Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Adv Mater ; 36(23): e2310875, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38450765

RESUMO

Photodynamic therapy (PDT) has been approved for clinic. However, powerless efficiency for deep hypoxic tumor therapy remains an enormous challenge for PDT. Herein, a hypoxia-sensitive nanotherapeutic system (FTCD-SRGD) based on fullerene (C70) and anoxic activating chemical prodrug tirapazamine (TPZ) is rationally designed for multimodal therapy of deep hypoxic tumors. To enhance the accumulation and achieve specific drug release in tumor, the FTCD-SRGD is modified with cyclo(Arg-Gly-Asp-d-Phe-Lys) (cRGDfK) peptide and disulfide bonds. With the exacerbated hypoxic microenvironment created by C70 consuming O2 for generating reactive oxygen species (ROS), TPZ is activated to produce toxic radical species to ablate deep tumors, which achieves a synergistic treatment of C70-mediated PDT and hypoxia-enhanced chemotherapy. Additionally, given this hypoxia-sensitive system-induced immunogenic cell death (ICD) activating anticancer cytotoxic T lymphocyte to result in more susceptible tumor to immunotherapy, FTCD-SRGD plus immune checkpoint inhibitor (anti-PD-L1) fully inhibit deep hypoxic tumors by promoting infiltration of effector T cells in tumors. Collectively, it is the first time to develop a multimodal therapy system with fullerene-based hypoxia-sensitive PS for deep tumors. The powerful multimodal nanotherapeutic system for combining hypoxia-enhanced PDT and immunotherapy to massacre deep hypoxic tumors can provide a paradigm to combat the present bottleneck of tumor therapy.


Assuntos
Fulerenos , Fotoquimioterapia , Fármacos Fotossensibilizantes , Tirapazamina , Fulerenos/química , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Animais , Fotoquimioterapia/métodos , Camundongos , Linhagem Celular Tumoral , Tirapazamina/química , Tirapazamina/farmacologia , Humanos , Terapia Combinada , Microambiente Tumoral/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Neoplasias/patologia , Hipóxia Tumoral/efeitos dos fármacos , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico
2.
Adv Mater ; 36(21): e2312440, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38332741

RESUMO

Delayed re-epithelization and weakened skin contractions are the two primary factors that hinder wound closure in large-scale acute or chronic wounds. However, effective strategies for targeting these two aspects concurrently are still lacking. Herein, an antioxidative active-shrinkage hydrogel (AHF@AS Gel) is constructed that can integratedly promote re-epithelization and skin constriction to accelerate large-scale acute and diabetic chronic wound closure. The AHF@AS Gel is encapsulated by antioxidative amino- and hydroxyl-modified C70 fullerene (AHF) and a thermosensitive active shrinkage hydrogel (AS Gel). Specifically, AHF relieves overactivated inflammation, prevents cellular apoptosis, and promotes fibroblast migration in vitro by reducing excessive reactive oxygen species (ROS). Notably, the AHF@AS Gel achieved ≈2.7-fold and ≈1.7-fold better re-epithelization in acute wounds and chronic diabetic wounds, respectively, significantly contributing to the promotion of wound closure. Using proteomic profiling and mechanistic studies, it is identified that the AHF@AS Gel efficiently promoted the transition of the inflammatory and proliferative phases to the remodeling phase. Notably, it is demonstrated that AS Gel alone activates the mechanosensitive epidermal growth factor receptor/Akt (EGFR/Akt) pathway and promotes cell proliferation. The antioxidative active shrinkage hydrogel offers a comprehensive strategy for acute wound and diabetic chronic wound closure via biochemistry regulation integrating with mechanical forces stimulation.


Assuntos
Antioxidantes , Hidrogéis , Pele , Cicatrização , Hidrogéis/química , Antioxidantes/química , Antioxidantes/farmacologia , Animais , Pele/metabolismo , Pele/efeitos dos fármacos , Pele/patologia , Camundongos , Cicatrização/efeitos dos fármacos , Fulerenos/química , Fulerenos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Receptores ErbB/metabolismo , Reepitelização/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Movimento Celular/efeitos dos fármacos , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Apoptose/efeitos dos fármacos
3.
Adv Sci (Weinh) ; 10(35): e2302910, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37884486

RESUMO

Tumor immunotherapy offers a new paradigm to treat cancer; however, the existing regimens are accompanied by the dilemma of insufficient therapeutic outcomes and off-target adverse effects. The intestinal immune system contains a bulk of immune cells, which can be important contributors to the maintenance of systemic immune homeostasis. However, manipulating intestinal immunity to achieve systemic anti-tumor immunity is extremely challenging. Here, an oral immunotherapy strategy is reported using immune-enhancing fullerenes (IEF) that can reinvigorate anti-tumor immunity via immune cell-metabolic reprogramming of intestinal immune cells. Findings show that IEF can remodel anti-inflammatory macrophages into tumor-killing macrophages by regulating the energy metabolism pathway from oxidative phosphorylation (OXPHOS) to glycolysis. Consequently, IEF can reprogram the immunosuppressive intestinal immunity and enhance sys temic immunity in vivo, thereby boosting anti-tumor immunity and converting "cold" tumors into "hot" tumors. Oral immunotherapy strategy, modulating autoimmune cells in the intestine and achieving systemic anti-tumor immunity, can ensure safe and efficient tumor immunotherapy.


Assuntos
Neoplasias , Humanos , Imunoterapia , Terapia de Imunossupressão , Neoplasias/tratamento farmacológico , Intestinos
4.
J Mater Chem B ; 11(31): 7401-7409, 2023 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-37431674

RESUMO

Myelosuppression is a predominant side-effect of radiotherapy, which manifests as the lower activity of blood cell precursors in bone marrow. Though progress in anti-myelosuppression has been made by the application of growth factors e.g., the granulocyte colony-stimulating factor (G-CSF), the side-effects (e.g., bone-pain, liver injury, and lung toxicity) limit their applications in clinic. Herein, we developed a strategy of efficiently normalizing leukopoiesis using gadofullerene nanoparticles (GFNPs) against myelosuppression triggered by radiation. Specifically, GFNPs with high radical-scavenging abilities elevated the generation of leukocytes and alleviated the bone marrow's pathological state under myelosuppression. Notably, GFNPs potentiated the differentiation, development, and maturation of leukocytes (neutrophils, lymphocytes) in radiation bearing mice even better than what G-CSF did. In addition, GFNPs had little toxicity towards the main organs including the heart, liver, spleen, lung, and kidney. This work provides an in-depth understanding of how advanced nanomaterials mitigate myelosuppression by regulating leukopoiesis.


Assuntos
Medula Óssea , Fulerenos , Camundongos , Animais , Medula Óssea/patologia , Leucopoese , Fator Estimulador de Colônias de Granulócitos/farmacologia , Fulerenos/farmacologia
5.
Sci Bull (Beijing) ; 68(15): 1651-1661, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37453828

RESUMO

Tumor vascular normalization (TVN) reverses abnormal tumor vasculatures, which could boost anti-cancer efficiency and especially increase drug intratumoral delivery. Endothelial cells play a vital role in angiogenesis, yet continuous modulating endothelial cell migration to improve TVN is ingenious but challenging. Here we propose a potential strategy for TVN based on inhibiting endothelial migration using antioxidative fullerene nanoparticles (FNPs). We demonstrate that FNPs inhibit cell migration upon their anti-oxidation effects in vitro. The optimized alanine-modified gadofullerene (GFA) exhibits superior TVN ability and inhibits tumor growth in vivo. Mechanically, facilitated with the protein microarray, we confirm that GFA could suppress the focal adhesion pathway to restrain endothelial migration. Subsequently, remarkable anti-tumor efficacy of chemotherapy synergy was obtained, which benefited from a more normalized vascular network by GFA. Together, our study introduces the potential of FNPs as promising TVN boosters to consider in cancer nanomedicine design.


Assuntos
Células Endoteliais , Neoplasias de Tecido Vascular , Humanos , Linhagem Celular Tumoral , Neoplasias de Tecido Vascular/metabolismo , Oxirredução
6.
Adv Healthc Mater ; 12(28): e2301306, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37506058

RESUMO

Metal ion-facilitated chemodynamic therapy (CDT) is an emerging method for treating cancer. However, its potential is hindered by its low catalytic performance in weakly acidic tumor microenvironments (TMEs) and the severe toxicity of free metal ions. A new approach to tumor therapy, chemodynamic vascular disruption (CVD), is introduced using metal-free, peroxidase (POD)-mimetic multihydroxylated [70] fullerene (MHF) nanocatalysts. The research shows that MHF contains C···O active sites, as demonstrated by density functional theory (DFT) calculations, and converts H2 O2 into ∙OH across a pH range of 6.0-10.0. The generation of ∙OH and the dismantling of tumor blood vessels are observed in real-time using mouse dorsal skin-fold chamber (DSFC) models. Applying proteomics, it is discovered that the CVD mechanism involves the nanocatalytic MHF enhancing H2 O2 decomposition in the TME, producing ∙OH. This damages tumor vascular endothelial junction proteins, causing vascular leakage and subsequently cutting off the vascular supply to the tumor cells. This method deviates from the traditional CDT that targets tumor cells. Instead, the proficient MHF nanocatalysts aim to directly disrupt the tumor vasculature, enhancing anti-tumor efficiency without triggering harmful toxicity. The proposed CVD therapeutic strategy enhances the application of gentle carbon nanocatalysts in cancer therapy, offering new perspectives on nanocatalytic medicine.


Assuntos
Doenças Cardiovasculares , Neoplasias , Animais , Camundongos , Peroxidase , Peroxidases , Endotélio Vascular , Neoplasias/tratamento farmacológico , Modelos Animais de Doenças , Linhagem Celular Tumoral , Peróxido de Hidrogênio , Microambiente Tumoral
7.
J Mater Chem B ; 10(45): 9457-9465, 2022 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-36346268

RESUMO

The development and progression of colorectal cancer (CRC) are highly dependent on the long-term inflammatory microenvironment with immune dysregulation in the colorectum. However, effective therapeutics are limited to targeting CRC. Here, we developed oral fullerene tablets (OFTs) that can act directly on the colorectal site by oral administration and reduce the inflammatory state at the tumor site for effective CRC therapy. In detail, OFTs scavenged reactive oxygen species (ROS), restrained the mutation of the wild-type P53, inhibited the activation of the inflammatory pathway nuclear factor-κB (NF-κB) and the signal transducer and activator of transcription 3 (STAT3) in the colorectum of CRC mice. Subsequently, OFTs could greatly reduce the infiltration of pro-inflammatory M1 macrophages and neutrophils at the tumor site, restoring the inflammatory microenvironment and immune homeostasis in the colorectal region, and ultimately achieving the inhibition of CRC. In addition, there were no significant toxic side effects of the long-term administration of OFTs. Our work provides an effective oral therapeutic strategy for CRC therapy by modulating the colorectal tumor inflammatory microenvironment and sheds light on the route for oral nano-materials in the clinical treatment of CRC.


Assuntos
Neoplasias Colorretais , Fulerenos , Camundongos , Animais , Fulerenos/farmacologia , Fulerenos/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Transdução de Sinais , NF-kappa B/metabolismo , Comprimidos , Microambiente Tumoral
8.
Adv Sci (Weinh) ; 9(29): e2201541, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36031401

RESUMO

Malignant proliferation and metastasis are the hallmarks of cancer cells. Aminated [70]fullerene exhibits notable antineoplastic effects, promoting it a candidate for multi-targeted cancer drugs. It is an urgent need to reveal the structure-activity relationship for antineoplastic aminated fullerenes. Herein, three amphiphilic derivatives of [60]fullerene with clarified molecular structures are synthesized: TAPC-4, TAPC-3, and TCPC-4. TAPC-4 inhibits the proliferation of diverse tumor cells via G0/G1 cell cycle arrest, reverses the epithelial-mesenchymal transition, and abrogates the high mobility of tumor cells. TAPC-4 can be excreted from the organism and achieves an in vivo inhibition index of 75.5% in tumor proliferation and 87.5% in metastatic melanoma with a wide safety margin. Molecular dynamics simulations reveal that the amphiphilic molecular structure and the ending amino groups promote the targeting of TAPC-4 to heat shock protein Hsp90-beta, vimentin, and myosin heavy chain 9 (MYH9), probably resulting in the alteration of cyclin D1 translation, vimentin expression, and MYH9 location, respectively. This work initially emphasizes the dominant role of the amphiphilic structure and the terminal amino moieties in the antineoplastic effects of aminated fullerenes, providing fundamental support for their anti-tumor drug development.


Assuntos
Antineoplásicos , Fulerenos , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Ciclina D1 , Fulerenos/química , Fulerenos/farmacologia , Fulerenos/uso terapêutico , Proteínas de Choque Térmico , Cadeias Pesadas de Miosina , Vimentina
9.
J Mater Chem B ; 9(28): 5722-5728, 2021 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-34231648

RESUMO

Hepatocellular carcinoma has become one of the most prevalent cancers, with a high mortality rate. Accurate diagnosis at an earlier stage is regarded as an effective measure to reduce the disease-related mortality of liver cancer. Magnetic resonance imaging (MRI) as a non-invasive checking mode has become a powerful tool in medical diagnosis. However, MRI contrast agents for liver-specific imaging either have some side effects or the imaging effect is not ideal. Thus, development of more efficient and security MRI contrast agents for the early diagnosis of hepatocellular carcinoma is urgent. Herein, a kind of water-soluble gadofullerene nanoparticle (GFNP) with high efficiency and security has been successfully used to achieve in situ liver cancer imaging. By comparing GFNPs with different functional groups, Gd@C82 modified by a hydroxyl group (GF-OH) presents the highest contrast efficiency both in vitro and in vivo. Notably, the smallest tumor with a diameter of only 0.5 mm could be clearly observed by GF-OH using MRI. Moreover, the imaging window of GF-OH is more than 3-6 hours. In addition, GF-OH can be mostly excreted from the living body and causes no serious toxicity. These results demonstrate that GF-OH is a safe, efficient MRI contrast agent for the diagnosis of early orthotopic hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular/diagnóstico por imagem , Meios de Contraste/química , Fulerenos/química , Neoplasias Hepáticas/diagnóstico por imagem , Imageamento por Ressonância Magnética , Animais , Meios de Contraste/farmacocinética , Feminino , Fulerenos/farmacocinética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Tamanho da Partícula , Distribuição Tecidual
10.
Langmuir ; 37(8): 2740-2748, 2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33586439

RESUMO

Hydrophilic fullerene derivatives get notable performance in various biological applications, especially in cancer therapy and antioxidation. The biological behaviors of functional fullerenes are much dependent on their surface physicochemical properties. The excellent reactive oxygen species-scavenging capabilities of functional fullerenes promote their outstanding performances in inhibiting pathological symptoms associated with oxidative stress, including neurodegenerative diseases, cardiovascular diseases, acute and chronic kidney disease, and diabetes. Herein, fullerene derivatives with reversed surface charges in aqueous solutions are prepared: cationic C60-EDA and anionic C60-(EDA-EA). Under the driving force of membrane potential (negative inside) in the cell and mitochondria, C60-EDA is much rapidly taken in by cells and transported into mitochondria compared with C60-(EDA-EA) that is enriched in lysosomes. With high cellular uptake and mitochondrial enrichment, C60-EDA exhibits stronger antioxidation capabilities in vitro than C60-(EDA-EA), indicating its better performance in the therapy of oxidation-induced diseases. It is revealed that the cellular uptake rate, subcellular location, and intracellular antioxidation behavior of fullerene derivatives are primarily mediated by their surface charges, providing new strategies for the design of fullerene drugs and their biological applications.


Assuntos
Fulerenos , Antioxidantes/farmacologia , Organelas , Estresse Oxidativo , Espécies Reativas de Oxigênio
11.
ACS Appl Mater Interfaces ; 12(51): 56862-56873, 2020 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-33305958

RESUMO

Functional fullerene derivatives exhibit fantastic inhibitory capabilities against cancer survival and metastasis, but the absence of clarified biological molecular targets and ambiguous regulation mechanisms set barriers for their clinical transformation. Cancer metastasis is the primary cause of mortality and initiated with increased cell migration, making cell motility regulation a high-value therapeutic target in precision medicine. Herein, a critical molecular target of the aminated fullerene derivative (C70-EDA), myosin heavy chain 9 (MYH9), was initially identified by a pull-down assay and MS screening. MYH9 is a cytoplasm-located protein and is responsible for cell motility and epithelial-mesenchymal transition regulation. Omics data from large-scale clinical samples reveals that MYH9 gets overexpressed in various cancers and correlates with unfavorable prognosis, indicating that it is a potential antineoplastic target. It is unveiled that C70-EDA binds to the C-terminal of MYH9, triggering the transport of MYH9 from the cytoplasm to the cell edge, blocking the MYH9-involved cell mobility, and inhibiting the metastasis-associated EMT process. This work provides a precise biological target and new strategies for fullerene applications in cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Movimento Celular/efeitos dos fármacos , Fulerenos/farmacologia , Cadeias Pesadas de Miosina/metabolismo , Células A549 , Antineoplásicos/química , Antineoplásicos/metabolismo , Biotina/análogos & derivados , Biotina/metabolismo , Biotina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Fluoresceína-5-Isotiocianato/farmacologia , Fulerenos/química , Fulerenos/metabolismo , Humanos , Ligação Proteica
12.
Theranostics ; 10(15): 6886-6897, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32550910

RESUMO

Aplastic anemia (AA) is characterized as hypoplasia of bone marrow hematopoietic cells and hematopenia of peripheral blood cells. Though the supplement of exogenous erythropoietin (EPO) has been clinically approved for AA treatment, the side-effects hinder its further application. Here a robust treatment for AA induced by chemotherapy drugs is explored using gadofullerene nanoparticles (GFNPs). Methods: The gadofullerene were modified with hydrogen peroxide under alkaline conditions to become the water-soluble nanoparticles (GFNPs). The physicochemical properties, in vitro chemical construction, stability, hydroxyl radical scavenging ability, in vitro cytotoxicity, antioxidant activity, in vivo treatment efficacy, therapeutic mechanism and biological distribution, metabolism, toxicity of GFNPs were examined. Results: GFNPs with great stability and high-efficiency antioxidant activity could observably increase the number of red blood cells (RBC) in the peripheral blood of AA mice and relieve the abnormal pathological state of bone marrow. The erythropoiesis mainly includes hemopoietic stem cells (HSCs) differentiation, erythrocyte development in bone marrow and erythrocyte maturation in peripheral blood. The positive control-EPO promotes erythropoiesis by regulating HSCs differentiation and erythrocyte development in bone marrow. Different from the anti-AA mechanism of EPO, GFNPs have little impact on both the differentiation of HSCs and the myeloid erythrocyte development, but notably improve the erythrocyte maturation. Besides, GFNPs can notably decrease the excessive reactive oxygen species (ROS) and inhibit apoptosis of hemocytes in blood. In addition, GFNPs are mostly excreted from the living body and cause no serious toxicity. Conclusion: Our work provides an insight into the advanced nanoparticles to powerfully treat AA through ameliorating the erythrocyte maturation during erythropoiesis.


Assuntos
Anemia Aplástica/tratamento farmacológico , Células da Medula Óssea/efeitos dos fármacos , Ciclofosfamida/toxicidade , Eritropoetina/farmacologia , Fulerenos/química , Células-Tronco Hematopoéticas/efeitos dos fármacos , Nanopartículas/administração & dosagem , Anemia Aplástica/induzido quimicamente , Anemia Aplástica/patologia , Animais , Antineoplásicos Alquilantes/toxicidade , Células da Medula Óssea/metabolismo , Bussulfano/toxicidade , Diferenciação Celular , Modelos Animais de Doenças , Eritropoese/efeitos dos fármacos , Feminino , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Nanopartículas/química
13.
Nano Lett ; 20(6): 4487-4496, 2020 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-32407113

RESUMO

Cancer immunotherapy as a novel cancer therapeutic strategy has shown enormous promise. However, the immunosuppressive tumor microenvironment (ITM) is a primary obstacle. Tumor-associated macrophages (TAMs) as a major component of immune cells in a tumor microenvironment are generally polarized to the M2 phenotype that not only accelerates tumor growth but also influences the infiltration of lymphocytes and leads to immunosuppression. Thus, rebuilding ITM by re-educating TAMs and increasing infiltration of lymphocytes is a promising strategy. Herein, gadofullerene (GF-Ala) nanoparticles are demonstrated to reprogram TAMs to M1-like and increase the infiltration of cytotoxic T lymphocytes (CTLs), achieving effective inhibition of tumor growth. Notably, the modulation of ITM by GF-Ala promotes the anticancer efficacy of anti-PD-L1 immune checkpoint inhibitor, achieving superior synergistic treatment. Additionally, GF-Ala nanoparticles can be mostly excreted from the body and cause no obvious toxicity. Together, this study provides an effective immunomodulation strategy using gadofullerene nanoparticles by rebuilding ITM and synergizing immune checkpoint blockade therapy.


Assuntos
Fulerenos , Nanopartículas , Neoplasias , Microambiente Tumoral , Humanos , Imunoterapia , Neoplasias/tratamento farmacológico
14.
J Mater Chem B ; 8(12): 2508-2518, 2020 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-32124888

RESUMO

The traditional photodynamic therapy (PDT) using a photosensitizer and oxygen under light generates reactive oxygen species (ROS) to kill tumor cells. However, its treatment efficiency is limited by insufficient oxygen in tumor cells. Herein, ß-alanine modified gadofullerene nanoparticles (GFNPs) were explored to disrupt tumor vasculatures assisted by light for potent melanoma treatment. As tumor vasculatures are oxygen-rich, the yields of photo-induced singlet oxygen (1O2) by GFNPs are not subjected to the hypoxemia of tumor tissues. Different from the small molecule photosensitizer Chlorin e6 (Ce6), GFNPs realize high-efficiency tumor vascular disruption under light observed by using the mice tumor vascular dorsal skin fold chamber (DSFC) model. The tumor vascular disruption efficiency of GFNPs is size-dependent, and the smallest one (hydration diameter of ca. 126 nm) is more efficient. Mechanistically, the high yields of photo-induced 1O2 by GFNPs can lead to the destruction of the tumor vascular endothelial adherent junction protein-VE cadherin and the decrease of tumor vascular endothelial cells-CD31 proteins, inducing rapid tumor necrosis. In conclusion, our work provides an insight into the design of well-sized nanoparticles to powerfully treat melanoma assisted by light, as well as greatly extending the applications of PDT for robust tumor therapy.


Assuntos
Fulerenos/farmacologia , Luz , Melanoma/tratamento farmacológico , Nanopartículas/química , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Animais , Morte Celular/efeitos dos fármacos , Clorofilídeos , Feminino , Fulerenos/química , Melanoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Tamanho da Partícula , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/química , Porfirinas/química , Porfirinas/farmacologia , Oxigênio Singlete/análise , Neoplasias Cutâneas/patologia , Propriedades de Superfície , beta-Alanina/química
15.
ACS Appl Bio Mater ; 3(1): 450-457, 2020 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-35019461

RESUMO

Radiotherapy is the current frontline method for cancer treatment, while the severe systemic side effects (e.g., myelosuppression) limit its application because it generates excessive reactive oxygen species. Therefore, there is a pressing need to develop effective strategies for radiotherapy protection. Here, we explored a robust myelosuppressive protector using gadofullerene nanocrystals (GFNCs) to protect mice against radiation injury, which was induced by different doses of X-rays (3, 4.5, and 6 Gy). Our data show that the radiotherapy-induced myelosuppression was remarkably reduced by the high radical scavenging abilities of GFNCs. In addition, GFNCs could normalize the oxidative stress-related indexes, such as malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT). Of note, GFNCs provided protection of the bone marrow in tumor-bearing mice without interfering with the antitumor properties of radiotherapy. Thus, GFNCs may play a promising role in radioprotection during radiotherapy.

16.
Dalton Trans ; 48(22): 7884-7890, 2019 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-31080968

RESUMO

Metallofullerene Gd@C82 derivatives have been found to exhibit excellent performance in the treatment of chronic diseases and in tumor therapy due to their excellent anti-oxidation ability. Thus, there is a great need to clarify the specific protection mechanism of Gd@C82 derivatives against oxidative stress at the cellular and molecular level. Herein, we optimize the preparation of amino acid derivatives of Gd@C82 (GF-Ala) and investigated the potential mechanism of anti-oxidative stress induced by H2O2 in L02 human hepatic cells. GF-Ala with excellent biocompatibility and high production yield was obtained by adjusting the size of solid Gd@C82 as a starting material. Pretreatment with GF-Ala significantly improved the cell viability of oxidatively damaged cells. Furthermore, we found that GF-Ala prominently reduced intracellular reactive oxygen species (ROS) production, stabilized mitochondrial membrane potential (MMP) and inhibited cell apoptosis. Moreover, GF-Ala up-regulated the expression of Bcl-2 and down-regulated the expression of Bax, which further prevented the activation of pro-caspase 3 and PARP. Thus, we showed that GF-Ala protected the cells from oxidative stress by blocking the mitochondria-mediated apoptosis pathway.


Assuntos
Alanina/química , Apoptose/efeitos dos fármacos , Complexos de Coordenação/farmacologia , Fulerenos/química , Gadolínio/química , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Linhagem Celular , Complexos de Coordenação/química , Humanos , Peróxido de Hidrogênio/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos
17.
ACS Appl Mater Interfaces ; 11(24): 21343-21352, 2019 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-31140277

RESUMO

Water-soluble gadofullerene nanomaterials have been extensively investigated as magnetic resonance imaging (MRI) contrast agents, radical scavengers, sensitizers for photodynamic therapy, and inherent antineoplastic agents. Most recently, an alanine-modified gadofullerene nanoparticle (Gd@C82-Ala) with excellent anticancer activity has been reported; however, the absolute tumor uptake of Gd@C82-Ala is still far from being satisfactory, and its dynamic pharmacokinetics and long-term metabolic behaviors remain to be elucidated. Herein, Gd@C82-Ala was chemically modified with eight-arm polyethylene glycol amine to improve its biocompatibility and provide the active sites for the attachment of a tumor-homing ligand (cRGD) and positron emission tomography (PET) isotopes (i.e., 64Cu or 89Zr). The physical and chemical properties (e.g., size, surface functionalization condition, radiochemical stability, etc.) of functionalized Gd@C82-Ala were properly characterized. Also, its glioblastoma cell targeting capacity was evaluated in vitro by flow cytometry, confocal fluorescence microscopy, and dynamic cellular interaction assays. Because of the presence of gadolinium ions, the gadofullerene conjugates can act simultaneously as T1* MRI contrast agents and PET probes. Thus, the pharmacokinetic behavior of functionalized Gd@C82-Ala was investigated by PET/MRI, which combines the merits of high resolution and excellent sensitivity. The functionalized Gd@C82-Ala-PEG-cRGD-NOTA-64Cu (NOTA stands for 1,4,7-triazacyclononane-triacetic acid) demonstrated much higher accumulation in U87-MG tumor than its counterpart without cRGD attachment from in vivo PET observation, consistent with observation at the cellular level. In addition, Gd@C82-Ala-PEG-Df-89Zr (Df stands for desferrioxamine) was employed to investigate the metabolic behavior of gadofullerene conjugates in vivo for up to 30 days. It was estimated that nearly 70% of Gd@C82-Ala-PEG-Df-89Zr was excreted from the test subjects primarily through renal pathways within 24 h. With proper surface engineering, functionalized Gd@C82-Ala nanoparticles can show an improved accumulation in glioblastoma. Pharmacokinetic studies also confirmed the safety of this nanoplatform, which can be used as an image-guidable therapeutic agent for glioblastoma.


Assuntos
Glioblastoma/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Nanopartículas/química , Tomografia por Emissão de Pósitrons/métodos , Animais , Citometria de Fluxo , Fulerenos/química , Gadolínio/química , Humanos , Células MCF-7 , Camundongos , Microscopia Confocal
18.
Sci Rep ; 8(1): 16573, 2018 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-30410075

RESUMO

Despite the great efforts for tumor therapy in the last decades, currently chemotherapy induced toxicity remains a formidable problem for cancer patients, and it usually prohibits the cancer therapy from successful completion due to severe side effects. In general, the main side effects of chemotherapeutic agents are from the as-produced reactive oxygen species (ROS) that not only harm the tumor cells but also damage the patients' organs. Here we report the application of amino acid derivatives of fullerene (AADF) in the chemotherapy which strongly scavenge the excess ROS to protect the tested mice against the chemotherapy-induced hepatotoxicity and cardiotoxicity. Two amino acids, i.e., L-lysine and ß-alanine were separately employed to chemically modify C70 fullerene, and L-lysine derivative of fullerene (C70-Lys) exhibits superior radical scavenging activity to ß-alanine derivative of C70 (C70-Ala). As expected, C70-Lys show much better protective effect than C70-Ala against the chemotherapy injuries in vivo, which is verified by various histopathological, haematological examinations and antioxidative enzyme studies. Moreover, the L-glutathione level is increased and the cytochrome P-450 2E1 expression is inhibited. They are potentially developed as promising bodyguards for chemotherapy protection.


Assuntos
Aminoácidos/química , Antineoplásicos/efeitos adversos , Cardiotoxicidade/prevenção & controle , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Sequestradores de Radicais Livres/administração & dosagem , Fulerenos/administração & dosagem , Animais , Citocromo P-450 CYP2E1/metabolismo , Doxorrubicina/efeitos adversos , Feminino , Sequestradores de Radicais Livres/química , Sequestradores de Radicais Livres/farmacologia , Fulerenos/química , Fulerenos/farmacologia , Glutationa/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Lisina/química , Camundongos , Espécies Reativas de Oxigênio/metabolismo , beta-Alanina/química
19.
Biomaterials ; 163: 142-153, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29459323

RESUMO

The tumor vasculature with unique characteristics offers an attractive target for anti-cancer therapy. Herein, we put forward a novel antitumor therapeutic mechanism based on the gadofullerene nanocrystals (GFNCs), the agent we have previously shown to efficiently disrupt tumor vasculature by size-expansion with assistance of radiofrequency (RF). However, the tumor vascular disrupting mechanism of RF-assisted GFNCs treatment was not further studied. In the present work, a rapid tumor blood flow shutdown has been observed by the vascular perfusion imaging in vivo and vascular damages were evident 6 h after the RF-assisted GFNCs treatment. Importantly, a significant down-expression of tumor vascular endothelial cadherin (VE-cadherin) treated by RF-assisted GFNCs was further investigated, which caused vascular collapse, blood flow shut-down and subsequent tumor hemorrhagic necrosis. These findings set forth a systematic mechanism on the superior anti-tumor efficiency by RF-assisted GFNCs treatment.


Assuntos
Antineoplásicos/administração & dosagem , Vasos Sanguíneos/metabolismo , Fulerenos/administração & dosagem , Gadolínio/química , Nanopartículas/química , Animais , Antígenos CD/metabolismo , Antineoplásicos/química , Caderinas/metabolismo , Carcinoma Hepatocelular/irrigação sanguínea , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/terapia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Fulerenos/química , Células Hep G2 , Xenoenxertos , Células Endoteliais da Veia Umbilical Humana , Humanos , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/uso terapêutico , Tamanho da Partícula , Ondas de Rádio
20.
ACS Appl Mater Interfaces ; 9(41): 35539-35547, 2017 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-28945341

RESUMO

Chemotherapy as a conventional cancer treatment suffers from critical systemic side effects, which is generally considered as the consequence of reactive oxygen species (ROS). Fullerenes have been widely studied for their excellent performance in radicals scavenging. In the present study, we report a solid-liquid reaction to synthesize fullerenols and their application as ROS scavengers in chemotherapy protection. The solid-liquid reaction is carried out without catalyst and suitable for mass production. The novel [60]/[70] fullerenols show a high stability in water, and the [70] fullerenols (C70-OH) exhibit radical scavenging capability superior to that of [60] fullerenols (C60-OH) in chemotherapy protection. The mouse model for single and reduplicative chemotherapy-induced liver injury demonstrates their protective effects in the chemotherapeutic process, which is confirmed by histopathological examinations and hematological index. The increase of the hepatic l-glutathione (GSH) level and downregulated expression of the cytochrome P-450 2E1 (CYP2E1) give the possible mechanism associated with the impact of fullerenols on the metabolism of doxorubicin. The novel fullerenols may be promising protective agents to satisfy the demand for future clinical chemotherapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA