Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Small ; 17(51): e2103303, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34643054

RESUMEN

Pathogenic infections seriously threaten public health and have been considered as one of the most critical challenges in clinical therapy. Construction of a safe and efficient photothermal antibacterial platform is a promising strategy for treatment of bacterial infections. Considering that high temperature does harm to the normal tissues and cells, herein, a bacteria-triggered multifunctional hydrogel is constructed for low-temperature photothermal sterilization with high efficiency by integrating localized chemodynamic therapy (L-CDT). The hydrogel is constructed by incorporating copper sulfide nanoparticles (CuSNPs ) with photothermal profile into the network of hyaluronic acid (HA) and Fe3+ -EDTA complexes, named as CHFH (CuSNPs -HA-Fe3+ -EDTA hydrogel). Bacteria can be accumulated on the surface of CHFH, which secretes hyaluronidase to decompose the HA and release Fe3+ . The Fe3+ is reduced into Fe2+ in microenvironment of bacteria to trigger Fenton reaction. The generated hydroxyl radicals result in sterilization based on L-CDT within short range. By integrating with photothermal property of CuSNPs , low-temperature photothermal therapy (LT-PTT) for sterilization is realized, which improves the antibacterial efficiency while minimizes damage to normal tissues. The CHFH is further used to prepare Band aid which effectively promotes the Staphylococcus aureus-infected wound healing process in vivo, confirming the great potential for clinical application.


Asunto(s)
Hidrogeles , Nanopartículas , Staphylococcus aureus , Esterilización , Temperatura
2.
J Nanobiotechnology ; 19(1): 390, 2021 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-34823543

RESUMEN

BACKGROUND: Although lower temperature (< 45 °C) photothermal therapy (LPTT) have attracted enormous attention in cancer therapy, the therapeutic effect is still unsatisfying when applying LPTT alone. Therefore, combining with other therapies is urgently needed to improve the therapeutic effect of LPTT. Recently reported oxygen-irrelevant free radicals based thermodynamic therapy (TDT) exhibit promising potential for hypoxic tumor treatment. However, overexpression of glutathione (GSH) in cancer cells would potently scavenge the free radicals before their arrival to the specific site and dramatically diminish the therapeutic efficacy. METHODS AND RESULTS: In this work, a core-shell nanoplatform with an appropriate size composed of arginine-glycine-aspartate (RGD) functioned polydopamine (PDA) as a shell and a triphenylphosphonium (TPP) modified hollow mesoporous manganese dioxide (H-mMnO2) as a core was designed and fabricated for the first time. This nanostructure endows a size-controllable hollow cavity mMnO2 and thickness-tunable PDA layers, which effectively prevented the pre-matured release of encapsulated azo initiator 2,2'-azobis[2-(2-imidazolin-2-yl) propane] dihydrochloride (AIBI) and revealed pH/NIR dual-responsive release performance. With the mitochondria-targeting ability of TPP, the smart nanocomposites (AIBI@H-mMnO2-TPP@PDA-RGD, AHTPR) could efficiently induce mitochondrial associated apoptosis in cancer cells at relatively low temperatures (< 45 °C) via selectively releasing oxygen-irrelevant free radicals in mitochondria and facilitating the depletion of intracellular GSH, exhibiting the advantages of mitochondria-targeted LPTT/TDT. More importantly, remarkable inhibition of tumor growth was observed in a subcutaneous xenograft model of osteosarcoma (OS) with negligible side effects. CONCLUSIONS: The synergistic therapy efficacy was confirmed by effectively inducing cancer cell death in vitro and completely eradicating the tumors in vivo. Additionally, the excellent biosafety and biocompatibility of the nanoplatforms were confirmed both in vitro and in vivo. Taken together, the current study provides a novel paradigm toward oxygen-independent free-radical-based cancer therapy, especially for the treatment of hypoxic solid tumors.


Asunto(s)
Radicales Libres , Nanopartículas del Metal/química , Mitocondrias , Sistema de Administración de Fármacos con Nanopartículas , Terapia Fototérmica , Animales , Compuestos Azo/química , Línea Celular Tumoral , Frío , Femenino , Radicales Libres/análisis , Radicales Libres/metabolismo , Humanos , Imidazoles/química , Compuestos de Manganeso/química , Ratones , Ratones Desnudos , Mitocondrias/química , Mitocondrias/metabolismo , Óxidos/química
3.
Acta Biomater ; 159: 324-337, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36706851

RESUMEN

Collateral damage to healthy tissue, uneven heat distribution, inflammatory diseases, and tumor metastasis induction hinder the translation of high-temperature photothermal therapy (PTT) from bench to practical clinical applications. In this report, a multifunctional gold nanorod (GNR)-based nanosystem was designed by attaching siRNA against B7-H3 (B7-H3si), glucose oxidase (GOx), and hyaluronic acid (HA) for efficient low-temperature PTT. Herein, GOx can not only exhaust glucose to induce starvation therapy but also reduce the heat shock protein (HSP), realizing the ablation of tumors without damage to healthy tissues. Evidence shows that B7-H3, a type I transmembrane glycoprotein molecule, plays essential roles in growth, metastasis, and drug resistance. By initiating the downregulation of B7-H3 by siRNA, siRNA-GOx/GNR@HA NPs may promote the effectiveness of treatment. By targeting cluster of differentiation 44 (CD44) and depleting B7-H3 and HSPs sequentially, siRNA-GOx/GNR@HA NPs showed 12.9-fold higher lung distribution than siRNA-GOx/GNR NPs. Furthermore, 50% of A549-bearing mice in the siRNA-GOx/GNR NPs group survived over 50 days. Overall, this low-temperature phototherapeutic nanosystem provides an appropriate strategy for eliminating cancer with high treatment effectiveness and minimal systemic toxicity. STATEMENT OF SIGNIFICANCE: To realize efficient tumor ablation under mild low-temperature (42-45 â„ƒ) and RNA interference simultaneously, here we developed a multifunctional gold nanorod (GNR)-based nanosystem (siRNA-GOx/GNR@HA NPs). This nanoplatform can significantly inhibit tumor cell proliferation and induce cell apoptosis by downregulation of HSP90α, HSP70, B7-H3, p-AKT, and p-ERK and upregulation of cleaved caspase-9 at mild low-temperature due to its superior tumor homing ability and the combined effect of photothermal effect, glucose deprivation-initiated tumor starvation, and B7-H3 gene silence effect. It is believed that this multifunctional low-temperature photothermal nanosystem with efficient and specific anticancer properties, shows a potential application in clinical tumor treatment.


Asunto(s)
Nanotubos , Neoplasias , Animales , Ratones , Fototerapia , Temperatura , Oro/farmacología , Interferencia de ARN , Neoplasias/terapia , ARN Interferente Pequeño/genética , Glucosa , Línea Celular Tumoral
4.
Int J Nanomedicine ; 18: 7533-7541, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38106449

RESUMEN

Introduction: Photothermal therapy (PTT) has a significant potential for its application in precision tumour therapy. However, PTT-induced hyperthermia may damage healthy tissues and trigger the expression of heat shock proteins (HSPs), thereby compromising the long-term therapeutic efficacy of PTT. Methods: In this study, a biomimetic drug delivery system comprising CuP nanozymes as the inner core and platelet membrane (PM) as the outer shell was successfully developed for administering synergistic chemodynamic therapy and mild PTT. PM is encapsulated on CuP to form this biomimetic nanoparticle (PM-coated CuP nanoparticles, PC). PC possesses peroxidase (POD) activity, can facilitate the conversion of hydrogen peroxide into ·OH, thereby inhibiting the expression of HSPs. Results: Upon exposure to low-power laser irradiation (0.5 W/cm2, 1064 nm), PC can convert near-infrared II laser energy into heat energy, thereby enabling the administration of enhanced mild PTT. In vitro and in vivo experiments have demonstrated that this synergistic approach can induce over 90% tumour eradication with favourable biocompatibility. Discussion: PC exhibits high efficacy and biocompatibility, making it a promising candidate for future applications.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Polímeros , Pirroles , Fototerapia , Cobre , Terapia Fototérmica , Biomimética , Temperatura , Neoplasias/tratamiento farmacológico , Línea Celular Tumoral
5.
ACS Appl Mater Interfaces ; 15(38): 44631-44640, 2023 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-37706663

RESUMEN

In photothermal treatments (PTTs), normal tissues around cancerous tumors get injured by excessive heat, whereas damaged cancer cells are easily restored by stress-induced heat shock proteins (HSPs) at low temperatures. Therefore, to achieve a unique tumor microenvironment (TME), it is imperative to increase PTT efficiency and reduce normal tissue injury by adopting appropriate reactive oxygen species (ROS) and lipid peroxides (LPO) cross-linked with HSPs. In the present research, a potential strategy for mild photothermal treatments (mPTTs) was proposed by initiating localized catalytic chemical reactions in TME based on Pd nanozyme-modified hydrogenated TiO2 (H-TiO2@Pd). In vitro and in vivo evaluations demonstrated that H-TiO2@Pd had good peroxidase-like activities (POD), glutathione oxidase-like activities (GSHOx), and photodynamic properties and also satisfactory biocompatibility for 4T1 cells. Localized catalytic chemical reactions in H-TiO2@Pd significantly depleted GSH to downregulate the protein expression of GPX4 and promoted the accumulation of LPO and ROS, which consumed HSP70 or inhibited its function in 4T1 cells. Hence, the as-constructed low-temperature photothermal therapeutic platform based on Pd nanozyme-modified H-TiO2 can be a promising candidate to develop a safe and effective mPTT for cancer treatments.


Asunto(s)
Peróxidos Lipídicos , Terapia Fototérmica , Especies Reactivas de Oxígeno , Temperatura , Catálisis
6.
ACS Appl Mater Interfaces ; 15(47): 54207-54220, 2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-37974457

RESUMEN

Tumor drug resistance caused by the tumor microenvironment is an extremely difficult problem for researchers to solve. Nanoplatforms that integrate diagnosis and treatment have great advantages in tumor treatment, but the design and synthesis of simple and efficient nanoplatforms still face tremendous challenges. In this study, a novel Mn/Au@ir820/GA-CD133 nanoprobe was developed. The manganese dioxide/gold particles were prepared by coprecipitation/assembly, chemically coupled with CD133 antibody, and finally loaded with the photosensitive drug IR820 and the heat shock protein inhibitor Ganetespib. The nanoprobe demonstrated good tumor-targeting ability, increased the level of singlet oxygen produced from laser irradiation by effectively alleviating tumor hypoxia, and decreased the threshold of heat tolerance by downregulating the expression of HSP90 in tumor tissues. This nanoprobe successfully inhibited the growth and progression of tumor tissues in a tumor-bearing mouse model by improving the effectiveness of photodynamic and low-temperature photothermal combination therapy.


Asunto(s)
Neoplasias Pulmonares , Fotoquimioterapia , Animales , Ratones , Oro/farmacología , Temperatura , Compuestos de Manganeso/farmacología , Línea Celular Tumoral , Microambiente Tumoral
7.
ACS Appl Mater Interfaces ; 14(40): 45178-45188, 2022 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-36178205

RESUMEN

Noninvasive photothermal therapy (PTT) is an emerging strategy for eliminating multidrug-resistant (MDR) bacteria that achieve sterilization by generating temperatures above 50 °C; however, such a high temperature also causes collateral damage to healthy tissues. In this study, we developed a low-temperature PTT based on borneol-containing polymer-modified MXene nanosheets (BPM) with bacteria-targeting capabilities. BPM was fabricated through the electrostatic coassembly of negatively charged two-dimensional MXene nanosheets (2DM) and positively charged quaternized α-(+)-borneol-poly(N,N-dimethyl ethyl methacrylate) (BPQ) polymers. Integrating BPQ with 2DM improved the stability of 2DM in physiological environments and enabled the bacterial membrane to be targeted due to the presence of a borneol group and the partially positive charge of BPQ. With the aid of near-infrared irradiation, BPM was able to effectively eliminate methicillin-resistant Staphylococcus aureus (MRSA) and Escherichia coli (E. coli) through targeted photothermal hyperthermia. More importantly, BPM effectively eradicated more than 99.999% (>5 orders of magnitude) of MRSA by localized heating at a temperature that is safe for the human body (≤40 °C). Together, these findings suggest that BPM has good biocompatibility and that membrane-targeting low-temperature PTT could have great therapeutic potential against MDR infections.


Asunto(s)
Hipertermia Inducida , Staphylococcus aureus Resistente a Meticilina , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Canfanos , Escherichia coli , Humanos , Hipertermia Inducida/métodos , Metacrilatos/farmacología , Terapia Fototérmica , Polímeros/farmacología , Temperatura
8.
Biomaterials ; 286: 121593, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35635897

RESUMEN

Cellular barriers such as the cell membranes, lysosomes or nuclear pores of tumor cells hinder the drugs delivery and weaken the efficiency of traditional tumor therapies. Targeted destructing tumor cell membranes can quickly destroy cell homeostasis and kill cells without facing intracellular delivery barriers. Herein, we designed a self-delivery phototherapeutic chimeric peptide (CCP) for high efficient cell membrane-targeting combinational low-temperature photothermal therapy (LTPTT) and photodynamic therapy (PDT). The self-assembled CCP nanoparticles display remarkable tumor accumulation after systemic administration without additional carriers, avoiding the carriers related side toxicities. The CCPs are able to generate reactive oxygen species (ROS) and mild heat (<45 °C) locally at cell membrane and quickly induce immunogenic cell death to achieve efficient combinational LTPTT/PDT. The damage-associated molecular patterns released after cell membrane rupture effectively elicit antitumor immunity to eradicate residual tumor cells. With a single dosage and short-term near-infrared (NIR) light irradiation, CCPs significantly inhibit growth and metastasis of tumor, and prolong survival time of tumor-bearing mice. This work presents a unique cell membrane-targeting phototherapy strategy to kill tumor and suppress metastasis in an effective, safe and minimally invasive manner.


Asunto(s)
Nanopartículas , Neoplasias , Fotoquimioterapia , Animales , Línea Celular Tumoral , Membrana Celular , Ratones , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Péptidos/uso terapéutico , Fototerapia , Temperatura
9.
Adv Healthc Mater ; 11(3): e2101633, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34741792

RESUMEN

Urinary tract infections (UTIs) caused by uropathogenic Escherichia coli (UPEC) remain a matter of concern, as the clinical use of multiple antibiotics induces antibiotic resistance in bacteria, resulting in the failure of treatments. Despite the emergence of anti-adhesion strategies that can prevent the development of bacterial drug resistance, these strategies are mainly used for disease prevention rather than effective treatment. Photothermal therapy (PTT) has emerged as an efficient alternative for the elimination of bacteria. Nevertheless, high local temperatures related to PTT probably cause damage to surrounding healthy tissue. Herein, a biomimetic nonantibiotic nanoplatform for low-temperature photothermal treatment of UTIs is developed. The nanoplatform comprises polydopamine (PDA) photothermal core and biphenyl mannoside (Man) shell with multivalent high-affinity to UPEC. Scanning electron microscope (SEM) shows PDA-Man possessed ultra-strong targeting binding ability toward UPEC. It is the fact that this impulse UPEC to form a large bacterial cluster. Consequently, the high photothermal energy of the PDA-Man appears predominantly in the affected bacterial area, while the overall environment remains at a low temperature. The fabricated nanoplatform shows excellent photothermal bactericidal effects, approximately 100% in a UTI model. Overall, this low-temperature photothermal nanoplatform provides an appropriate strategy for the elimination of bacteria in clinical applications.


Asunto(s)
Infecciones por Escherichia coli , Infecciones Urinarias , Escherichia coli Uropatógena , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Biomimética , Infecciones por Escherichia coli/tratamiento farmacológico , Humanos , Temperatura , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/metabolismo
10.
J Colloid Interface Sci ; 614: 436-450, 2022 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-35108635

RESUMEN

Although low-temperature photothermal therapy (PTT) can sensitize tumors to immune checkpoint inhibition, its efficacy is still restricted in the deep and internal tumors without enough oxygen and lymphocytic infiltration. Non-oxygen-dependent alkyl radicals have been demonstrated to synergistically enhance PTT through up-regulating lipid peroxidation and reactive oxygen species (ROS). Herein, an innovative strategy based on alkyl radicals to augment immunogenetic cell death (ICD) caused by mild PTT was proposed to improve poor efficacy of immunotherapy, which composed of a photothermal material of Chinse ink, an azo-initiator of 2,2-azobis[2-(2-imidazoline-2-acyl)propane]dihydrochloride (AIPH) and a PD-L1 inhibitor of HY19991 (HY). Upon near-infrared-II laser irradiation, low-temperature (<45℃) stimulation induced a high expression of immune checkpoint receptor (PD-L1) in tumors and triggered a large amount alkyl radicals generated by AIPH. Significantly, the alkyl radicals augmented the ICD and increased the recruitment of tumor-infiltrating lymphocytes against tumors after transformation of the immunologically cold tumor microenvironment into hot by mild PTT. The released HY further enhanced the immunotherapy effect by blocking the binding of activated T lymphocytes and PD-L1. In vivo studies exhibited that the all-in-one hydrogel with synergistic mechanisms had an extraordinary ability to reverse the immunosuppressive microenvironment, stimulate innate and adaptive immune responses to eliminate tumors and prevent metastasis.


Asunto(s)
Inmunoterapia , Neoplasias , Línea Celular Tumoral , Humanos , Fototerapia , Temperatura , Microambiente Tumoral
11.
ACS Appl Mater Interfaces ; 13(26): 30274-30283, 2021 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-34170100

RESUMEN

In this work, an iron self-boosting polymer nanoenzyme was prepared by using pyrrole-3-carboxylic acid as a monomer and iron as an oxidizing agent via a simple and one-step method [hereafter referred to as FePPy nanoparticles (NPs)]. In fact, researchers previously paid negligible attention on the iron element during the polymerization reaction of polypyrrole, thus the intrinsically catalytic functions and enzymatic activities of the high iron content (wt %: 21.11%) are ignored and not fully explored. As expected, results demonstrate that the as-synthesized FePPy NPs can decompose H2O2 to generate hydroxyl radicals (•OH) which exhibit enzyme characteristics, further inducing a nonapoptotic ferroptosis pathway. Moreover, the nanoenzyme shows impressive photothermal properties which can accelerate the Fenton reactions to enhance ferroptosis. The combined photothermal and ferroptosis therapy of FePPy NPs was found to have high efficacy. With the properties of easy synthesis, high efficacy, and good biocompatibility, the FePPy NPs are considered as potential agents for cancer treatments.


Asunto(s)
Antineoplásicos/uso terapéutico , Ferroptosis/efectos de los fármacos , Nanoestructuras/uso terapéutico , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/uso terapéutico , Animales , Antineoplásicos/química , Antineoplásicos/efectos de la radiación , Ácidos Carboxílicos/química , Ácidos Carboxílicos/efectos de la radiación , Ácidos Carboxílicos/uso terapéutico , Catálisis , Femenino , Células HeLa , Humanos , Peróxido de Hidrógeno/química , Radical Hidroxilo/metabolismo , Hierro/química , Hierro/efectos de la radiación , Luz , Ratones Endogámicos BALB C , Ratones Desnudos , Nanoestructuras/química , Nanoestructuras/efectos de la radiación , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/efectos de la radiación , Terapia Fototérmica , Polímeros/química , Polímeros/efectos de la radiación , Polímeros/uso terapéutico , Pirroles/química , Pirroles/efectos de la radiación , Pirroles/uso terapéutico , Temperatura
12.
ACS Appl Mater Interfaces ; 12(4): 4265-4275, 2020 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-31903741

RESUMEN

Conventional therapeutic approaches to treat malignant tumors such as surgery, chemotherapy, or radiotherapy often lead to poor therapeutic results, great pain, economic burden, and risk of recurrence and may even increase the difficulty in treating the patient. Long-term drug administration and systemic drug delivery for cancer chemotherapy would be accompanied by drug resistance or unpredictable side effects. Thus, the use of photothermal therapy, a relatively rapid tumor elimination technique that regulates autophagy and exerts an antitumor effect, represents a novel solution to these problems. Heat shock protein 90 (HSP90), a protein that reduces photothermal or hypothermic efficacy, is closely related to AKT (protein kinase B) and autophagy. Therefore, it was hypothesized that autophagy could be controlled to eliminate tumors by combining exogenous light with a selective HSP90 inhibitor, for example, SNX-2112. In this study, an efficient tumor-killing strategy using graphene oxide loaded with SNX-2112 and folic acid for ultrafast low-temperature photothermal therapy (LTPTT) is reported. A unique mechanism that achieves remarkable therapeutic performance was discovered, where overactivated autophagy induced by ultrafast LTPTT led to direct apoptosis of tumors and enabled functional recovery of T cells to promote natural immunity for actively participating in the attack against tumors. This LTPTT approach resulted in residual tumor cells being rendered in an "injured" state, opening up the possibility of concurrent antitumor and antirecurrence treatment.


Asunto(s)
Antineoplásicos/administración & dosificación , Autofagia/efectos de los fármacos , Autofagia/efectos de la radiación , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Neoplasias/tratamiento farmacológico , Fotoquimioterapia/métodos , Animales , Línea Celular Tumoral , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Masculino , Ratones Endogámicos BALB C , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/fisiopatología , Fotoquimioterapia/instrumentación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Temperatura
13.
Adv Healthc Mater ; 9(2): e1901307, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31814332

RESUMEN

A nanoplatform that integrates diagnostic and therapeutic functions with intrinsic tumor microenvironment-responsive biodegradability is highly desired. Herein, a biodegradable nanotheranostic agent based on hollow mesoporous organosilica nanoparticles (HMONs), followed by encapsulating of heat shock protein 90 (Hsp 90) inhibitor is described. Then, the pore-engineering including gating with bovine serum albumin-iridium oxide nanoparticles (BSA-IrO2 ) and conjugation of polyethylene glycol (PEG) is conducted to yield 17AAG@HMONs-BSA-IrO2 -PEG (AHBIP) nanotheranostics for multimode computed tomography (CT)/photoacoustic (PA) imaging-guided photodynamic therapy (PDT) and low-temperature photothermal therapy (PTT). Such nanoplatforms show extraordinary photothermal conversion efficiency, high cargo loading (35.4% for 17AAG), and stimuli-responsive release of 17AAG for inhibition of Hsp90, which induces cell apoptosis at low-temperatures (≈41 °C). Also, the IrO2 simultaneously endows the nanotheranostics with catalytic activity in triggering the decomposition of H2 O2 into O2 and thus reducing the tumor hypoxia, as well as protecting normal tissues against H2 O2 -induced inflammation. AHBIP shows good photocatalysis activity for PDT as a result of the generation of superoxide anion by laser irradiation. The resulting AHBIP-mediated synergistic PTT/PDT offers an outstanding therapeutic outcome both in vitro and in vivo. Overall, the incorporation of the BSA-IrO2 and biodegradable HMONs into one nanoplatform has great potential for clinical applications.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Benzoquinonas/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Lactamas Macrocíclicas/administración & dosificación , Nanopartículas/química , Nanomedicina Teranóstica/métodos , Animales , Antiinflamatorios no Esteroideos/química , Benzoquinonas/farmacocinética , Materiales Biocompatibles/química , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular , Femenino , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Peróxido de Hidrógeno/química , Iridio/química , Lactamas Macrocíclicas/farmacocinética , Ratones Endogámicos C57BL , Ratones Desnudos , Oxígeno/farmacocinética , Técnicas Fotoacústicas , Fotoquimioterapia/métodos , Polietilenglicoles/química , Albúmina Sérica Bovina/química , Superóxidos/metabolismo , Nanomedicina Teranóstica/instrumentación , Tomografía Computarizada por Rayos X , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Adv Healthc Mater ; 8(16): e1900250, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31290616

RESUMEN

Hyperthemia (>50 °C) induced heating damage of nearby normal organs and inflammatory diseases are the main challenges for photothermal therapy (PTT) of cancers. To overcome this limitation, a redox-responsive biomodal tumor-targeted nanoplatform is synthesized, which can achieve multispectral optoacoustic tomography/X-ray computed tomography imaging-guided low-temperature photothermal-radio combined therapy (PTT RT). In this study, Bi2 Se3 hollow nanocubes (HNCs) are first fabricated based on a mild cation exchange way and Kirkendall effect and then modified with hyaluronic acid (HA) through redox-cleavable linkage (-s-s-), thus enabling the HNC to target cancer cells overexpressing CD-44 and control the cargo release profile. Finally, gambogic acid (GA), a type of heat-shock protein (HSP) inhibitor, which is vital to cells resisting heating-caused damage is loaded, into Bi2 Se3 HNC. Such HNC-s-s-HA/GA under a mild NIR laser irradiation can induce efficient cancer cell apoptosis, achieving PTT under relatively low temperature (≈43 °C) with remarkable cancer cell damage efficiency. Furthermore, enhanced radiotherapy (RT) can also be experienced without depth limitation based on RT sensitizer Bi2 Se3 HNC. This research designs a facile way to synthesize Bi2 Se3 HNC-s-s-HA/GA possessing theranostic functionality and cancer cells-specific GSH, but also shows a low-temperature PTT RT method to cure tumors in a minimally invasive and highly efficient way.


Asunto(s)
Nanoestructuras/química , Fototerapia/métodos , Bismuto/química , Receptores de Hialuranos/química , Oxidación-Reducción , Selenio/química , Temperatura , Xantonas/química
15.
ACS Appl Mater Interfaces ; 10(49): 42115-42126, 2018 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-30462492

RESUMEN

Noninvasive physical treatment with relatively low intensity stimulation and the development of highly efficient anticancer medical strategy are still desirable for cancer therapy. Herein a versatile, biodegradable, hollow mesoporous organosilica nanocapsule (HMONs) nanoplatform that is capped by the gemcitabine (Gem) molecule through a pH-sensitive acetal covalent bond is designed. The fabricated nanocapsule exhibits desirable small molecule release at the tumor tissues/cell sites and shows a reduced risk for drug accumulation. After loading indocyanine green (ICG), the heat-shock protein 90 (Hsp 90) inhibitor, and 17AAG and modification with polyethylene glycol (NH2-PEG), the resulting ICG-17AAG@HMONs-Gem-PEG exhibited a precisely controlled release of ICG and 17AAG and low-temperature photothermal therapy (PTT) (∼41 °C) with excellent tumor destruction efficacy. In addition, ICG loading conferred the nanoplatform with near-infrared fluorescence imaging (FL) and photoaccoustic (PA) imaging capability. In short, this work not only presents a smart drug self-controlled nanoplatform with pH-responsive payload release and theranostic performance but also provides an outstanding low-temperature PTT strategy, which is highly valid in the inhibition of cancer cells with minimal damage to the organism. Therefore, this research provides a paradigm that has a chemodrug-gated HMONs-based theranostic nanoplatform with intrinsic biodegradability, multimodal imaging capacity, high low-temperature PTT/chemotherapy efficacy, and reduced systemic toxicity.


Asunto(s)
Doxorrubicina , Hipertermia Inducida , Verde de Indocianina , Nanocápsulas , Compuestos de Organosilicio , Fototerapia , Animales , Línea Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Humanos , Verde de Indocianina/química , Verde de Indocianina/farmacocinética , Verde de Indocianina/farmacología , Ratones , Ratones Desnudos , Nanocápsulas/química , Nanocápsulas/uso terapéutico , Compuestos de Organosilicio/química , Compuestos de Organosilicio/farmacocinética , Compuestos de Organosilicio/farmacología , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Polietilenglicoles/farmacología
16.
ACS Nano ; 12(3): 2858-2872, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29510031

RESUMEN

Tumor hypoxia is one of the major challenges for the treatment of tumors, as it may negatively affect the efficacy of various anticancer modalities. In this study, a tumor-targeted redox-responsive composite biocatalyst is designed and fabricated, which may combine tumor starvation therapy and low-temperature photothermal therapy for the treatment of oxygen-deprived tumors. The nanosystem was prepared by loading porous hollow Prussian Blue nanoparticles (PHPBNs) with glucose oxidase (GOx) and then coating their surface with hyaluronic acid (HA) via redox-cleavable linkage, therefore allowing the nanocarrier to bind specifically with CD44-overexpressing tumor cells while also exerting control over the cargo release profile. The nanocarriers are designed to enhance the efficacy of the hypoxia-suppressed GOx-mediated starvation therapy by catalyzing the decomposition of intratumoral hydroperoxide into oxygen with PHPBNs, and the enhanced glucose depletion by the two complementary biocatalysts may consequently suppress the expression of heat shock proteins (HSPs) after photothermal treatment to reduce their resistance to the PHPBN-mediated low-temperature photothermal therapies.


Asunto(s)
Ferrocianuros/uso terapéutico , Glucosa Oxidasa/uso terapéutico , Hipertermia Inducida/métodos , Nanopartículas/uso terapéutico , Neoplasias/terapia , Animales , Sistemas de Liberación de Medicamentos , Glucosa/metabolismo , Células Hep G2 , Humanos , Peróxido de Hidrógeno/metabolismo , Ratones , Ratones Desnudos , Nanopartículas/ultraestructura , Neoplasias/metabolismo , Neoplasias/patología , Oxígeno/metabolismo , Fototerapia/métodos , Temperatura
17.
Adv Mater ; 29(40)2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28833643

RESUMEN

Near-infrared (NIR)-light-triggered photothermal therapy (PTT) usually requires hyperthermia to >50 °C for effective tumor ablation, which can potentially induce inflammatory disease and heating damage of normal organs nearby, while tumor lesions without sufficient heating (e.g., the internal part) may survive after treatment. Achieving effective tumor killing under relatively low temperatures is thus critical toward successful clinical use of PTT. Herein, we design a simple strategy to fabricate poly(ethylene glycol) (PEG)-modified one-dimensional nanoscale coordination polymers (1D-NCPs) with intrinsic biodegradability, large surface area, pH-responsive behaviors, and versatile theranostic functions. With NCPs consisting of Mn2+/indocyanine green (ICG) as the example, Mn-ICG@pHis-PEG display efficient pH-responsive tumor retention after systemic administration and then load Gambogic acid (GA), a natural inhibitor of heat-shock protein 90 (Hsp90) that plays an essential role for cells to resist heating-induced damage. Such Mn-ICG@pHis-PEG/GA under a mild NIR-triggered heating is able to induce effective apoptosis of tumor cells, realizing low-temperature PTT (~43 °C) with excellent tumor destruction efficacy. This work not only develops a facile approach to fabricate PEGylated 1D-NCPs with tumor-specific pH responsiveness and theranostic functionalities, but also presents a unique low-temperature PTT strategy to kill cancer in a highly effective and minimally invasive manner.


Asunto(s)
Nanofibras , Animales , Ratones Endogámicos BALB C , Polímeros , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA