Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Molecules ; 26(19)2021 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-34641524

RESUMEN

Photothermal therapy (PTT) is one of the most promising cancer treatment methods because hyperthermal effects and immunogenic cell death via PTT are destructive to cancer. However, PTT requires photoabsorbers that absorb near-infrared (NIR) light with deeper penetration depth in the body and effectively convert light into heat. Gold nanoparticles have various unique properties which are suitable for photoabsorbers, e.g., controllable optical properties and easy surface modification. We developed gold nanodot swarms (AuNSw) by creating small gold nanoparticles (sGNPs) in the presence of hydrophobically-modified glycol chitosan. The sGNPs assembled with each other through their interaction with amine groups of glycol chitosan. AuNSw absorbed 808-nm laser and increased temperature to 55 °C. In contrast, AuNSw lost its particle structure upon exposure to thiolated molecules and did not convert NIR light into heat. In vitro studies demonstrated the photothermal effect and immunogenic cell death after PTT with AuNSW. After intratumoral injection of AuNSw with laser irradiation, tumor growth of xenograft mouse models was depressed. We found hyperthermal damage and immunogenic cell death in tumor tissues through histological and biochemical analyses. Thiol-responsive AuNSw showed feasibility for PTT, with advanced functionality in the tumor microenvironment.


Asunto(s)
Quitosano/química , Nanopartículas del Metal/química , Terapia Fototérmica/métodos , Animales , Oro/química , Humanos , Terapia por Láser , Masculino , Nanopartículas del Metal/uso terapéutico , Ratones Endogámicos BALB C , Neoplasias/terapia , Tamaño de la Partícula , Terapia Fototérmica/instrumentación , Compuestos de Sulfhidrilo/química , Temperatura , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Bioconjug Chem ; 31(4): 1012-1024, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32163277

RESUMEN

Enzyme-activatable anticancer prodrugs are modified medications that are composed of an anticancer drug, cleavable linker, and functional moiety. The purpose of such a prodrug structure is to generate multipurpose functions that traditional drugs cannot perform and to reduce the toxicity of conventional anticancer drugs by the mask of the cleavable linker. Once the cleavable linker is degraded via a specific chemical reaction in the cancer microenvironment, the cytotoxicity of the degraded prodrugs is selectively recovered. Among many factors that cleave the linker, we focus on the overexpressed enzymes in cancer. Because of the selective enzymatic degradation of the cleavable linker and the high local concentration of specific enzymes in cancer, the enzyme-activatable prodrugs could show low toxicity in normal tissues, while showing comparable anticancer effect in tumors. In addition, some prodrugs provide additional features, such as cancer imaging, drug release monitoring, tumor targeting, and enhanced stability, which conventional anticancer drugs cannot possess. In this review, we summarize currently developed enzyme-activatable prodrugs according to their activating enzymes, and categorize them by their additional functions, e.g. targeting, imaging, and delivery. This summary of enzyme-activatable prodrugs may help in the design of anticancer prodrugs, and in the establishment of a personalized cancer treatment strategy.


Asunto(s)
Enzimas/metabolismo , Terapia Molecular Dirigida/métodos , Neoplasias/tratamiento farmacológico , Profármacos/metabolismo , Animales , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Humanos , Neoplasias/metabolismo
3.
Ann Neurol ; 73(5): 617-25, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23495101

RESUMEN

OBJECTIVE: Advancing the understanding and management of thromboembolic stroke requires simple and robust new methods that would be useful for the in vivo assessment of thrombus burden/distribution and for characterizing its evolution in a prompt and quantitative manner. METHODS: Animals (n=127) with experimental models of thrombosis were imaged with microcomputed tomography 5 minutes (and/or ~3 weeks) after intravenous injection of glycol chitosan (GC) gold nanoparticles (AuNPs). RESULTS: Nanoparticles accumulated in the thrombus, allowing computed tomography visualization of both the presence and extent of primary and recurrent thrombi in mouse carotid arteries without a single failure of detection. Nanoparticle thrombus imaging was also effective in monitoring the therapeutic efficacy of thrombolysis (n=118 tissue plasminogen activator [tPA] therapies). Thrombus evolution (either spontaneous or post-tPA) could be mapped at high resolution in both space and time. Due to a long circulating half-life, GC-AuNPs remain available for entrapment into fibrin matrix for an extended period of time (up to 3 weeks), allowing repetition or ongoing monitoring of thrombogenesis and thrombolysis. INTERPRETATION: This is the first report on a hyperacute direct thrombus imaging technique using thrombus-seeking AuNPs and computed tomography. When translated into stroke practice, the thrombus imaging may allow us to advance to personalized thrombolytic therapy by demonstrating thrombus burden, distribution, and character in a prompt and quantitative manner. Further study into this area is indicated.


Asunto(s)
Oro/uso terapéutico , Nanopartículas , Trombosis/diagnóstico por imagen , Tomografía Computarizada por Rayos X , Animales , Animales Recién Nacidos , Anticolesterolemiantes/uso terapéutico , Mapeo Encefálico , Quitosano/uso terapéutico , Modelos Animales de Enfermedad , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Trombosis/tratamiento farmacológico , Factores de Tiempo , Activador de Tejido Plasminógeno/uso terapéutico
4.
Pharm Res ; 31(6): 1418-25, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23934255

RESUMEN

PURPOSE: The application of gold nanoparticles (AuNPs) in biomedical field was limited due to the low stability in the biological condition. Herein, to enhance stability and tumor targeting ability of AuNPs, their surface was modified with biocompatible glycol chitosan (GC) and the in vivo biodistribution of GC coated AuNPs (GC-AuNPs) were studied through computed tomography (CT). METHODS: Polymer-coated gold nanoparticles were produced using GC as a reducing agent and a stabilizer. Their feasibility in biomedical application was explored through CT in tumor-bearing mice. RESULTS: Stability of gold nanoparticles increased in the physiological condition due to the GC coating layer on the surface. Tomographic images of tumor were successfully obtained in the tumor-xenografted animal model when the GC-AuNPs were used as a CT contrast agent. The tumor targeting property of the gold nanoparticles was due to the properties of GC because GC-AuNPs were accumulated in the tumor, while most of heparin-coated nanoparticles were found in the liver and spleen. CONCLUSIONS: The polymer properties on the surface played an important role in the behavior of gold nanoparticles in the biological condition and the enhanced stability and tumor targeting property of nanoparticles were inherited from GC on the surface.


Asunto(s)
Materiales Biocompatibles , Quitosano/química , Medios de Contraste/química , Oro/química , Nanopartículas del Metal/química , Neoplasias/diagnóstico , Animales , Supervivencia Celular/efectos de los fármacos , Quitosano/toxicidad , Medios de Contraste/farmacocinética , Humanos , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/secundario , Ratones , Neoplasias/patología , Radiografía , Distribución Tisular
5.
Pharm Res ; 31(12): 3371-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24879466

RESUMEN

PURPOSE: To develop an MRI/optical multimodal imaging probe based on dye-conjugated iron oxide/silica core/shell nanoparticle, and investigate the distance-dependent fluorescence quenching through careful control of the distance between the iron oxide core and fluorescent dyes. METHODS: Different size of core/shell nanoparticles were prepared by varying the silica shell width. PEGylation on the surface of silica shell was followed to improve the stability of particles in the physiological condition. In vitro cytotoxicity was evaluated by the MTT assay on a HeLa cell line and in vivo imaging of subcutaneous SCC7 xenografted mice was performed using MRI/optical imaging modalities. RESULTS: Diameter and ζ-potential of the nanoparticles were measured, and TEM images demonstrated the mono-disperse nature of the particles. Quenching efficiency of the dyes on the surface was nearly 100% in the smallest nanoparticle, while almost no quenching effect was observed for the largest nanoparticle. In vitro cytotoxicity showed nearly 90% cell viability at 0.15 Fe mg/mL, a comparable concentration for clinical use. The tumor area was significantly darkened after the nanoparticle injection due to the high transverse relaxivity value of the nanoparticles. Fluorescence signal was affected by the particle size due to the distance-dependent quenching/dequenching behaviour.


Asunto(s)
Compuestos Férricos/química , Colorantes Fluorescentes/química , Imagen Multimodal/métodos , Nanopartículas/química , Dióxido de Silicio/química , Animales , Supervivencia Celular/efectos de los fármacos , Medios de Contraste , Células HeLa , Humanos , Imagen por Resonancia Magnética/métodos , Ratones , Tamaño de la Partícula , Polietilenglicoles , Sales de Tetrazolio , Tiazoles , Ensayos Antitumor por Modelo de Xenoinjerto
6.
ACS Nano ; 18(4): 3575-3582, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38235729

RESUMEN

The applications of ultrasound imaging are often limited due to low contrast, which arises from the comparable acoustic impedance of normal tissues and disease sites. To improve the low contrast, we propose a contrast agent called gas-generating laser-activatable nanorods for contrast enhancement (GLANCE), which enhances ultrasound imaging contrast in two ways. First, GLANCE absorbs near-infrared lasers and generates nitrogen gas bubbles through the photocatalytic function of gold nanorods and photolysis of azide compounds. These gas bubbles decrease the acoustic impedance and highlight the injection site from the surrounding tissues. Second, GLANCE exhibits photoacoustic properties owing to the gold nanorods that emit photoacoustic signals upon laser irradiation. Additionally, GLANCE offers several benefits for biomedical applications such as nanometer-scale size, adjustable optical absorption, and biocompatibility. These distinctive features of GLANCE would overcome the limitations of conventional ultrasound imaging and facilitate the accurate diagnosis of various diseases.


Asunto(s)
Nanotubos , Técnicas Fotoacústicas , Oro , Técnicas Fotoacústicas/métodos , Diagnóstico por Imagen , Ultrasonografía/métodos , Medios de Contraste
7.
J Funct Biomater ; 15(4)2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38667538

RESUMEN

Copper-based nanomaterials have been employed as therapeutic agents for cancer therapy and diagnosis. Nevertheless, persistent challenges, such as cellular toxicity, non-uniform sizes, and low photothermal efficiency, often constrain their applications. In this study, we present Cu2+-loaded silica nanoparticles fabricated through the chelation of Cu2+ ions by silanol groups. The integration of Cu2+ ions into uniformly sized silica nanoparticles imparts a photothermal therapy effect. Additionally, the amine functionalization of the silica coating facilitates the chemical conjugation of tumor-specific fluorescence probes. These probes are strategically designed to remain in an 'off' state through the Förster resonance energy transfer mechanism until exposed to cysteine enzymes in cancer cells, inducing the recovery of their fluorescence. Consequently, our Cu2+-loaded silica nanoparticles demonstrate an efficient photothermal therapy effect and selectively enable cancer imaging.

8.
J Nanosci Nanotechnol ; 13(5): 3223-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23858834

RESUMEN

Due to their efficient conversion of absorbed light energy to heat gold nanorods have been proved to be an amazing tool for minimally invasive photo-thermal cancer therapy. The present in vitro study demonstrates the ability of silica coated Au nanorods to function as a dual probe for cancer-cell therapy and imaging without any toxic side-effects. HeLa cells were incubated with silica coated Au nanorods and imaged inside the cell just after 1 hour of incubation by a dark field set up due to strong surface enhanced Raman scattering. To induce hyperthermia, silica coated Au nanorod incubated HeLa cells were illuminated with a diode laser (671 nm, 200 mW, 10 min). Cell destruction was observed even at a very low dose of nanorods, whereas none was observed in the absence of nanorods. Silica coated Au nanorods thus offer a promising, novel class of selective photo-thermal agents for cancer therapy and diagnosis.


Asunto(s)
Hipertermia Inducida/métodos , Nanopartículas del Metal/uso terapéutico , Neoplasias Experimentales/diagnóstico , Neoplasias Experimentales/terapia , Dióxido de Silicio/química , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/uso terapéutico , Oro/química , Oro/uso terapéutico , Células HeLa , Humanos , Luz , Ensayo de Materiales , Nanopartículas del Metal/química , Tamaño de la Partícula , Fototerapia/métodos , Resultado del Tratamiento
9.
Chem Soc Rev ; 41(7): 2656-72, 2012 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-22189429

RESUMEN

Nanomedicine is the biomedical application of nanoscale materials for diagnosis and therapy of disease. Recent advances in nanotechnology and biotechnology have contributed to the development of multifunctional nanoparticles as representative nanomedicine. They were initially developed to enable the target-specific delivery of imaging or therapeutic agents for biomedical applications. Due to their unique features including multifunctionality, large surface area, structural diversity, and long circulation time in blood compared to small molecules, nanoparticles have emerged as attractive preferences for optimized therapy through personalized medicine. Multimodal imaging and theragnosis are the cutting-edge technologies where the advantages of nanoparticles are maximized. Because each imaging modality has its pros and cons, the integration of several imaging agents with different properties into multifunctional nanoparticles allows precise and fast diagnosis of disease through synergetic multimodal imaging. Moreover, nanoparticles are not only used for molecular imaging but also applied to deliver therapeutic agents to the disease site in order to accomplish the simultaneous imaging and therapy called theragnosis. This tutorial review will highlight the recent advances in the development of multifunctional nanoparticles and their biomedical applications to multimodal imaging and theragnosis as nanomedicine.


Asunto(s)
Diagnóstico por Imagen , Sistemas de Liberación de Medicamentos , Nanomedicina/métodos , Nanopartículas/química , Humanos
10.
ACS Appl Mater Interfaces ; 15(1): 120-137, 2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-35184560

RESUMEN

In view of the fact that the blood-brain barrier (BBB) prevents the transport of imaging probes and therapeutic agents to the brain and thus hinders the diagnosis and treatment of brain-related disorders, methods of circumventing this problem (e.g., ultrasound-mediated nanoparticle delivery) have drawn much attention. Among the related techniques, focused ultrasound (FUS) is a favorite means of enhancing drug delivery via transient BBB opening. Photoacoustic brain imaging relies on the conversion of light into heat and the detection of ultrasound signals from contrast agents, offering the benefits of high resolution and large penetration depth. The extensive versatility and adjustable physicochemical properties of nanoparticles make them promising therapeutic agents and imaging probes, allowing for successful brain imaging and treatment through the combined action of ultrasound and nanoparticulate agents. FUS-induced BBB opening enables nanoparticle-based drug delivery systems to efficiently access the brain. Moreover, photoacoustic brain imaging using nanoparticle-based contrast agents effectively visualizes brain morphologies or diseases. Herein, we review the progress in the simultaneous use of nanoparticles and ultrasound in brain research, revealing the potential of ultrasound-mediated nanoparticle delivery for the effective diagnosis and treatment of brain disorders.


Asunto(s)
Medios de Contraste , Nanopartículas , Medios de Contraste/farmacología , Imagen por Resonancia Magnética/métodos , Encéfalo/diagnóstico por imagen , Barrera Hematoencefálica , Sistemas de Liberación de Medicamentos/métodos , Microburbujas , Neuroimagen
11.
Acc Chem Res ; 44(10): 1018-28, 2011 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-21851104

RESUMEN

Therapy and diagnosis are two major categories in the clinical treatment of disease. Recently, the word "theranosis" has been created, combining the words to describe the implementation of these two distinct pursuits simultaneously. For successful theranosis, the efficient delivery of imaging agents and drugs is critical to provide sufficient imaging signal or drug concentration in the targeted disease site. To achieve this purpose, biomedical researchers have developed various nanoparticles composed of organic or inorganic materials. However, the targeted delivery of these nanoparticles in animal models and patients remains a difficult hurdle for many researchers, even if they show useful properties in cell culture condition. In this Account, we review our strategies for developing theranostic nanoparticles to accomplish in vivo targeted delivery of imaging agents and drugs. By applying these rational strategies, we achieved fine multimodal imaging and successful therapy. Our first strategy involves physicochemical optimization of nanoparticles for long circulation and an enhanced permeation and retention (EPR) effect. We accomplished this result by testing various materials in mouse models and optimizing the physical properties of the materials with imaging techniques. Through these experiments, we developed a glycol chitosan nanoparticle (CNP), which is suitable for angiogenic diseases, such as cancers, even without an additional targeting moiety. The in vivo mechanism of this particle was examined through rationally designed experiments. In addition, we evaluated and compared the biodistribution and target-site accumulation of bare and drug-loaded nanoparticles. We then focus on the targeting moieties that bind to cell surface receptors. Small peptides were selected as targeting moieties because of their stability, low cost, size, and activity per unit mass. Through phage display screening, the interleukin-4 receptor binding peptide was discovered, and we combined it with our nanoparticles. This product accumulated efficiently in atherosclerotic regions or tumors during both imaging and therapy. We also developed hyaluronic acid nanoparticles that can bind efficiently to the CD44 antigen receptors abundant in many tumor cells. Their delivery mechanism is based on both physicochemical optimization for the EPR effect and receptor-mediated endocytosis by their hyaluronic acid backbone. Finally, we introduce the stimuli-responsive system related to the chemical and biological changes in the target disease site. Considering the relatively low pH in tumors and ischemic sites, we applied pH-sensitive micelle to optical imaging, magnetic resonance imaging, anticancer drug delivery, and photodynamic therapy. In addition, we successfully evaluated the in vivo imaging of enzyme activity at the target site with an enzyme-specific peptide sequence and CNPs. On the basis of these strategies, we were able to develop self-assembled nanoparticles for in vivo targeted delivery, and successful results were obtained with them in animal models for both imaging and therapy. We anticipate that these rational strategies, as well as our nanoparticles, will be applied in both the diagnosis and therapy of many human diseases. These theranostic nanoparticles are expected to greatly contribute to optimized therapy for individual patients as personalized medicine, in the near future.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/uso terapéutico , Animales , Fenómenos Químicos , Humanos , Inyecciones Intravenosas , Nanopartículas/administración & dosificación , Nanopartículas/química , Receptores de Superficie Celular/metabolismo
12.
Adv Drug Deliv Rev ; 183: 114177, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35245568

RESUMEN

As immunogenic cell death (ICD) inducers initiating antitumor immune responses, certain chemotherapeutic drugs have shown considerable potential to reverse the immunosuppressive tumor microenvironment (ITM) into immune-responsive tumors. The application of these drugs in nanomedicine provides a more enhanced therapeutic index by improving unfavorable pharmacokinetic (PK) profiles and inefficient tumor targeting. However, the clinical translation of conventional nanoparticles is restricted by fundamental problems, such as risks of immunogenicity and potential toxicity by carrier materials, premature drug leakage in off-target sites during circulation, low drug loading contents, and complex structure and synthetic processes that hinder quality control (QC) and scale-up industrial production. To address these limitations, tumor-activated carrier-free prodrug nanoparticles (PDNPs), constructed only by the self-assembly of prodrugs without any additional carrier materials, have been widely investigated with distinct advantages for safe and more effective drug delivery. In addition, combination immunotherapy based on PDNPs with other diverse modalities has efficiently reversed the ITM to immune-responsive tumors, potentiating the response to immune checkpoint blockade (ICB) therapy. In this review, the trends and advances in PDNPs are outlined, and each self-assembly mechanism is discussed. In addition, various combination immunotherapies based on PDNPs are reviewed. Finally, a physical tumor microenvironment remodeling strategy to maximize the potential of PDNPs, and key considerations for clinical translation are highlighted.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Profármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Excipientes , Humanos , Inmunoterapia , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Profármacos/química , Microambiente Tumoral
13.
Adv Drug Deliv Rev ; 186: 114344, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35580813

RESUMEN

Photodynamic nanomedicines have significantly enhanced the therapeutic efficacy of photosensitizers (PSs) by overcoming critical limitations of PSs such as poor water solubility and low tumor accumulation. Furthermore, functional photodynamic nanomedicines have enabled overcoming oxygen depletion during photodynamic therapy (PDT) and tissue light penetration limitation by supplying oxygen or upconverting light in targeted tumor tissues, resulting in providing the potential to overcome biological therapeutic barriers of PDT. Nevertheless, their localized therapeutic effects still remain a huddle for the effective treatment of metastatic- or recurrent tumors. Recently, newly designed photodynamic nanomedicines and their combination chemo- or immune checkpoint inhibitor therapy enable the systemic treatment of various metastatic tumors by eliciting antitumor immune responses via immunogenic cell death (ICD). This review introduces recent advances in photodynamic nanomedicines and their applications, focusing on overcoming current limitations. Finally, the challenges and future perspectives of the clinical translation of photodynamic nanomedicines in cancer PDT are discussed.


Asunto(s)
Neoplasias , Fotoquimioterapia , Línea Celular Tumoral , Humanos , Nanomedicina/métodos , Neoplasias/tratamiento farmacológico , Oxígeno , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/uso terapéutico
14.
Cancers (Basel) ; 14(8)2022 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-35454950

RESUMEN

One of the promising cancer treatment methods is photothermal therapy (PTT), which has achieved good therapeutic efficiency through nanoparticle-based photoabsorbers. Because of the various functions of nanoparticles, such as targeting properties, high light-to-heat conversion, and photostability, nanoparticle-mediated PTT successfully induces photothermal damage in tumor tissues with minimal side effects on surrounding healthy tissues. The therapeutic efficacy of PTT originates from cell membrane disruption, protein denaturation, and DNA damage by light-induced heat, but these biological impacts only influence localized tumor areas. This conventional nanoparticle-mediated PTT still attracts attention as a novel cancer immunotherapy, because PTT causes immune responses against cancer. PTT-induced immunogenic cell death activates immune cells for systemic anti-cancer effect. Additionally, the excellent compatibility of PTT with other treatment methods (e.g., chemotherapy and immune checkpoint blockade therapy) reinforces the therapeutic efficacy of PTT as combined immunotherapy. In this review, we investigate various PTT agents of nanoparticles and compare their applications to reveal how nanoparticle-mediated PTT undergoes a transition from thermotherapy to immunotherapy.

15.
Nanomaterials (Basel) ; 12(19)2022 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-36234538

RESUMEN

Gold nanoparticles (AuNPs) with various sizes and morphologies have been extensively investigated for effective photothermal therapy (PTT) against multiple cancer types. However, a highly dynamic and complex tumor microenvironment (TME) considerably reduces the efficacy of PTT by limiting deep tumor penetration of AuNPs. Herein, we propose a mesenchymal stem cell (MSC)-mediated deep tumor delivery of gold nanorod (AuNR) for a potent PTT. First, MSCs are treated with tetraacylated N-azidomannosamine (Ac4ManNAz) to introduce modifiable azide (N3) groups on the cell surface via metabolic glycoengineering. Then, AuNRs modified with bio-orthogonal click molecules of bicyclo[6.1.0]nonyne (AuNR@BCN) are chemically conjugated to the N3 groups on the MSC surface by copper-free click chemistry reaction, resulting in AuNR@MSCs. In cultured MSCs, the appropriate condition to incorporate the AuNR into the MSCs is optimized; in addition, the photothermal efficiency of AuNR-MSCs under light irradiation are assessed, showing efficient heat generation in vitro. In colon tumor-bearing mice, intravenously injected AuNR@MSCs efficiently accumulate within the tumor tissues by allowing deep tissue penetration owing to the tumor homing effect by natural tumor tropism of AuNR@MSCs. Upon localized light irradiation, the AuNR@MSCs significantly inhibit colon tumor growth by the enhanced photothermal effect compared to conventional AuNRs. Collectively, this study shows a promising approach of MSCs-mediated deep tumor delivery of AuNR for effective PTT.

16.
J Mater Chem B ; 9(19): 3983-4001, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-33909000

RESUMEN

Immunogenic cell death (ICD) occurring by chemical and physical stimuli has shown the potential to activate an adaptive immune response in the immune-competent living body through the release of danger-associated molecular patterns (DAMPs) into the tumor microenvironment (TME). However, limitations to the long-term immune responses and systemic toxicity of conventional ICD inducers have led to unsatisfactory therapeutic efficacy in ICD-based cancer immunotherapy. Until now, various nanoparticle-based ICD-inducers have been developed to induce an antitumor immune response without severe toxicity, and to efficiently elicit an anticancer immune response against target cancer cells. In this review, we introduce a recent advance in the designs and applications of nanoparticle-based therapeutics to elicit ICD for effective cancer immunotherapy. In particular, combination strategies of nanoparticle-based ICD inducers with typical theranostic modalities are introduced intensively. Subsequently, we discuss the expected challenges and future direction of nanoparticle-based ICD inducers to provide strategies for boosting ICD in cancer immunotherapy. These versatile designs and applications of nanoparticle-based therapeutics for ICD can provide advantages to improve the therapeutic efficacy of cancer immunotherapy.


Asunto(s)
Inmunoterapia/métodos , Nanopartículas/química , Neoplasias/terapia , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Humanos , Muerte Celular Inmunogénica/efectos de los fármacos , Nanopartículas/uso terapéutico , Nanopartículas/toxicidad , Fármacos Fotosensibilizantes/química , Linfocitos T Citotóxicos/inmunología , Nanomedicina Teranóstica
17.
Nanomaterials (Basel) ; 11(7)2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-34203541

RESUMEN

Lymph node mapping is important in cancer immunotherapy because the morphology of lymph nodes is one of the crucial evaluation criteria of immune responses. We developed new theragnostic glycol-chitosan-coated gold nanoparticles (GC-AuNPs), which highlighted lymph nodes in ultrasound-guided photoacoustic (US/PA) imaging. Moreover, the ovalbumin epitope was conjugated GC-AuNPs (OVA-GC-AuNPs) for delivering tumor antigen to lymph node resident macrophage. In vitro studies proved the vigorous endocytosis activity of J774A.1 macrophage and consequent strong photoacoustic signals from them. The macrophages also presented a tumor antigen when OVA-GC-AuNPs were used for cellular uptake. After the lingual injection of GC-AuNPs into healthy mice, cervical lymph nodes were visible in a US/PA imaging system with high contrast. Three-dimensional analysis of lymph nodes revealed that the accumulation of GC-AuNPs in the lymph node increased as the post-injection time passed. Histological analysis showed GC-AuNPs or OVA-GC-AuNPs located in subcapsular and medullar sinuses where macrophages are abundant. Our new theragnostic GC-AuNPs present a superior performance in US/PA imaging of lymph nodes without targeting moieties or complex surface modification. Simultaneously, GC-AuNPs were able to deliver tumor antigens to cause macrophages to present the OVA epitope at targeted lymph nodes, which would be valuable for cancer immunotherapy.

18.
Bioconjug Chem ; 21(11): 1939-42, 2010 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-20936793

RESUMEN

We developed a new apoptosis imaging probe with gold nanoparticles (AuNPs). A near-infrared fluorescence dye was attached to AuNP surface through the bridge of peptide substrate (DEVD). The fluorescence was quenched in physiological conditions due to the quenching effect of AuNP, and the quenched fluorescence was recovered after the DEVD had been cleaved by caspase-3, the enzyme involved in apoptotic process. The adhesion of DEVD substrates on AuNP surface was accomplished by conjugation of the 3,4-dihydroxy phenylalanine (DOPA) groups which are adhesive to inorganic surface and rich in mussels. This surface modification with DEVD substrates by DOPA groups resulted in increased stability of AuNP in cytosol condition for hours. Moreover, the cleavage of substrate and the dequenching process are very fast, and the cells did not need to be fixed for imaging. Therefore, the real-time monitoring of caspase activity could be achieved in live cells, which enabled early detection of apoptosis compared to a conventional apoptosis kit such as Annexin V-FITC. Therefore, our apoptosis imaging has great potential as a simple, inexpensive, and efficient apoptosis imaging probe for biomedical applications.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Oro/farmacología , Nanopartículas del Metal/química , Imagen Molecular/métodos , Caspasa 3/química , Supervivencia Celular/efectos de los fármacos , Colorantes Fluorescentes/química , Oro/química , Células HeLa , Humanos , Levodopa/química , Óptica y Fotónica , Tamaño de la Partícula , Péptidos/química , Propiedades de Superficie
19.
Bioconjug Chem ; 21(12): 2173-7, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21062090

RESUMEN

Herein, we developed matrix metalloprotease (MMP) sensitive gold nanorods (MMP-AuNR) for cancer imaging and therapy. It was feasible to absorb NIR laser and convert into heat as well as visualize MMP activity. We showed the possibility of gold nanorods as a hyperthermal therapeutic agent and MMP sensitive imaging agent both in vitro and in vivo condition. The results suggested potential application of MMP-AuNR for simultaneous cancer diagnosis and therapy.


Asunto(s)
Carcinoma de Células Escamosas , Diagnóstico por Imagen/métodos , Oro , Terapia por Láser/métodos , Metaloproteinasas de la Matriz/metabolismo , Fototerapia/métodos , Neoplasias Cutáneas , Animales , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/terapia , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Fluorescencia , Oro/química , Oro/uso terapéutico , Calor , Luz , Inhibidores de la Metaloproteinasa de la Matriz , Metaloproteinasas de la Matriz/química , Ratones , Ratones Desnudos , Microscopía Electrónica de Transmisión , Nanotubos/química , Trasplante de Neoplasias , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/terapia , Espectrometría de Fluorescencia , Espectroscopía Infrarroja Corta , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA