Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Adv Healthc Mater ; : e2401743, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39015058

RESUMO

The transformation of monotherapy into multimodal combined targeted therapy to fully exploit synergistic efficacy is of increasing interest in tumor treatment. In this work, a novel nanodrug-carrying platform based on iron-based MOFs, which is loaded with doxorubicin hydrochloride (DOX), dihydroartemisinin (DHA), and glucose oxidase (GOx), and concurrently covalently linked to the photosensitizer 5,10,15,20-tetrakis(4-carboxyphenyl)porphyrin (TCPP) in polydopamine (PDA)-encapsulated MIL-101(Fe) (denoted as MIL-101(Fe)-DOX-DHA@TCPP/GOx@PDA, MDDTG@P), is successfully developed. Upon entering the tumor microenvironment, MDDTG@P catalyzes the hydrogen peroxide (H2O2) into hydroxyl radicals (·OH) and depletes glutathione (GSH); thus, exerting the role of chemodynamic therapy (CDT). The reduced Fe2+ can also activate DHA, further expanding CDT and promoting tumor cell apoptosis. The introduced GOx will rapidly consume glucose and oxygen (O2) in the tumor; while, replenishing H2O2 for Fenton reaction, starving the cancer cells; and thus, realizing starvation and chemodynamic therapy. In addition, the covalent linkage of TCPP endows MDDTG@P with good photodynamic therapeutic (PDT) properties. Therefore, this study develops a nanocarrier platform for triple synergistic chemodynamic/photodynamic/starvation therapy, which has promising applications in the efficient treatment of tumors.

2.
Small ; : e2402312, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39077967

RESUMO

Reactive oxygen species (ROS)-dependent monotherapy usually demonstrates poor therapeutic outcomes, due to the accompanied activation of protective autophagy in tumor cells, which results in ROS tolerance and immune suppression. In this study, a bimetallic electro-sensitizer, Pt-Ir NPs is constructed, loaded with the autophagy inhibitor chloroquine (Pt-Ir-CQ NPs), to enhance the effectiveness of electrotherapy by inhibiting autophagy and activating anti-tumor immune responses. This novel electrotherapy platform demonstrates unique advantages, particularly in the treatment of hypoxic and immunosuppressive tumors. First, the electro-sensitizer catalyzes water molecules into ROS under electric field, achieving tumor ablation through electrotoxicity. Second, the incorporated CQ inhibits the protective autophagy induced by electrotherapy, restoring the sensitivity of tumor cells to ROS and thereby enhancing the anti-tumor effects of electrotherapy. Third, Pt-Ir-CQ NPs enhance the functionality of antigen-presenting cells and immunogenic cells through inhibiting autophagy, synergistically activating the anti-tumor immune responses along with the immunogenic cell death (ICD) effect induced by electrotherapy. This study provides a novel approach for the effective ablation and long-term inhibition of solid tumors through flexible modulation by an exogenous electric field.

3.
ACS Nano ; 18(26): 16359-16394, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38861272

RESUMO

Glioblastoma (GBM) remains the epitome of aggressiveness and lethality in the spectrum of brain tumors, primarily due to the blood-brain barrier (BBB) that hinders effective treatment delivery, tumor heterogeneity, and the presence of treatment-resistant stem cells that contribute to tumor recurrence. Nanoparticles (NPs) have been used to overcome these obstacles by attaching targeting ligands to enhance therapeutic efficacy. Among these ligands, peptides stand out due to their ease of synthesis and high selectivity. This article aims to review single and multiligand strategies critically. In addition, it highlights other strategies that integrate the effects of external stimuli, biomimetic approaches, and chemical approaches as nanocatalytic medicine, revealing their significant potential in treating GBM with peptide-functionalized NPs. Alternative routes of parenteral administration, specifically nose-to-brain delivery and local treatment within the resected tumor cavity, are also discussed. Finally, an overview of the significant obstacles and potential strategies to overcome them are discussed to provide a perspective on this promising field of GBM therapy.


Assuntos
Barreira Hematoencefálica , Neoplasias Encefálicas , Glioblastoma , Nanopartículas , Peptídeos , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/metabolismo , Humanos , Peptídeos/química , Peptídeos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Nanopartículas/química , Barreira Hematoencefálica/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/administração & dosagem , Animais , Sistemas de Liberação de Medicamentos
4.
Angew Chem Int Ed Engl ; 63(6): e202316858, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38095801

RESUMO

Nanocatalytic tumor therapy based on Fenton nanocatalysts has attracted considerable attention because of its therapeutic specificity, enhanced outcomes, and high biocompatibility. Nevertheless, the rate-determining step in Fenton chemistry, which involves the transition of a high-valence metallic center (FeIII ) to a Fenton-active low-valence metallic center (FeII ), has hindered advances in nanocatalyst-based therapeutics. In this study, we constructed mesoporous single iron atomic nanocatalysts (mSAFe NCs) by employing catechols from dopamine to coordinate and isolate single iron atoms. The catechols also serve as reductive ligands, generating a field-effect-based cocatalytic system that instantly reduces FeIII species to FeII species within the mSAFe NCs. This self-motivated cocatalytic strategy enabled by mSAFe NCs accelerates the kinetics of the Fenton catalytic reaction, resulting in remarkable performance for nanocatalytic tumor therapy both in vitro and in vivo.


Assuntos
Compostos Férricos , Neoplasias , Humanos , Ferro , Neoplasias/tratamento farmacológico , Compostos Ferrosos , Catecóis , Peróxido de Hidrogênio , Catálise
5.
ACS Appl Mater Interfaces ; 15(51): 59175-59188, 2023 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-38095444

RESUMO

Generating lethal reactive oxygen species (ROS) within tumors by nanocatalytic medicines is an advanced strategy for tumor-specific therapy in recent years. Nevertheless, the low yield of ROS restrains its therapeutic efficiency. Herein, a dual-catalytic nanomedicine based on tumor microenvironment (TME)-responsive liposomal nanosystem co-delivering CuO2 and dihydroartemisinin (DHA) (LIPSe@CuO2&DHA) is developed to boost ROS generation against tumor. The liposomal nanosystem can degrade in the ROS-overexpressed TME and liberate CuO2 and DHA to initiate Cu-based dual-catalytic ROS generation. Serving as generators of H2O2 and Cu2+, CuO2 can self-produce plenty of toxic hydroxyl radicals via Fenton-like reaction in the acidic TME. Meanwhile, the released Cu2+ can catalyze DHA to generate cytotoxic C-centered radicals. Together, the self-supplied H2O2 and Cu-based dual-catalytic reaction greatly increase the intratumoral level of lethal ROS. Importantly, Cu2+ can decrease the GSH-mediated scavenging effect on the produced ROS via a redox reaction and undergo a Cu2+-to-Cu+ conversion to enhance the Fenton-like reaction, further guaranteeing the high efficiency of ROS generation. Resultantly, LIPSe@CuO2&DHA induces remarkable cancer cell death and tumor growth inhibition, which may present a promising nanocatalytic medicine for cancer therapy.


Assuntos
Nanomedicina , Neoplasias , Humanos , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular Tumoral , Peróxido de Hidrogênio/farmacologia , Neoplasias/patologia , Fototerapia , Microambiente Tumoral , Glutationa/farmacologia
6.
J Nanobiotechnology ; 21(1): 425, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37968644

RESUMO

BACKGROUND: Chemodynamic therapy (CDT) based on Fenton/Fenton-like reaction has emerged as a promising cancer treatment strategy. Yet, the strong anti-oxidation property of tumor microenvironment (TME) caused by endogenous glutathione (GSH) still severely impedes the effectiveness of CDT. Traditional CDT nanoplatforms based on core@shell structure possess inherent interference of different subunits, thus hindering the overall therapeutic efficiency. Consequently, it is urgent to construct a novel structure with isolated functional units and GSH depletion capability to achieve desirable combined CDT therapeutic efficiency. RESULTS: Herein, a surface curvature-induced oriented assembly strategy is proposed to synthesize a sushi-like novel Janus therapeutic nanoplatform which is composed of two functional units, a FeOOH nanospindle serving as CDT subunit and a mSiO2 nanorod serving as drug-loading subunit. The FeOOH CDT subunit is half covered by mSiO2 nanorod along its long axis, forming sushi-like structure. The FeOOH nanospindle is about 400 nm in length and 50 nm in diameter, and the mSiO2 nanorod is about 550 nm in length and 100 nm in diameter. The length and diameter of mSiO2 subunit can be tuned in a wide range while maintaining the sushi-like Janus structure, which is attributed to a Gibbs-free-energy-dominating surface curvature-induced oriented assembly process. In this Janus therapeutic nanoplatform, Fe3+ of FeOOH is firstly reduced to Fe2+ by endogenous GSH, the as-generated Fe2+ then effectively catalyzes overexpressed H2O2 in TME into highly lethal ·OH to achieve efficient CDT. The doxorubicin (DOX) loaded in the mSiO2 subunit can be released to achieve combined chemotherapy. Taking advantage of Fe3+-related GSH depletion, Fe2+-related enhanced ·OH generation, and DOX-induced chemotherapy, the as-synthesized nanoplatform possesses excellent therapeutic efficiency, in vitro eliminating efficiency of tumor cells is as high as ~ 87%. In vivo experiments also show the efficient inhibition of tumor, verifying the synthesized sushi-like Janus nanoparticles as a promising therapeutic nanoplatform. CONCLUSIONS: In general, our work provides a successful paradigm of constructing novel therapeutic nanoplatform to achieve efficient tumor inhibition.


Assuntos
Nanopartículas Multifuncionais , Neoplasias , Humanos , Peróxido de Hidrogênio , Protocolos de Quimioterapia Combinada Antineoplásica , Doxorrubicina/farmacologia , Glutationa , Neoplasias/tratamento farmacológico , Linhagem Celular Tumoral , Microambiente Tumoral
7.
Adv Mater ; 35(48): e2306469, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37669827

RESUMO

Nanotechnology is revolutionizing cancer therapy, and catalyzes the emerging of ion-involved cancer-therapeutic modality, which unfortunately suffers from undesirable nanocarriers for efficient intracellular ion delivery. To radically extricate from this critical issue, the glutathione (GSH)-responsive organosilica network is employed to lock the liquid drops at the nanoscale via a general bottom-up strategy to achieve the systemic delivery of "ion drugs". In this work, a sulfate radical generation donor (Na2 S2 O8 ), as a paradigm "ion drug", is entrapped into this liquid nanoparticle for efficiently delivering to the tumor region. After further surface engineering with pH-responsive tannic acid-Fe2+ framework, these liquid nanoparticles achieve tumor-microenvironmental pH/GSH-dual responsive ion release (Fe2+ /Na+ /S2 O8 2- ) after reaching the tumor sites, where the Fe2+ further triggers S2 O8 2- to generate toxic •SO4 - and •OH, effectively executing cancer cell ferroptosis (Fe2+ , reactive oxygen species-ROS) and pyroptosis (Na+ , ROS). Such a tumor-responsive/specific liquid nanoplatform is highly instructive for further ion-mediated nanomedicine and disease treatment.


Assuntos
Nanopartículas , Neoplasias , Humanos , Espécies Reativas de Oxigênio/uso terapêutico , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Sistemas de Liberação de Medicamentos , Nanomedicina , Linhagem Celular Tumoral , Microambiente Tumoral
8.
Acta Biomater ; 167: 463-472, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37302733

RESUMO

Nitric oxide (NO) is a crucial gaseous medium for tumor growth and progression, but it may also cause mitochondrial disorder and DNA damage by drastically increasing its concentration in tumor. Due to its challenging administration and unpredictable release, NO based gas therapy is difficult to eliminate malignant tumor at low safe doses. To address these issues, herein, we develop a multifunctional nanocatalyst called Cu-doped polypyrrole (CuP) as an intelligent nanoplatform (CuP-B@P) to deliver the NO precursor BNN6 and specifically release NO in tumors. Under the aberrant metabolic environment of tumors, CuP-B@P catalyzes the conversion of antioxidant GSH into GSSG and excess H2O2 into ·OH through Cu+/Cu2+ cycle, which results in oxidative damage to tumor cells and the concomitant release of cargo BNN6. More importantly, after laser exposure, nanocatalyst CuP can absorb and convert photons into hyperthermia, which in turn, accelerates the aforesaid catalytic efficiency and pyrolyzes BNN6 into NO. Under the synergistic effect of hyperthermia, oxidative damage, and NO burst, almost complete tumor elimination is achieved in vivo with negligible toxicity to body. Such an ingenious combination of NO prodrug and nanocatalytic medicine provides a new insight into the development of NO based therapeutic strategies. STATEMENT OF SIGNIFICANCE: A hyperthermia-responsive NO delivery nanoplatform (CuP-B@P) based on Cu-doped polypyrrole was designed and fabricated, in which CuP catalyzed the conversion of H2O2 and GSH into ·OH and GSSG to induce intratumoral oxidative damage. After laser irradiation, hyperthermia ablation and responsive release of NO further coupled with oxidative damage to eliminate malignant tumors. This versatile nanoplatform provides new insights into the combined application of catalytic medicine and gas therapy.


Assuntos
Hipertermia Induzida , Nanopartículas , Neoplasias , Humanos , Polímeros , Pirróis , Óxido Nítrico , Fototerapia , Hipertermia Induzida/métodos , Peróxido de Hidrogênio , Dissulfeto de Glutationa , Catálise , Linhagem Celular Tumoral
9.
Natl Sci Rev ; 10(5): nwad063, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37056424

RESUMO

It is a great challenge to effectively eradicate biofilm and cure biofilm-infected diseases because dense extracellular polymeric substance matrix prevents routine antibacterial agents from penetrating into biofilm. H2 is an emerging energy-regulating molecule possessing both high biosafety and high tissue permeability. In this work, we propose a concept of sonocatalytic hydrogen/hole-combined 'inside/outside-cooperation' anti-biofilm for promoting bacteria-infected diabetic wound healing based on two-dimensional piezoelectric nanomaterials. Proof-of-concept experiments using C3N4 nanosheets as a representative piezoelectric catalyst with wide band gap and high biosafety have verified that sonocatalytically generated H2 and holes rapidly penetrate into biofilm to inhibit bacterial energy metabolism and oxidatively deprive polysaccharides/NADH in biofilm to destroy the bacterial membrane/electron transport chain, respectively, inside/outside-cooperatively eradicating biofilm. A bacteria-infected diabetic wound model is used to confirm the excellent in vivo antibacterial performance of sonocatalytic hydrogen/hole-combined therapy, remarkably improving bacteria-infected diabetic wound healing. The proposed strategy of sonocatalytic hole/hydrogen-combined 'inside/outside-cooperation' will make a highway for treatment of deep-seated biofilm infection.

10.
Mater Today Bio ; 17: 100462, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36325424

RESUMO

As one of the most common representations of articular cartilage damage, osteoarthritis (OA) is characterized by the apoptosis and dysfunction of chondrocytes as well as the progressive degradation of extracellular matrix, of which the main components are glycosaminoglycan and type Ⅱ collagen. Few-layered phosphorene (FLP) has been attracting great attentions in biomedical fields owing to the excellent capability of in-situ catalysis for scavenging oxidate-associated molecules, especially the reactive oxygen species (ROS) and reactive nitrogen species (RNS). Herein, FLP has been fabricated and employed for articular cartilage protection by means of deleting oxidate-associated molecules. The in vitro results show that as low as 200 â€‹µg/mL FLP is capable of diminishing oxidative damages on the osteoarthritic chondrocytes through the efficient elimination of ROS, H2O2 and NO. Meanwhile, the cartilage matrix protection has also been achieved at 200 â€‹µg/mL FLP by the uniform restoration of glycosaminoglycan and type Ⅱ collagen. FLP enables the nanocatalytic treatment for the overloaded oxidative stress in the injured articular cartilage and represents a promising alternative for osteoarthritis therapy.

11.
Natl Sci Rev ; 9(10): nwac022, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36415318

RESUMO

Nanocatalysts with enzyme-like catalytic activities, such as oxidase mimics, are extensively used in biomedicine and environmental treatment. Searching for enzyme-like nanomaterials, clarifying the origins of catalytic activity and developing activity assessment methodologies are therefore of great significance. Here, we report that oxidase catalysis and oxygen reduction reaction (ORR) electrocatalysis can be well bridged based on their identical activity origins, which makes facile electrocatalytic ORR activity measurements intrinsically applicable to oxidase-like activity evaluations. Inspired by natural heme-copper oxidases, Cu/Fe-doped single-atom catalysts (SACs) were first synthesized and used as model catalysts. Chromogenic reactions, electrochemical voltammetric measurements and density functional theory calculations further verified the linear relationship between the oxidase-like and ORR catalytic activities of the catalysts; thus, an effective descriptor ([Formula: see text]) is proposed for rapid enzymatic catalyst evaluation. Evidence suggests that the enhanced tumour therapeutic efficacy of SACs is a result of their oxidase-like/ORR activities, which proves that numerous ORR electrocatalysts are promising candidates for oxidase mimics and tumour therapy. The synergistic catalytic effect of the biomimetic heterobinuclear Cu-Fe centres has also been thoroughly probed.

12.
Biomaterials ; 290: 121816, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36201946

RESUMO

The absence of lymphatic vessels in tumors leads to the retention of interstitial fluid, and the formation of an inverse pressure difference between the tumor and blood vessels hinders drug delivery deep into the tumor, which leads to tumor recurrence and metastasis. Therefore, we designed a novel strategy to downregulate tumor interstitial fluid pressure (TIFP) by water splitting in the tumor interstitium based on piezoelectric catalysis nanomedicine. First, the chemotherapeutic drug doxorubicin (DOX) was loaded on the piezoelectric catalytic material MoS2 and then encapsulated with tumor cell membrane (CM) to obtain MD@C. MD@C could not only target the tumor through homologous targeting but, more importantly, also triggered piezoelectric catalytic water splitting under ultrasound (US) stimulation; as a result, the TIFPs of U14 and PAN02 tumor-bearing mice were reduced to 57.14% and 45.5%, respectively, and the tumor inhibition rates of MD@C were 96.75% and 99.21%, which increased the perfusion of blood-derived drugs in the tumors. Moreover, the hydroxyl radicals generated by piezoelectric catalysis could effectively inhibit the growth of tumors in combination with DOX. Consequently, the piezoelectric catalytic water splitting strategy of MD@C can enhance drug delivery, providing a new universal platform for the treatment of solid malignant tumors.


Assuntos
Nanopartículas , Neoplasias , Camundongos , Animais , Molibdênio , Doxorrubicina/uso terapêutico , Doxorrubicina/farmacologia , Nanomedicina , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Catálise , Água , Linhagem Celular Tumoral , Nanopartículas/uso terapêutico
13.
Small ; 18(29): e2202964, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35717674

RESUMO

Nanocatalytic medicine is a burgeoning disease treatment model with high specificity and biosafety in which the nanocatalyst is the core of driving catalytic reaction to generate therapeutic outcomes. However, the robust defense systems in the pathological region would counteract nanocatalyst-initiated therapeutics. Here, a Cu-doped polypyrrole is innovatively developed by a facile oxidative polymerization reaction, which exhibits intriguing multi-catalytic activities, including catalyzing H2 O2 to generate O2 and · OH, and consuming reduced glutathione by a Cu(II)-Cu(I) transition approach. By decorating with sonosensitizers and DSPE-PEG, the obtained CuPPy-TP plus US irradiation can induce severe oxidative damage to tumor cells by amplifying oxidative stress and simultaneously relieving antioxidant capacity in tumors based on the highly effective sonochemical and redox reactions. The notable tumor-specific biodegradability, remarkable cell apoptosis in vitro, and tumor suppression in vivo are demonstrated in this work, which not only present a promising biocompatible antitumor nanocatalyst but also broaden the perspective in oxidative stress-based antitumor therapy.


Assuntos
Polímeros , Pirróis , Catálise , Linhagem Celular Tumoral , Peróxido de Hidrogênio/farmacologia , Polímeros/farmacologia , Microambiente Tumoral
14.
ACS Nano ; 15(10): 16286-16297, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34652919

RESUMO

Nanocatalytic medicine is one of the most recent advances in the development of nanomedicine, which catalyzes intratumoral chemical reactions to produce toxins such as reactive oxygen species in situ for cancer specific treatment by using exogenous-delivered catalysts such as Fenton agents. However, the overexpression of reductive glutathione and Cu-Zn superoxide dismutase in cancer cells will significantly counteract the therapeutic efficacy by reactive oxygen species-mediated oxidative damages. Additionally, the direct delivery of iron-based Fenton agents may arouse undesired detrimental effects such as anaphylactic reactions. In this study, instead of exogenously delivering Fenton agents, the endogenous copper ions from intracellular Cu-Zn superoxide dismutase have been employed as the source of Fenton-like agents by chelating the Cu ions from the superoxide dismutase using a common metal ion chelator, N,N,N',N'-tetrakis(2-pyridinylmethyl)-1,2-ethanediamine (TPEN), followed by the TPEN-Cu(II) chelate reduction to TPEN-Cu(I) by reductive glutathione. Briefly, TPEN was loaded in a disulfide bond-containing link poly(acrylic acid) shell-coated hybrid mesoporous silica/organosilicate (MSN@MON) nanocomposite as a reductive glutathione-responsive nanoplatform, which features inter-related triple functions: intratumoral reductive glutathione-responsive link poly(acrylic acid) disruption and TPEN release; the accompanying reductive glutathione consumption and Cu-Zn superoxide dismutase deactivation by TPEN chelating Cu ions from this superoxide dismutase; and the Fenton reaction catalyzed by TPEN-Cu(I) chelate as a Fenton-like agent generated from TPEN-Cu(II) reduction by the remaining reductive glutathione in cancer cells, thereby cutting off the self-protection pathway of cancer cells under severe oxidation stress and ensuring cancer cell apoptosis by reactive oxygen species produced by the catalytic Fenton-like reactions. Such a nanocatalyst demonstrates excellent biosafety and augmented therapeutic efficacy by simultaneous nanocatalytic oxidative damage and intrinsic protection pathway breakage of cancer cells.


Assuntos
Cobre , Neoplasias , Quelantes , Humanos , Neoplasias/tratamento farmacológico , Estresse Oxidativo , Superóxido Dismutase/metabolismo
15.
Int J Nanomedicine ; 16: 5193-5209, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34354353

RESUMO

BACKGROUND: Recently, nanocatalyst-induced endoplasmic reticulum (ER) stress for cancer therapy has been attracting considerable attention. However, cancer cells are often able to overcome ER stress-induced death by activating the unfolded protein response (UPR), making nanocatalytic monotherapy a poor defense against cancer progression. PURPOSE: In this study, to improve the nanocatalytic treatment efficacy, a phase change material (PCM) was used to encapsulate the upstream ER stress initiator, iron oxide nanoparticles (Fe3O4 NPs), and the downstream UPR modulator, PR-619. Subsequently, the tumor-homing peptide tLyP-1 was coupled to it to form tLyP-1/PR-619/Fe3O4@PCM (tPF@PCM) theranostic platform. MATERIALS AND METHODS: tPF@PCM was synthesized using nanoprecipitation and resolidification methods followed by the EDC/NHS cross-linking method. The targeting capacity of tPF@PCM was evaluated in vitro and in vivo using flow cytometry and magnetic resonance imaging, respectively. The therapeutic efficacy of tPF@PCM was investigated in a renal cell carcinoma mouse model. Moreover, we explored the synergistic anti-tumor mechanism by examining the intracellular reactive oxygen species (ROS), aggregated proteins, ER stress response levels, and type of cell death. RESULTS: tPF@PCM had excellent tumor-targeting properties and exhibited satisfactory photothermal-enhanced tumor inhibition efficacy both in vitro and in vivo. Specifically, the phase transition temperature (45 °C) maintained using 808 nm laser irradiation significantly increased the release and catalytic activity of the peroxidase mimic Fe3O4 NPs. This strongly catalyzed the generation of hydroxyl radicals (•OH) via the Fenton reaction in the acidic tumor microenvironment. The redox imbalance subsequently resulted in an increase in the level of damaged proteins in the ER and initiated ER stress. Moreover, the pan-deubiquitinase inhibitor PR-619 blocked the "adaptive" UPR-mediated degradation of these damaged proteins, exacerbating the ER burden. Consequently, irremediable ER stress activated the "terminal" UPR, leading to apoptosis in cancer cells. CONCLUSION: This ER stress-exacerbating strategy effectively suppresses tumorigenesis, offering novel directions for advances in the treatment of conventional therapy-resistant cancers.


Assuntos
Retículo Endoplasmático , Neoplasias , Animais , Apoptose , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Humanos , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Resposta a Proteínas não Dobradas
16.
Adv Sci (Weinh) ; 8(9): 2002816, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33977044

RESUMO

Elevating intratumoral levels of highly toxic reactive oxygen species (ROS) by nanocatalytic medicine for tumor-specific therapy without using conventional toxic chemodrugs is recently of considerable interest, which, however, still suffers from less satisfactory therapeutic efficacy due to the relatively poor accumulation at the tumor site and largely blocked intratumoral infiltration of nanomedicines. Herein, an ultrasound (US)-triggered dual size/charge-switchable nanocatalytic medicine, designated as Cu-LDH/HMME@Lips, is constructed for deep solid tumor therapy via catalytic ROS generations. The negatively charged liposome outer-layer of the nanomedicine enables much-prolonged blood circulation for significantly enhanced tumoral accumulation, while the positively charged Fenton-like catalyst Cu-LDH released from the liposome under the US stimulation demonstrates much enhanced intratumoral penetration via transcytosis. In the meantime, the co-released sonosensitizer hematoporphyrin monomethyl ether (HMME) catalyze the singlet oxygen (1O2) generation upon the US irradiation, and deep-tumoral infiltrated Cu-LDH catalyzes the H2O2 decomposition to produce highly toxic hydroxyl radical (·OH) specifically within the mildly acidic tumor microenvironment (TME). The efficient intratumoral accumulation and penetration via the dual size/charge switching mechanism, and the ROS generations by both sonosensitization and Fenton-like reactions, ensures the high therapeutic efficacy for the deep tumor therapy by the nanocatalytic medicine.


Assuntos
Cobre/administração & dosagem , Hematoporfirinas/administração & dosagem , Hidróxidos/administração & dosagem , Nanomedicina/métodos , Neoplasias/terapia , Espécies Reativas de Oxigênio/metabolismo , Terapia por Ultrassom/métodos , Catálise , Linhagem Celular Tumoral , Humanos , Técnicas In Vitro , Nanomedicina/instrumentação , Nanopartículas , Microambiente Tumoral/efeitos dos fármacos
17.
Adv Healthc Mater ; 10(11): e2001819, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33857353

RESUMO

Nanocatalytic medicine has been emerging as a highly promising strategy for cancer therapeutics since it enables tumor suppression by in situ generating toxic agents within tumors through catalytic reactions without using conventional highly toxic and nonselective chemodrugs. In the last several years, a number of nanocatalytic medicines have been used to steer catalytic reactions in endogenous or exogenous stimuli-activated cancer therapy, such as chemodynamic therapy, photodynamic therapy, and sonodynamic therapy. In particular, transitional metal-based nanocatalytic medicines with excellent catalytic activity and selectivity show significant clinical potentials, and significant progress has been achieved very recently. In this review, three types of typical transitional metal (Fe, Mn, and Cu)-based nanocatalytic medicines are summarized, followed by detailed discussions on their catalytic mechanisms. Of note, the obstacles and challenges that will be encountered in the design and further clinical conversion of transitional metal-based nanocatalytic medicine in the future are also outlooked.


Assuntos
Hipertermia Induzida , Neoplasias , Catálise , Humanos , Peróxido de Hidrogênio , Neoplasias/tratamento farmacológico , Fototerapia
18.
Adv Mater ; 32(36): e2002246, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32705751

RESUMO

The oxidation of intracellular biomolecules by reactive oxygen species (ROS) forms the basis for ROS-based tumor therapy. However, the current therapeutic modalities cannot catalyze H2 O2 and O2 concurrently for ROS generation, thereby leading to unsatisfactory therapeutic efficacy. Herein, it is reported a bioinspired hollow N-doped carbon sphere doped with a single-atom copper species (Cu-HNCS) that can directly catalyze the decomposition of both oxygen and hydrogen peroxide to ROS, namely superoxide ion (O2 •- ) and the hydroxyl radical (•OH), respectively, in an acidic tumor microenvironment for the oxidation of intracellular biomolecules without external energy input, thus resulting in an enhanced tumor growth inhibitory effect. Notably, the Fenton reaction turnover frequency of Cu species in Cu-HNCS is ≈5000 times higher than that of Fe in commercial Fe3 O4 nanoparticles. Experimental results and density functional theory calculations reveal that the high catalytic activity of Cu-HNCS originates from the single-atom copper, and the calculation predicts a next-generation Fenton catalyst. This work provides an effective paradigm of tumor parallel catalytic therapy for considerably enhanced therapeutic efficacy.


Assuntos
Materiais Biomiméticos/química , Cobre/química , Materiais Biomiméticos/uso terapêutico , Catálise , Linhagem Celular Tumoral , Teoria da Densidade Funcional , Humanos , Peróxido de Hidrogênio/metabolismo , Modelos Moleculares , Conformação Molecular , Oxirredução
19.
J Control Release ; 321: 734-743, 2020 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-32145265

RESUMO

Tumor microenvironment (TME)-responsive nanoformulations that catalyze a cascade of intracellular redox reactions showed promise for tumor treatment with high specificity and efficiency. In this study, we report Cu2+-doped zeolitic imidazolate frameworks-coated polydopamine nanoparticles (PDA@Cu/ZIF-8 NPs) for glutathione-triggered and photothermal-reinforced sequential catalytic therapy against breast cancer. In the TME, the PDA@Cu/ZIF-8 NPs could initially react with antioxidant glutathione (GSH), inducing GSH depletion and Cu+ generation. Whereafter, the generated Cu+ would catalyze local H2O2 to produce highly toxic hydroxyl radicals (·OH) through an efficient Fenton-like reaction even in weakly acidity. Importantly, the PDA could exert excellent photothermal conversion effect to simultaneously accelerate GSH consumption and improve the Fenton-like reaction for further expanding the intracellular oxidative stress, which innovatively achieves a synergistic photothermal-chemodynamic therapy for highly efficient anticancer treatment.


Assuntos
Neoplasias da Mama , Glutationa , Nanopartículas , Microambiente Tumoral , Neoplasias da Mama/terapia , Catálise , Linhagem Celular Tumoral , Glutationa/farmacologia , Humanos , Peróxido de Hidrogênio
20.
Adv Mater ; 32(12): e1907152, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32053261

RESUMO

Nanocatalytic medicine has been developed recently to trigger intratumoral generation of highly toxic reactive oxygen species (ROS) for cancer therapy, which, unfortunately, suffers from compromised therapeutic efficacy due to a self-protective mechanism, autophagy, of cancer cells to mitigate oxidative damage. In this work, during the efforts of ROS generation by nanocatalytic medicine, a pharmacological autophagy inhibition strategy is implemented for augmenting ROS-induced oxidative damage for synergetic cancer therapy. An iron-containing metal-organic framework [MOF(Fe)] nanocatalyst as a peroxidase mimic is used to catalyze the generation of highly oxidizing •OH radicals specifically within cancer cells, while chloroquine is applied to deacidify lysosomes and inhibit autophagy, cutting off the self-protection pathway under severe oxidative stress. Cancer cells fail to extract their components to detoxicate and strengthen themselves, finally succumbing to the ROS-induced oxidative damage during nanocatalytic therapy. Both in vitro and in vivo results demonstrate the synergy between nanocatalytic therapy and autophagy inhibition, suggesting that such a combined strategy is applicable to amplify tumor-specific oxidative damage and may be informative to future design of therapeutic regimen.


Assuntos
Autofagia/efeitos dos fármacos , Cloroquina/farmacologia , Estruturas Metalorgânicas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Catálise , Linhagem Celular Tumoral , Cloroquina/química , Cloroquina/uso terapêutico , Sinergismo Farmacológico , Feminino , Humanos , Peróxido de Hidrogênio/química , Radical Hidroxila/química , Radical Hidroxila/metabolismo , Radical Hidroxila/farmacologia , Ferro/química , Estimativa de Kaplan-Meier , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/uso terapêutico , Camundongos , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/mortalidade , Neoplasias/patologia , Estresse Oxidativo , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA