therapy strategies based on biomedical nanotechnology induced ferroptosis for cancer therapeutics. Given the new chances and challenges in this emerging area, we need more attention to the combination of nanotechnology and ferroptosis in the treatment of cancer in the future.


Asunto(s)
Ferroptosis , Neoplasias , Ferroptosis/efectos de los fármacos , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Animales , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Nanopartículas , Nanotecnología/métodos , Sistema de Administración de Fármacos con Nanopartículas , Sistemas de Liberación de Medicamentos/métodos , Terapia Combinada , Especies Reactivas de Oxígeno/metabolismo , Nanomedicina/métodos
17.
J Nanobiotechnology ; 22(1): 365, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918839

RESUMEN

Bacteriophages (phages) represent a unique category of viruses with a remarkable ability to selectively infect host bacteria, characterized by their assembly from proteins and nucleic acids. Leveraging their exceptional biological properties and modifiable characteristics, phages emerge as innovative, safe, and efficient delivery vectors. The potential drawbacks associated with conventional nanocarriers in the realms of drug and gene delivery include a lack of cell-specific targeting, cytotoxicity, and diminished in vivo transfection efficiency. In contrast, engineered phages, when employed as cargo delivery vectors, hold the promise to surmount these limitations and attain enhanced delivery efficacy. This review comprehensively outlines current strategies for the engineering of phages, delineates the principal types of phages utilized as nanocarriers in drug and gene delivery, and explores the application of phage-based delivery systems in disease therapy. Additionally, an incisive analysis is provided, critically examining the challenges confronted by phage-based delivery systems within the domain of nanotechnology. The primary objective of this article is to furnish a theoretical reference that contributes to the reasoned design and development of potent phage-based delivery systems.


Asunto(s)
Bacteriófagos , Sistemas de Liberación de Medicamentos , Nanomedicina , Bacteriófagos/genética , Humanos , Nanomedicina/métodos , Sistemas de Liberación de Medicamentos/métodos , Animales , Técnicas de Transferencia de Gen , Portadores de Fármacos/química , Nanopartículas/química , Nanotecnología/métodos
18.
J Nanobiotechnology ; 22(1): 364, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38915007

RESUMEN

Photothermal therapy (PTT) is a promising cancer treatment method due to its ability to induce tumor-specific T cell responses and enhance therapeutic outcomes. However, incomplete PTT can leave residual tumors that often lead to new metastases and decreased patient survival in clinical scenarios. This is primarily due to the release of ATP, a damage-associated molecular pattern that quickly transforms into the immunosuppressive metabolite adenosine by CD39, prevalent in the tumor microenvironment, thus promoting tumor immune evasion. This study presents a photothermal nanomedicine fabricated by electrostatic adsorption among the Fe-doped polydiaminopyridine (Fe-PDAP), indocyanine green (ICG), and CD39 inhibitor sodium polyoxotungstate (POM-1). The constructed Fe-PDAP@ICG@POM-1 (FIP) can induce tumor PTT and immunogenic cell death when exposed to a near-infrared laser. Significantly, it can inhibit the ATP-adenosine pathway by dual-directional immunometabolic regulation, resulting in increased ATP levels and decreased adenosine synthesis, which ultimately reverses the immunosuppressive microenvironment and increases the susceptibility of immune checkpoint blockade (aPD-1) therapy. With the aid of aPD-1, the dual-directional immunometabolic regulation strategy mediated by FIP can effectively suppress/eradicate primary and distant tumors and evoke long-term solid immunological memory. This study presents an immunometabolic control strategy to offer a salvage option for treating residual tumors following incomplete PTT.


Asunto(s)
Inmunoterapia , Nanomedicina , Terapia Fototérmica , Microambiente Tumoral , Animales , Terapia Fototérmica/métodos , Inmunoterapia/métodos , Ratones , Nanomedicina/métodos , Microambiente Tumoral/efectos de los fármacos , Línea Celular Tumoral , Humanos , Verde de Indocianina/química , Verde de Indocianina/farmacología , Neoplasias/terapia , Adenosina Trifosfato/metabolismo , Adenosina/farmacología , Adenosina/química , Ratones Endogámicos C57BL , Apirasa/metabolismo , Femenino , Fototerapia/métodos
19.
Nano Lett ; 24(27): 8217-8231, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38848540

RESUMEN

Theranostic medicine combines diagnostics and therapeutics, focusing on solid tumors at minimal doses. Optically activated photosensitizers are significant examples owing to their photophysical and chemical properties. Several optotheranostics have been tested that convert light to imaging signals, therapeutic radicals, and heat. Upon light exposure, conjugated photosensitizers kill tumor cells by producing reactive oxygen species and heat or by releasing cancer antigens. Despite clinical trials, these molecularly conjugated photosensitizers require protection from their surroundings and a localized direction for site-specific delivery during blood circulation. Therefore, cell membrane biomimetic ghosts have been proposed for precise and safe delivery of these optically active large molecules, which are clinically relevant because of their biocompatibility, long circulation time, bypass of immune cell recognition, and targeting ability. This review focuses on the role of biomimetic nanoparticles in the treatment and diagnosis of tumors through light-mediated diagnostics and therapy, providing insights into their preclinical and clinical status.


Asunto(s)
Materiales Biomiméticos , Neoplasias , Fármacos Fotosensibilizantes , Nanomedicina Teranóstica , Humanos , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Fármacos Fotosensibilizantes/uso terapéutico , Fármacos Fotosensibilizantes/química , Materiales Biomiméticos/química , Materiales Biomiméticos/uso terapéutico , Nanopartículas/química , Nanopartículas/uso terapéutico , Animales , Biomimética , Nanomedicina/métodos
20.
Nano Lett ; 24(27): 8351-8360, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38916238

RESUMEN

Targeting telomere maintenance has emerged as a promising strategy for hepatocellular carcinoma (HCC) treatment. However, given the duality of the telomere-telomerase axis in telomere maintenance, a comprehensive strategy is urgently needed. Herein, we develop a poly(amino acid) (D-PAAs)-based strategy for spatiotemporal codelivery of telomerase inhibitor, BIBR1523, and AKT inhibitor, isobavachalcone. By leveraging D-PAAs' modifiability, we synthesize polymer-inhibitor conjugates (PB and PI) and a folic acid-decorated tumor-targeting vector (PF). These building blocks undergo micellization to fabricate a codelivery nanomedicine (P-BI@P-FA) by exploiting D-PAAs' noncovalent assembly. P-BI@P-FA improves the pharmacokinetics, tumor selectivity, and bioavailability of small molecule inhibitors and initiates a dual telomere-specific inhibition by combining telomerase deactivation with telomere disruption. Furthermore, a hybrid tumor-targeting magnetic nanosystem is designed using D-PAAs and manganese dioxide to showcase magnetic resonance imaging capacities. Our D-PAAs-based strategy addresses the pressing need for telomere-specific HCC treatment while allowing for diagnostic application, presenting a promising avenue for nanomedicine design.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Imagen por Resonancia Magnética , Nanomedicina , Telomerasa , Telómero , Carcinoma Hepatocelular/diagnóstico por imagen , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Telomerasa/antagonistas & inhibidores , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/tratamiento farmacológico , Humanos , Nanomedicina/métodos , Telómero/metabolismo , Imagen por Resonancia Magnética/métodos , Animales , Ratones , Línea Celular Tumoral , Aminoácidos/química , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA