Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 337
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Anal Chem ; 95(7): 3761-3768, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36757879

RESUMEN

Lanthanide nanoparticles exhibit unique photophysical properties and thus emerge as promising second near-infrared (NIR-II) optical agents. However, the limited luminescence brightness hampers their construction of activatable NIR-II probes. Herein, we report the synthesis of dye-sensitized lanthanide nanoprobes (NaGdF4:Nd/ICG; indocyanine green (ICG)) and their further development for in vivo activatable imaging of hypochlorite (ClO-). Dye sensitization using ICG not only shifts the optimal doping concentration of Nd3+ from 5 to 20 mol % but also leads to a 5-fold NIR-II enhancement relative to the ICG-free counterpart. Mechanistic studies reveal that such a luminescence enhancement of NaGdF4:Nd at high Nd3+ concentration is ascribed to an alleviated cross-relaxation effect due to the broad absorption of ICG and faster energy transfer process. Taking advantage of dye oxidation, the nanoprobes enable activatable NIR-II imaging of hypochlorous acid (ClO-) in a drug-induced lymphatic inflammation mouse model. This work thus provides a simple, yet effective luminescence enhancement strategy for constructing lanthanide nanoprobes at higher activator doping concentration toward activatable NIR-II molecular imaging.


Asunto(s)
Elementos de la Serie de los Lantanoides , Nanopartículas del Metal , Animales , Ratones , Luminiscencia , Diagnóstico por Imagen , Verde de Indocianina/farmacología
2.
Small ; 19(49): e2303541, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37608451

RESUMEN

Extracellular vesicles (EVs) have emerged as potential tools for tumor-target therapy accompanied with activating anticancer immune responses by serving as an integrated platform, but usually suffered from the limited cross presentation of tumor-associated antigen by dendritic cells (DCs). Here, a straightforward engineering strategy to construct heat shock proteins 70 (HSP70) highly expressed EVs incapsulated with Te nanoparticles (Te@EVsHSP70 ) for tumor photothermal therapy triggering improved immunotherapy is proposed. Tumor cells are firstly used as bioreactors for intracellular synthesis of Te nanoparticles, and NIR irradiation is subsequently introduced to upregulate the expression of HSP70 to give engineered Te@EVsHSP70 through exocytosis. Te@EVsHSP70 exhibits excellent photothermal performance and enhanced tumor antigen capture capability, which induces significant immunogenic death of tumor cells and improves DCs maturation both in vitro and in vivo. Thus, the engineered EVs demonstrate superior antitumor efficacy through photothermal effect and following provoked antitumor immune responses. This work provides a facile method to fabricate multifunctional EVs-based drug delivery system for improving photothermal-triggered tumor immunotherapy.


Asunto(s)
Vesículas Extracelulares , Nanopartículas , Neoplasias , Humanos , Presentación de Antígeno/fisiología , Inmunoterapia , Antígenos de Neoplasias , Línea Celular Tumoral
3.
Acc Chem Res ; 55(4): 526-536, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35077133

RESUMEN

Cancer stem cells (CSCs), also known as tumor initiating cells or tumor repopulating cells, which comprise only a small fraction of tumor, have received tremendous attention during the past two decades, as they are considered as the ringleader for initiation and progression of tumors, therapy resistance, metastasis, and recurrence in the clinic. Hence, eradicating CSCs is critical for successful cancer treatment. To that end, various CSC-targeting therapeutic agents have been pursued. However, these CSC-specific drugs are ineffective toward bulk cancer cells. Furthermore, these anti-CSC drugs not only eradicate CSCs but also affect conventional stem cells in normal organs or tissues. By virtue of the enhanced permeability and retention (EPR) effect, nanomaterial drug delivery systems (NDDSs) passively accumulate in tumor tissues, thereby alleviating severe side effects toward normal viscera. NDDSs can be further functionalized with CSC-specific binding molecules to promote targeted drug delivery toward CSCs. Moreover, NDDSs have unique advantages in encapsulating CSC-specific drugs and cytotoxic agents, realizing synchronized killing of CSCs and bulk cancer cells both temporally and spatially. For these reasons, leveraging nanotherapeutic strategies to target CSCs has gained tremendous attention recently.Some ten years ago, we summarized five basic features of efficient nanotherapeutics (the five features principle), which consist of long circulation, tumor accumulation, deep penetration, cellular internalization, and drug release. Based on this design rationale, we constructed several NDDSs, including nanogels with adaptive hydrophobicity, CSC-derived microparticles with tailored softness, and tumor exosome sheathed porous silicon biomimetic nanoparticles, for targeted drug delivery to tumor. To our astonishment, these NDDSs that possess the five basic features achieve decent drug delivery efficiency toward not only bulk tumor cells but more importantly CSCs. Consequently, such nanotherapeutics as-designed based on the five features principle are potent in eradicating CSCs, even with only cytotoxic drugs, for instance, doxorubicin. Furthermore, commercialized nanomedicines, such as Doxil and Abraxane, can be endowed with these five basic features by hyperbaric oxygen therapy and therefore achieve outstanding drug delivery efficiency, potent CSC elimination, and efficient cancer therapy. These studies suggest that intractable CSCs can be tackled with a material-based approach, highlight the critical role of the five features principle in designing effective nanotherapeutics, and pinpoint the significance of drug delivery efficiency in eliminating CSCs and bulk cancer cells.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos , Humanos , Nanopartículas/química , Neoplasias/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología
4.
Pharmacol Res ; 190: 106740, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36958408

RESUMEN

Cancer stem cells (CSCs) have been blamed as the main culprit of tumor initiation, progression, metastasis, chemoresistance, and recurrence. However, few anti-CSCs agents have achieved clinical success so far. Here we report a novel derivative of lonidamine (LND), namely HYL001, which selectively and potently inhibits CSCs by targeting mitochondria, with 380-fold and 340-fold lower IC50 values against breast cancer stem cells (BCSCs) and hepatocellular carcinoma stem cells (HCSCs), respectively, compared to LND. Mechanistically, we reveal that HYL001 downregulates glutaminase (GLS) expression to block glutamine metabolism, blunt tricarboxylic acid cycle, and amplify mitochondrial oxidative stress, leading to apoptotic cell death. Therefore, HYL001 displays significant antitumor activity in vivo, both as a single agent and combined with paclitaxel. Furthermore, HYL001 represses CSCs of fresh tumor tissues derived from liver cancer patients. This study provides critical implications for CSCs biology and development of potent anti-CSCs drugs.


Asunto(s)
Antineoplásicos , Neoplasias Hepáticas , Humanos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/metabolismo , Glutamina/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Mitocondrias/metabolismo , Células Madre Neoplásicas , Línea Celular Tumoral
5.
J Nanobiotechnology ; 21(1): 413, 2023 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-37946199

RESUMEN

Various X-ray imaging technologies like computed tomography (CT) and digital subtraction angiography (DSA) are widely used in transcatheter arterial embolization (TAE) therapy for treating hepatocellular cancer (HCC) patients. Although they display high-contrast imaging, they have a few disadvantages, such as complex operation and exposure to ionizing radiation. Thus, ultrasound (US) imaging plays an important role in medical diagnosis because of its advantages, like simple and fast operation, no ionizing radiation exposure, and accurate real-time imaging. Subsequently, Poly N-isopropylacrylamide-co-2,2,3,4,4,4-Hexafluorobutyl methacrylate (PNF) nanogels were synthesized for stabilizing TGFPE, the Pickering emulsions of 2H, 3H-decafluoropentane (HDFP). These emulsions displayed dual abilities of thermosensitive sol-gel transition and long-term US imaging in vitro. Thus, it was concluded that these emulsions could achieve vascular embolization and long-term US imaging in vivo as per the TAE animal model results. The emulsion droplets' flow and accumulation were visualized under the US imaging guidance. In summary, the Pickering emulsions have the potential to be used as US-guided embolization material for mediating TAE surgeries.


Asunto(s)
Carcinoma Hepatocelular , Embolización Terapéutica , Neoplasias Hepáticas , Animales , Humanos , Neoplasias Hepáticas/terapia , Carcinoma Hepatocelular/terapia , Nanogeles , Temperatura , Emulsiones , Embolización Terapéutica/métodos
6.
J Nanobiotechnology ; 21(1): 183, 2023 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-37291573

RESUMEN

Typical chemo-immunotherapy against malignant carcinoma, is characterized by the combined application of chemotherapeutic agents and monoclonal antibodies for immune checkpoint blockade (ICB). Temporary ICB with antibodies would not depress tumor intrinsic PD-L1 expression and potential PD-L1 adaptive upregulation during chemotherapy, thus exerting limited immunotherapy efficacy. Herein, we developed novel polymer-lipid hybrid nanoparticles (2-BP/CPT-PLNs) for inducing PD-L1 degradation by inhibiting palmitoylation with bioactive palmitic acid analog 2-bromopalmitate (2-BP) to replace PD-L1 antibody (αPD-L1) for ICB therapy, thus achieving highly efficient antitumor immune via immunogenic cell death (ICD) induced by potentiated chemotherapy. GSH-responsive and biodegradable polymer-prodrug CPT-ss-PAEEP10 assisted as a cationic helper polymer could help to stabilize 2-BP/CPT-PLNs co-assembled with 2-BP, and facilitate the tumor site-specific delivery and intracellular release of water-insoluble camptothecin (CPT) in vivo. 2-BP/CPT-PLNs would reinforce cytotoxic CD8+ T cell-mediated antitumor immune response via promoting intratumoral lymphocytes cells infiltration and activation. 2-BP/CPT-PLNs significantly prevented melanoma progression and prolonged life survival of mice beyond the conventional combination of irinotecan hydrochloride (CPT-11) and αPD-L1. Our work first provided valuable instructions for developing bioactive lipid analogs-derived nanoparticles via lipid metabolism intervention for oncotherapy.


Asunto(s)
Carcinoma , Melanoma , Nanopartículas , Ratones , Animales , Antígeno B7-H1 , Anticuerpos Monoclonales , Inmunoterapia , Nanopartículas/uso terapéutico , Polímeros , Lípidos , Ácidos Grasos , Línea Celular Tumoral
7.
Small ; 18(8): e2105716, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34889048

RESUMEN

Probiotic Escherichia coli Nissle 1917 (EcN) are employed as a bioreactor for intracellularly synthesizing tellurium nanorods (TeNRs) providing a biohybrid therapeutic platform (Te@EcN) for the elimination of advanced malignant tumor by photothermal immunotherapy. Te@EcN is found to possess superior photothermal property upon near-infrared irradiation, and can efficiently accumulate and retain in tumors, although EcN loses proliferation ability after the synthesis of TeNRs, thus inducing considerable immunogenic tumor cell death. Under co-stimulation by EcN acting as immunoadjuvants, maturation of dendritic cells and priming of cytotoxic T cells are largely promoted. In addition, Te@EcN can reprogram tumor-associated macrophages to ameliorate the immunosuppressive tumor microenvironment. Thus, tumor metastasis and recurrence can be efficiently suppressed. Most importantly, owing to the non-pathogenicity of probiotic EcN and their non-proliferative characteristics after TeNRs synthesis, Te@EcN is found to be rapidly metabolized and cleared from the normal tissues, showing very slight acute side effects in healthy mice even at a relatively high administration dose. Therefore, the proposed combined therapeutic strategy based on bacteria-synthesized TeNRs may find great potential in improving bacteria-mediated tumor therapy with increased antitumor efficacy and reduced toxicity.


Asunto(s)
Nanotubos , Neoplasias , Animales , Inmunoterapia , Ratones , Neoplasias/terapia , Telurio , Microambiente Tumoral
8.
Chem Soc Rev ; 50(10): 6013-6041, 2021 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-34027953

RESUMEN

Glutathione (GSH), the main redox buffer, has long been recognized as a pivotal modulator of tumor initiation, progression and metastasis. It is also implicated in the resistance of platinum-based chemotherapy and radiation therapy. Therefore, depleting intracellular GSH was considered a potent solution to combating cancer. However, reducing GSH within cancer cells alone always failed to yield desirable therapeutic effects. In this regard, the convergence of GSH-scavenging agents with therapeutic drugs has thus been pursued in clinical practice. Unfortunately, the therapeutic outcomes are still unsatisfactory due to untargeted drug delivery. Advanced nanomedicine of synergistic GSH depletion and cancer treatment has attracted tremendous interest because they promise to deliver superior therapeutic benefits while alleviating life-threatening side effects. In the past five years, the authors and others have demonstrated that numerous nanomedicines, by simultaneously delivering GSH-depleting agents and therapeutic components, boost not only traditional chemotherapy and radiotherapy but also multifarious emerging treatment modalities, including photodynamic therapy, sonodynamic therapy, chemodynamic therapy, ferroptosis, and immunotherapy, to name a few, and achieved decent treatment outcomes in a large number of rodent tumor models. In this review, we summarize the most recent progress in engineering nanomedicine for GSH depletion-enhanced cancer therapies. Biosynthesis of GSH and various types of GSH-consuming strategies will be briefly introduced. The challenges and perspectives of leveraging nanomedicine for GSH consumption-augmented cancer therapies will be discussed at the end.


Asunto(s)
Glutatión/biosíntesis , Nanomedicina , Neoplasias/terapia , Resistencia a Antineoplásicos , Ferroptosis/efectos de los fármacos , Glutatión/química , Glutatión/deficiencia , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Oxidantes/química , Fotoquimioterapia , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Especies Reactivas de Oxígeno/química , Especies Reactivas de Oxígeno/metabolismo
9.
Adv Exp Med Biol ; 1295: 77-95, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33543456

RESUMEN

Nanomedicine has been a hot topic in the field of tumor therapy in the past few decades. Because of the enhanced permeability and retention effect (EPR effect), nanomedicine can passively yet selectively accumulate at tumor tissues. As a result, it can improve drug concentration in tumor tissues and reduce drug distribution in normal tissues, thereby contributing to enhanced antitumor effect and reduced adverse effects. However, the therapeutic efficacy of anticancer nanomedicine is not satisfactory in clinical settings. Therefore, how to improve the clinical therapeutic effect of nanomedicine has become an urgent problem. The grand challenges of nanomedicine lie in how to overcome various pathophysiological barriers and simultaneously kill cancer cells effectively in hypoxic tumor microenvironment (TME). To this end, the development of novel stimuli-responsive nanomedicine has become a new research hotspot. While a great deal of progress has been made in this direction and preclinical results report many different kinds of promising multifunctional smart nanomedicine, the design of these intelligent nanomedicines is often too complicated, the requirements for the preparation processes are strict, the cost is high, and the clinical translation is difficult. Thus, it is more practical to find solutions to promote the therapeutic efficacy of commercialized nanomedicines, for example, Doxil®, Oncaspar®, DaunoXome®, Abraxane®, to name a few. Increasing attention has been paid to the combination of modern advanced medical technology and nanomedicine for the treatment of various malignancies. Recently, we found that hyperbaric oxygen (HBO) therapy could enhance Doxil® antitumor efficacy. Inspired by this study, we further carried out researches on the combination of HBO therapy with other nanomedicines for various cancer therapies, and revealed that HBO therapy could significantly boost antitumor efficacy of nanomedicine-mediated photodynamic therapy and photothermal therapy in different kinds of tumors, including hepatocellular carcinoma, breast cancer, and gliomas. Our results implicate that HBO therapy might be a universal strategy to boost therapeutic efficacy of nanomedicine against hypoxic solid malignancies.


Asunto(s)
Antineoplásicos , Oxigenoterapia Hiperbárica , Neoplasias , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos , Humanos , Nanomedicina , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
10.
Chem Soc Rev ; 49(8): 2273-2290, 2020 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-32215407

RESUMEN

Modulating nanomedicine mechanical properties for enhanced drug delivery to tumors has attracted increasing attention in the past few decades. In this tutorial review, we analyze the impact of nanomedicine mechanical properties on in vivo transport processes and highlight the most recent advances in drug delivery efficiency and antitumor efficacy. Typical nanoparticles that have been explored for this purpose since 2000 are summarized while the methods to tune and the techniques to characterize nanomedicine mechanical properties are introduced. In the end, challenges and perspectives on tailoring nanomedicine mechanical properties for tumor targeting delivery are discussed.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Fenómenos Mecánicos , Nanomedicina/métodos , Neoplasias/tratamiento farmacológico , Animales , Humanos
11.
Nano Lett ; 20(7): 5236-5242, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32501705

RESUMEN

Epitaxial growth of an inert shell around the optical active lanthanide upconversion nanoparticles (UCNPs) is a general strategy to enhance their brightness. Yet, its potential as a tool in multiplexing emission tailoring has rarely been reported. Here, by developing the atomic vacancies into color selectivity actuators, we present an efficient strategy to achieve inert-shell-modulated multiplexing upconversion in 1540 nm activated UCNPs. Artificially generated fluoride atomic vacancies, owing to the decreased NaOH/NH4F dosage during shell growth, reduce the coordination number of Y-F and lattice densities in the inert shell, leading to the core-engineered shell nanoparticles with distinctive emission profiles. The multicolor tailoring is independent of shell thickness and can be readily applied to Lu3+/Gd3+-based shells. The upconversion emission can be exploited to visualize in security decoding and in vivo multiplexing bioimaging. This method of regulating atomic vacancies based on the inert-shell engineering opens new insights of upconversion modulation in core-shell lanthanide nanostructures.

12.
Nano Lett ; 20(1): 261-271, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31786926

RESUMEN

Although a variety of advanced sterilization materials and treatments have emerged, the complete elimination of bacterial infection, especially drug-resistant bacterial infection, remains an immense challenge. Here, we demonstrate the use of neutrophils loaded with photocatalytic nanoparticles to reduce bacterial infection. This method activates the immune system to achieve an anti-infection response. We prepared the photocatalytic nanoparticle-laden neutrophils in vivo through neutrophil phagocytosis. The resulting loaded cells retained the cell membrane functionality of the source cell, as well as the complete immune cell function of neutrophils, particularly the ability to recruit macrophages to the target area. Photocatalytic nanoparticle-laden neutrophils can target infection sites and release reactive oxygen species to induce the secretion of chemokines, leading to the targeted recruitment of macrophages and enhancing a powerful immune cascade. In a severe mouse infection model induced by pathogenic bacteria, small doses of photocatalytic nanoparticle-laden neutrophils showed a remarkable therapeutic effect by enhancing macrophage recruitment and the immune cascade.


Asunto(s)
Óxido Ferrosoférrico , Nanopartículas/química , Activación Neutrófila/efectos de los fármacos , Neutrófilos/inmunología , Fagocitosis/efectos de los fármacos , Titanio , Animales , Femenino , Óxido Ferrosoférrico/química , Óxido Ferrosoférrico/farmacología , Staphylococcus aureus Resistente a Meticilina/inmunología , Ratones , Células RAW 264.7 , Titanio/química , Titanio/farmacología
13.
Angew Chem Int Ed Engl ; 60(25): 14051-14059, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-33797161

RESUMEN

Despite the promise of sonodynamic processes in cancer therapy, existing sonosensitizers often fail to regulate the generation of reactive oxygen species (ROS) against tumors, potentially leading to off-target toxicity to normal tissues. We report a transformable core-shell nanosonosensitizer (TiO2 @CaP) that reinvigorates ROS generation and dissolves its CaP shell to release Ca2+ in an acidic tumor microenvironment (TME) under ultrasound activation. Thus, TiO2 @CaP acts as a smart nanosonosensitizer that specifically induces mitochondrial dysfunction via overloading intracellular Ca2+ ions to synergize with the sonodynamic process in the TME. TiO2 @CaP substantially enhances immunogenic cell death, resulting in enhanced T-cell recruitment and infiltration into the immunogenic cold tumor (4T1). In conjunction with checkpoint blockade therapy (anti-PD 1), TiO2 @CaP-mediated sonodynamic therapy elicits systemic antitumor immunity, leading to regression of non-treated distant tumors and inhibition of lung metastasis. This work paves the way to development of "smart" TME-activatable sonosensitizers with temporospatial control over antitumor responses.


Asunto(s)
Neoplasias de la Mama/metabolismo , Calcio/metabolismo , Nanopartículas/metabolismo , Titanio/metabolismo , Terapia por Ultrasonido , Animales , Neoplasias de la Mama/terapia , Calcio/química , Línea Celular Tumoral , Femenino , Ratones , Nanopartículas/química , Tamaño de la Partícula , Titanio/química , Microambiente Tumoral
14.
Small ; 16(8): e1906797, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32003923

RESUMEN

The optogenetic neuron ablation approach enables noninvasive remote decoding of specific neuron function within a complex living organism in high spatiotemporal resolution. However, it suffers from shallow tissue penetration of visible light with low ablation efficiency. This study reports a upconversion nanoparticle (UCNP)-based multiplex proteins activation tool to ablate deep-tissue neurons for locomotion modulation. By optimizing the dopant contents and nanoarchitecure, over 300-fold enhancement of blue (450-470 nm) and red (590-610 nm) emissions from UCNPs is achieved upon 808 nm irradiation. Such emissions simultaneously activate mini singlet oxygen generator and Chrimson, leading to boosted near infrared (NIR) light-induced neuronal ablation efficiency due to the synergism between singlet oxygen generation and intracellular Ca2+ elevation. The loss of neurons severely inhibits reverse locomotion, revealing the instructive role of neurons in controlling motor activity. The deep penetrance NIR light makes the current system feasible for in vivo deep-tissue neuron elimination. The results not only provide a rapidly adoptable platform to efficient photoablate single- and multiple-cells, but also define the neural circuits underlying behavior, with potential for development of remote therapy in diseases.


Asunto(s)
Técnicas de Ablación , Locomoción , Nanopartículas , Neuronas , Técnicas de Ablación/métodos , Animales , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/efectos de la radiación , Rayos Infrarrojos , Luz , Locomoción/efectos de los fármacos , Nanopartículas/química , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/efectos de la radiación , Optogenética , Oxígeno Singlete/química
15.
Acc Chem Res ; 52(9): 2703-2712, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31433171

RESUMEN

The past decades have witnessed the development of a field dedicated to targeting tumor vasculature for cancer therapy. In contrast to conventional chemotherapeutics that need to penetrate into tumor tissues for killing tumor cells, the agents targeting tumor vascular system have two major advantages: direct contact with vascular endothelial cells or the blood and less possibility to induce drug resistance because of high gene stability of endothelial cells. More specifically, various angiogenesis inhibitors (AIs) and vascular disrupting agents (VDAs) that block tumor blood supply to inhibit tumor progression, some of which have been applied clinically, have been described. However, off-target effects and high effective doses limit the utility of these formulations in cancer patients. Thus, new strategies with improved therapeutic efficacy and safety are needed for tumor vessel targeting therapy. With the burgeoning developments in nanotechnology, smart nanotherapeutics now offer unprecedented potential for targeting tumor vasculature. Based on specific structural and functional features of the tumor vasculature, a number of different nanoscale delivery systems have been proposed for cancer therapy. In this Account, we summarize several distinct strategies to modulate tumor vasculature with various smart nanotherapeutics for safe and effective tumor therapy developed by our research programs. Inspired by the blood coagulation cascade, we generated nanoparticle-mediated tumor vessel infarction strategies that selectively block tumor blood supply to starve the tumor to death. By specifically delivering thrombin loaded DNA nanorobots (Nanorobot-Th) into tumor vessels, an intratumoral thrombosis is triggered to induce vascular infarction and, ultimately, tumor necrosis. Mimicking the coagulation cascade, a smart polymeric nanogel achieves permanent and peripheral embolization of liver tumors. Considering the critical role of platelets in maintaining tumor vessel integrity, a hybrid (PLP-D-R) nanoparticle selectively depleting tumor-associated platelets (TAP) to boost tumor vessel permeability was developed for enhancing intratumoral drug accumulation. In addition, benefiting from a better understanding of the molecular and cellular underpinnings of vascular normalization, several tumor acidity responsive nanotherapeutics, encapsulating therapeutic peptides, and small interfering RNA were developed to correct the abnormal features of the tumor vasculature. This made the tumor vessels more efficient for drug delivery. While we are still exploring the mechanisms of action of these novel nanoformulations, we expect that the strategies summarized here will offer a promising platform to design effective next-generation nanotherapeutics against cancer and facilitate the clinical translation of smart nanotherapeutics that target tumor vasculature.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Nanopartículas/química , Nanotecnología , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Humanos , Neoplasias/patología , Neovascularización Patológica/patología
16.
Nano Lett ; 19(11): 8234-8244, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31576757

RESUMEN

Photosensitizers (PSs) that are directly responsive to X-ray for radiodynamic therapy (RDT) with desirable imaging abilities have great potential applications in cancer therapy. Herein, the cerium (Ce)-doped NaCeF4:Gd,Tb scintillating nanoparticle (ScNP or scintillator) is first reported. Due to the sensitization effect of the Ce ions, Tb ions can emit fluorescence under X-ray irradiation to trigger X-ray excited fluorescence (XEF). Moreover, Ce and Tb ions can absorb the energy of secondary electrons generated by X-ray to produce reactive oxide species (ROS) for RDT. With the intrinsic absorption of X-ray by lanthanide elements, the NaCeF4:Gd,Tb ScNPs also act as a computed tomography (CT) imaging contrast agent and radiosensitizers for radiotherapy (RT) sensitization synchronously. Most importantly, the transverse relaxation time of Gd3+ ions is shortened due to the doping of Ce and Tb ions, leading to the excellent performance of our ScNPs in T2-weighted MR imaging for the first time. Both in vitro and in vivo studies verify that our synthesized ScNPs have good performance in XEF, CT, and T2-weighted MR imaging, and a synchronous RT/RDT is achieved with significant suppression on tumor progression under X-ray irradiation. Importantly, no systemic toxicity is observed after intravenous injection of ScNPs. Our work highlights that ScNPs have potential in multimodal imaging-guided RT/RDT of deep tumors.


Asunto(s)
Elementos de la Serie de los Lantanoides/uso terapéutico , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/terapia , Nanopartículas/uso terapéutico , Fármacos Fotosensibilizantes/uso terapéutico , Células A549 , Animales , Cerio/uso terapéutico , Medios de Contraste/uso terapéutico , Humanos , Imagen por Resonancia Magnética , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/ultraestructura , Imagen Óptica , Fotoquimioterapia , Especies Reactivas de Oxígeno/metabolismo , Tomografía Computarizada por Rayos X , Terapia por Rayos X
17.
Nano Lett ; 19(8): 5806-5817, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31331172

RESUMEN

Constructing a tumor microenvironment stimuli activatable theranostic nanoparticle with simple components and preparation procedures for multimodality imaging and therapy remains a major challenge for current theranostic systems. Here we report a novel and simple glutathione (GSH)-responsive turn-on theranostic nanoparticle for dual-modal imaging and combination therapy. The theranostic nanoparticle, DHP, consisting of a disulfide-bond-linked hydroxyethyl starch paclitaxel conjugate (HES-SS-PTX) and a near-infrared (NIR) cyanine fluorophore DiR, is prepared with a simple one-step dialysis method. As DiR is encapsulated within the hydrophobic core formed by HES-SS-PTX, the fluorescence of DiR is quenched by the aggregation-caused quenching (ACQ) effect. Nonetheless, once DHP is internalized by cancer cells, the disulfide bond of HES-SS-PTX can be cleaved by intracellular GSH, leading to the synchronized release of conjugated PTX and loaded DiR. The released PTX could exert its therapeutic effect, while DiR could adsorb onto nearby endosome/lysosome membranes and regain its fluorescence. Thus, DHP could monitor the release and therapeutic effect of PTX through the fluorescence recovery of DiR. Remarkably, DHP can also be used as an in vivo probe for both fluorescent and photoacoustic imaging and at the same time achieves potent antitumor efficacy through chemo-photothermal combination therapy. This study provides novel insights into designing clinically translatable turn-on theranostic systems.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Colorantes Fluorescentes/uso terapéutico , Glutatión/metabolismo , Nanopartículas/uso terapéutico , Neoplasias/terapia , Paclitaxel/uso terapéutico , Animales , Línea Celular Tumoral , Terapia Combinada , Hipertermia Inducida , Ratones , Ratones Endogámicos BALB C , Neoplasias/diagnóstico por imagen , Neoplasias/metabolismo , Imagen Óptica , Técnicas Fotoacústicas , Fototerapia , Nanomedicina Teranóstica
18.
Nano Lett ; 19(9): 5904-5914, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31389707

RESUMEN

Sensory neurons promote profound suppressive effects on neutrophils during Streptococcus pyogenes infection and contribute to the pathogenesis of necrotizing infection ("flesh-eating disease"). Thus, the development of new antibacterial agents for necrotizing infection is promising because of the clear streptococcal neuro-immune communication. Herein, based on the immune escape membrane exterior and competitive membrane functions of the glioma cell membrane, a novel nano neuro-immune blocker capsule was designed to prevent neuronal activation and improve neutrophil immune responses for necrotizing infection. These nano neuro-immune blockers could neutralize streptolysin S, suppress neuron pain conduction and calcitonin gene-related peptide release, and recruit neutrophils to the infection site, providing a strong therapeutic effect against necrotizing infection. Furthermore, nano neuro-immune blockers could serve as an effective inflammatory regulator and antibacterial agent via photothermal effects under near-infrared irradiation. In the Streptococcus pyogenes-induced necrotizing fasciitis mouse model, nano neuro-immune blockers showed significant therapeutic efficacy by ameliorating sensitivity to pain and promoting the antibacterial effect of neutrophils.


Asunto(s)
Antibacterianos/farmacología , Inflamación/tratamiento farmacológico , Necrosis/tratamiento farmacológico , Dolor/tratamiento farmacológico , Animales , Antibacterianos/química , Antibacterianos/efectos de la radiación , Proteínas Bacterianas/antagonistas & inhibidores , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/efectos de la radiación , Inflamación/microbiología , Luz , Ratones , Necrosis/microbiología , Neuroinmunomodulación/efectos de los fármacos , Neuroinmunomodulación/efectos de la radiación , Neuronas/efectos de los fármacos , Neuronas/microbiología , Neutrófilos/efectos de los fármacos , Neutrófilos/microbiología , Dolor/microbiología , Streptococcus pyogenes/efectos de los fármacos , Streptococcus pyogenes/patogenicidad , Estreptolisinas/antagonistas & inhibidores
19.
J Am Chem Soc ; 141(51): 20097-20106, 2019 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-31721575

RESUMEN

Simple, rapid, and sensitive assays of DNA sequence hold great importance in genetic analysis, clinical diagnosis, and molecular biology research. Most current methods for DNA detection, based on the complementary base pairing, require hybridization with intricately modified single-stranded DNA (ssDNA) probes or analytes. Herein, we have developed a powerful molecule with aggregation-induced emission (AIE) characteristic, namely, TPBT, which can specifically recognize double-stranded DNA (dsDNA) by emitting out a unique dual-color fluorescent signal of red (∼640 nm) and green (∼537 nm). The red-color emission at around 640 nm is observed when TPBT binds with dsDNA, ssDNA, proteins, and other polyanionic analytes. However, the green emission at around 537 nm is demonstrated to be the exclusive response of TPBT to dsDNA, which is closely related to the conformational change of TPBT upon groove binding. More strikingly, TPBT can distinguish single-nucleotide polymorphisms (SNPs) in a dsDNA sequence and detect the DNA damage suffered from UV light with ultrahigh sensitivity and specificity. This label-free, AIEgen-based dsDNA assay method is facile, robust, and universal, which will lead to major advances in genomic and disease diagnosis.


Asunto(s)
Color , ADN/análisis , Colorantes Fluorescentes/química , Fármacos Fotosensibilizantes/química , Estructura Molecular
20.
Small ; 15(46): e1902647, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31614073

RESUMEN

Integrating multiple strategies of antibacterial mechanisms into one has been proven to have tremendous promise for improving antimicrobial efficiency. Hence, dual-valent platinum nanoparticles (dvPtNPs) with a zero-valent platinum core (Pt0 ) and bi-valent platinum shell (Pt2+ ions), combining photothermal and photodynamic therapy, together with "chemotherapy," emerge as spatiotemporally light-activatable platinum nano-antibiotics. Under near-infrared (NIR) exposure, the multiple antibacterial modes of dvPtNPs are triggered. The Pt0 core reveals significant hyperthermia via effective photothermal conversion while an immediate release of chemotherapeutic Pt2+ ions occurs through hyperthermia-initiated destabilization of metallic interactions, together with reactive oxygen species (ROS) level increase, thereby resulting in synergistic antibacterial effects. The precise cooperative effects between photothermal, photodynamic, and Pt2+ antibacterial effects are achieved on both Gram-negative Escherichia coli and Gram-positive methicillin-resistant Staphylococcus aureus, where bacterial viability and colony-forming units are significantly reduced. Moreover, similar results are observed in mice subcutaneous abscess models. Significantly, after NIR treatment, dvPtNP exhibits a more robust bacteria-killing efficiency than other PtNP groups, owing to its integration of dramatic damage to the bacterial membrane and DNA, and alteration to ATP and ROS metabolism. This study broadens the avenues for designing and synthesizing antibacterial materials with higher efficiency.


Asunto(s)
Antibacterianos/farmacología , Luz , Nanopartículas/química , Platino (Metal)/farmacología , Animales , Escherichia coli/efectos de los fármacos , Escherichia coli/ultraestructura , Femenino , Nanopartículas del Metal/ultraestructura , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/ultraestructura , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Células 3T3 NIH
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA