Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Adv Mater ; : e2410031, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39246208

RESUMEN

During fractionated radiotherapy, DNA damage repair intensifies in tumor cells, culminating in cancer radioresistance and subsequent radiotherapy failure. Despite the recent development of nanoradiosensitizers targeting specific DNA damage repair pathways, the persistence of repair mechanisms involving multiple pathways remains inevitable. To address this challenge, a nucleophilicity-engineered DNA ligation blockade nanoradiosensitizer (DLBN) comprising Au/CeO2 heteronanostructure modified with trans-acting activator of transcription peptides is reported, which targets and inhibits the DNA ligation inside cancer cell nuclei via heterointerface-mediated dephosphorylation of DNA, a crucial step in overcoming cancer radioresistance. First, the Schottky-type heteronanostructure of cancer cell nucleus-targeting DLBN effectively intensifies radiation-induced DNA damage via catalase-mimetic activity and radiation-triggered catalytic reactions. Notably, by leveraging Au/CeO2 heterointerface, DLBN spontaneously dissociates H2O to hydroxide, a nucleophile with higher nucleophilicity, thereby exhibiting remarkable dephosphorylation capability at DNA nicks through facilitated nucleophilic attack. This enables the blockade of DNA ligation, a pivotal step in all DNA damage repair pathways, effectively interrupting the repair process. Consequently, DLBN resensitizes radioresistant cells by overcoming therapy-induced radioresistance, leading to a substantial accumulation of unrepaired DNA damage. These findings offer insight into the dephosphorylation of DNA within nuclei, and underscore the potential of heteronanostructure-based nanoradiosensitizer to block DNA ligation against therapy-induced radioresistance.

2.
Adv Sci (Weinh) ; 11(28): e2403116, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38816935

RESUMEN

To overcome current limitations in photoimmunotherapy, such as insufficient tumor antigen generation and a subdued immune response, a novel photo-/metallo dual-mode immunotherapeutic agent (PMIA) is introduced for potent near-infrared (NIR) light-triggered cancer therapy. PMIA features a dumbbell-like AuPt heterostructure decorated with starry Pt nanoclusters, meticulously engineered for enhancing plasmonic catalysis through multi-dimensional regulation of Pt growth on Au nanorods. Under NIR laser exposure, end-tipped Pt nanoclusters induce efficient electron-hole spatial separation along the longitudinal axis, resulting in radial and axial electron distribution polarization, conferring unique anisotropic properties to PMIA. Additionally, starry Pt nanoclusters on the sides of Au nanorods augment the local electron enrichment field. Validated through finite-difference time-domain analysis and Raman scattering, this configuration fosters local electron enrichment, facilitating robust reactive oxygen species generation for potent photoimmunotherapy. Moreover, Pt nanoclusters facilitate Pt2+ ion release, instigating intranuclear DNA damage and inducing synergistic immunogenic cell death (ICD) for metalloimmunotherapy. Consequently, PMIA elicits abundant danger-associated molecular patterns, promotes T cell infiltration, and triggers systemic immune responses, effectively treating primary and distant tumors, inhibiting metastasis in vivo. This study unveils a pioneering dual-mode ICD amplification strategy driven by NIR light, synergistically integrating photoimmunotherapy and metalloimmunotherapy, culminating in potent cancer photometalloimmunotherapy.


Asunto(s)
Oro , Inmunoterapia , Nanopartículas del Metal , Platino (Metal) , Inmunoterapia/métodos , Ratones , Animales , Platino (Metal)/química , Platino (Metal)/uso terapéutico , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Oro/química , Fototerapia/métodos , Neoplasias/terapia , Neoplasias/inmunología , Modelos Animales de Enfermedad , Anisotropía , Catálisis , Humanos , Línea Celular Tumoral
3.
Nat Commun ; 15(1): 460, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38212655

RESUMEN

Targeted assembly of nanoparticles in biological systems holds great promise for disease-specific imaging and therapy. However, the current manipulation of nanoparticle dynamics is primarily limited to organic pericyclic reactions, which necessitate the introduction of synthetic functional groups as bioorthogonal handles on the nanoparticles, leading to complex and laborious design processes. Here, we report the synthesis of tyrosine (Tyr)-modified peptides-capped iodine (I) doped CuS nanoparticles (CuS-I@P1 NPs) as self-catalytic building blocks that undergo self-propelled assembly inside tumour cells via Tyr-Tyr condensation reactions catalyzed by the nanoparticles themselves. Upon cellular internalization, the CuS-I@P1 NPs undergo furin-guided condensation reactions, leading to the formation of CuS-I nanoparticle assemblies through dityrosine bond. The tumour-specific furin-instructed intracellular assembly of CuS-I NPs exhibits activatable dual-modal imaging capability and enhanced photothermal effect, enabling highly efficient imaging and therapy of tumours. The robust nanoparticle self-catalysis-regulated in situ assembly, facilitated by natural handles, offers the advantages of convenient fabrication, high reaction specificity, and biocompatibility, representing a generalizable strategy for target-specific activatable biomedical imaging and therapy.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Furina , Fototerapia , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Nanopartículas/química , Catálisis , Cobre/química
4.
ACS Nano ; 17(18): 18548-18561, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37706454

RESUMEN

Replication stress (RS) induced by DNA damage plays a significant role in conferring the anticancer effects of radiotherapy and is tightly associated with radioresistance of cancer cells. Amplification of RS represents an effective approach to improving the efficacy of radiotherapy, although the development of selective RS amplifiers remains an unexplored frontier. We herein present an RS nano amplifier (RSNA) consisting of a catalytic FePt nanoparticle loaded with the chemotherapeutic doxorubicin (DOX), which selectively exacerbates RS in cancer cells by promoting replication fork (RF) catastrophe. RSNA converts the excessive reactive oxygen species (ROS) in cancer cells into oxygen, enhancing the DNA-damaging effects of radiotherapy to create more template lesions that impede RF progression in coalition with DOX. After radiation, ROS scavenging by RSNA accelerates RF progression through damaged template strands, increasing the frequency of RF collapse into double-strand breaks. Moreover, pretreatment with RSNA accumulates cancer cells in the S phase, exposing more RFs to radiation-induced RS. These effects of RSNA convergently maximize RS in cancer cells, effectively overcoming the radioresistance of cancer cells without affecting normal cells. Our study demonstrates the feasibility of selectively amplifying RS to boost radiotherapy.


Asunto(s)
Neoplasias , Humanos , Especies Reactivas de Oxígeno , División Celular , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Catálisis , Daño del ADN , Doxorrubicina/farmacología
5.
ACS Appl Mater Interfaces ; 15(31): 37193-37204, 2023 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-37493513

RESUMEN

Mesenchymal stem cell (MSC)-based therapy has emerged as a promising strategy for the treatment of spinal cord injury (SCI). However, the hostile microenvironment of SCI, which can adversely affect the survival and paracrine effect of the implanted MSCs, severely limits the therapeutic efficacy of this approach. Here, we report on a ceria nanozyme-integrated thermoresponsive in situ forming hydrogel (CeNZ-gel) that can enable dual enhancement of MSC viability and paracrine effect, leading to highly efficient spinal cord repair. The sol-gel transition property of the CeNZ-gel at body temperature ensures uniform coverage of the hydrogel in injured spinal cord tissues. Our results demonstrate that the CeNZ-gel significantly increases the viability of transplanted MSCs in the microenvironment by attenuating oxidative stress and, more importantly, promotes the secretion of angiogenic factors from MSCs by inducing autophagy of MSCs. The synergy between the oxidative stress-relieving effect of CeNZs and the paracrine effect of MSCs accelerates angiogenesis, nerve repair, and motor function recovery after SCI, providing an efficient strategy for MSC-based SCI therapy.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Traumatismos de la Médula Espinal , Regeneración de la Medula Espinal , Humanos , Hidrogeles/farmacología , Trasplante de Células Madre Mesenquimatosas/métodos , Traumatismos de la Médula Espinal/terapia
6.
Health Informatics J ; 28(4): 14604582221140975, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36413427

RESUMEN

A history of brain tumor strongly affects children's cognitive abilities, performance of daily activities, quality of life, and functional outcomes. In light of the difficulties in cognition, communication, physical skills, and behavior that these patients may encounter, occupational therapists should perform a comprehensive needs-led assessment of their global functioning after recovery. Such an assessment would ensure that the patients receive adequate support and services at school, at home, and in the community. By predicting the functional activity performance of children with a history of brain tumor, clinical workers can determine the progress of their ability recovery and the optimal treatment plan. We selected several features for testing and employed common machine learning models to predict Functional Independence Measure (WeeFIM) scores. The ensemble learning models exhibited stronger predictive performance than did the individual machine learning models. The ensemble learning models effectively predicted WeeFIM scores. Machine learning models can help clinical workers predict the functional assessment scores of patients with childhood brain tumors. This study used machine learning models to predict the WeeFIM scores of patients with childhood brain tumors and to demonstrate that ensemble machine learning models are more suitable for this task than are individual machine learning models.


Asunto(s)
Neoplasias Encefálicas , Estado Funcional , Niño , Humanos , Calidad de Vida , Aprendizaje Automático , Sobrevivientes , Neoplasias Encefálicas/terapia
7.
Natl Sci Rev ; 9(7): nwac080, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35832777

RESUMEN

Although molecular imaging probes have the potential to non-invasively diagnose a tumor, imaging probes that can detect a tumor and simultaneously identify tumor malignancy remain elusive. Here, we demonstrate a potassium ion (K+) sensitive dual-mode nanoprobe (KDMN) for non-invasive tumor imaging and malignancy identification, which operates via a cascaded 'AND' logic gate controlled by inputs of magnetic resonance imaging (MRI) and fluorescence imaging (FI) signals. We encapsulate commercial K+ indicators into the hollow cavities of magnetic mesoporous silica nanoparticles, which are subsequently coated with a K+-selective membrane that exclusively permits the passage of K+ while excluding other cations. The KDMN can readily accumulate in tumors and enhance the MRI contrast after systemic administration. Spatial information of the tumor lesion is thus accessible via MRI and forms the first layer of the 'AND' gate. Meanwhile, the KDMN selectively captures K+ and prevents interference from other cations, triggering a K+-activated FI signal as the second layer of the 'AND' gate in the case of a malignant tumor with a high extracellular K+ level. This dual-mode imaging approach effectively eliminates false positive or negative diagnostic results and allows for non-invasive imaging of tumor malignancy with high sensitivity and accuracy.

8.
PLoS One ; 16(12): e0257972, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34972111

RESUMEN

Cancer immunotherapies, such as checkpoint blockade of programmed cell death protein-1 (PD-1), represents a breakthrough in cancer treatment, resulting in unprecedented results in terms of overall and progression-free survival. Discovery and development of novel anti PD-1 inhibitors remains a field of intense investigation, where novel monoclonal antibodies (mAbs) and novel antibody formats (e.g., novel isotype, bispecific mAb and low-molecular-weight compounds) are major source of future therapeutic candidates. HLX10, a fully humanized IgG4 monoclonal antibody against PD-1 receptor, increased functional activities of human T-cells and showed in vitro, and anti-tumor activity in several tumor models. The combined inhibition of PD-1/PDL-1 and angiogenesis pathways using anti-VEGF antibody may enhance a sustained suppression of cancer-related angiogenesis and tumor elimination. To elucidate HLX10's mode of action, we solved the structure of HLX10 in complex with PD-1 receptor. Detailed epitope analysis showed that HLX10 has a unique mode of recognition compared to the clinically approved PD1 antibodies Pembrolizumab and Nivolumab. Notably, HLX10's epitope was closer to Pembrolizumab's epitope than Nivolumab's epitope. However, HLX10 and Pembrolizumab showed an opposite heavy chain (HC) and light chain (LC) usage, which recognizes several overlapping amino acid residues on PD-1. We compared HLX10 to Nivolumab and Pembrolizumab and it showed similar or better bioactivity in vitro and in vivo, providing a rationale for clinical evaluation in cancer immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/química , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/química , Receptor de Muerte Celular Programada 1/inmunología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/uso terapéutico , Bevacizumab/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Línea Celular Tumoral , Proliferación Celular , Transición Epitelial-Mesenquimal/efectos de los fármacos , Epítopos/inmunología , Humanos , Fragmentos Fab de Inmunoglobulinas/metabolismo , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Ligandos , Macaca fascicularis , Ratones Endogámicos NOD , Ratones SCID , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Nivolumab/química , Nivolumab/uso terapéutico , Unión Proteica , Ratas , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Expert Opin Biol Ther ; 21(11): 1491-1507, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34632911

RESUMEN

BACKGROUND: Cetuximab, the first approved EGFR targeting therapeutic antibody, is currently used to treat colorectal cancer and head and neck cancer. While effective, cetuximab is associated with a higher rate of skin rash, infusion reactions, and gastrointestinal toxicity, which was suggested to be linked to the presence of heterogeneous glycan contents on the Fab of the SP2/0-produced cetuximab. OBJECTIVE AND METHODS: To improve efficacy and minimize toxicity of EGFR inhibition treatment, we re-engineered cetuximab by humanizing its Fab regions and minimizing its glycan contents to generate HLX07. RESULTS: HLX07 binds to EGFR with similar affinity as cetuximab and shows better bioactivity compared to cetuximab in vitro. In vivo studies demonstrated that HLX07 significantly inhibited the growth of A431, FaDu, NCI-H292, and WiDr tumor cells and synergized them with chemotherapeutics and immune simulator agents such as anti-PD-1. In cynomolgus monkeys, 13-week repeat-dose GLP toxicokinetic studies showed minimal-to-mild toxicities in the dose range of up to 60 mg/kg/wk. In the preliminary phase 1 dose-escalation study, HLX07 had showed lower incidence of skin rashes with grade >2 severities. CONCLUSION: HLX07 is currently under phase 1/2 clinical development. We believe HLX07 would potentially be an alternative for patients who have been suffering from cetuximab-mediated toxicity.


Asunto(s)
Antineoplásicos , Neoplasias de Cabeza y Cuello , Anticuerpos Monoclonales , Antineoplásicos/efectos adversos , Línea Celular Tumoral , Cetuximab , Receptores ErbB , Humanos
10.
Acta Biomater ; 126: 15-30, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33652165

RESUMEN

With the advancement of nanochemistry, artificial nanozymes with high catalytic stability, low manufacturing and storage cost, and greater design flexibility over natural enzymes, have emerged as a next-generation nanomedicine. The catalytic activity and selectivity of nanozymes can be readily controlled and optimized by the rational chemical design of nanomaterials. This review summarizes the various chemical approaches to regulate the catalytic activity and selectivity of nanozymes for biomedical applications. We focus on the in-depth correlation between the physicochemical characteristics and catalytic activities of nanozymes from several aspects, including regulating chemical composition, controlling morphology, altering the size, surface modification and self-assembly. Furthermore, the chemically designed nanozymes for various biomedical applications such as biosensing, infectious disease therapy, cancer therapy, neurodegenerative disease therapy and injury therapy, are briefly summarized. Finally, the current challenges and future perspectives of nanozymes are discussed from a chemistry point of view. STATEMENT OF SIGNIFICANCE: As a kind of nanomaterials that performs enzyme-like properties, nanozymes perform high catalytic stability, low manufacturing and storage cost, attracting the attention of researchers from various fields. Notably, chemically designed nanozymes with robust catalytic activity, tunable specificity and multi-functionalities are promising for biomedical applications. It's crucial to define the correlation between the physicochemical characteristics and catalytic activities of nanozymes. To help readers understand this rapidly expanding field, in this review, we summarize various chemical approaches that regulate the catalytic activity and selectivity of nanozymes together with the discussion of related mechanisms, followed by the introduction of diverse biomedical applications using these chemically well-designed nanozymes. Hopefully our review will bridge the chemical design and biomedical applications of nanozymes, supporting the extensive research on high-performance nanozymes.


Asunto(s)
Nanoestructuras , Enfermedades Neurodegenerativas , Catálisis , Humanos , Nanomedicina
11.
Small ; 16(31): e2002537, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32519453

RESUMEN

Triple-negative breast cancer (TNBC) is highly aggressive and insensitive to conventional targeted therapies, resulting in poor therapeutic outcomes. Recent studies have shown that abnormal iron metabolism is observed in TNBC, suggesting an opportunity for TNBC treatment via the iron-dependent Fenton reaction. Nevertheless, the efficiency of current Fenton reagents is largely restricted by the lack of specificity and low intracellular H2 O2 level of cancer cells. Herein, core-shell-satellite nanomaces (Au @ MSN@IONP) are fabricated, as near-infrared (NIR) light-triggered self-fueling Fenton reagents for the amplified Fenton reaction inside TNBC cells. Specifically, the Au nanorod core can convert NIR light energy into heat to induce massive production of intracellular H2 O2 , thereby the surface-decorated iron oxide nanoparticles (IONP) are being fueled for robust Fenton reaction. By exploiting the vulnerability of iron efflux in TNBC cells, such a self-fueling Fenton reaction leads to highly specific anti-TNBC efficacy with minimal cytotoxicity to normal cells. The PI3K/Akt/FoxO axis, intimately involved in the redox regulation and survival of TNBC, is demonstrated to be inhibited after the treatment. Consequently, precise in vivo orthotopic TNBC ablation is achieved under the guidance of IONP-enhanced magnetic resonance imaging. The results demonstrate the proof-of-concept of NIR-light-triggered self-fueling Fenton reagents against TNBC with low ferroportin levels.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Línea Celular Tumoral , Humanos , Peróxido de Hidrógeno , Hierro , Fosfatidilinositol 3-Quinasas , Neoplasias de la Mama Triple Negativas/terapia
12.
Scanning ; 2018: 3697063, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29849858

RESUMEN

Multiphoton microscopy has attracted increasing attention and investigations in the field of breast cancer, based on two-photon excited fluorescence (TPEF) and second-harmonic generation (SHG). However, the incidence of breast benign diseases is about 5 to 10 times higher than breast cancer; up to 30% of women suffer from breast benign diseases and require treatment at some time in their lives. Thus, in this study, MPM was applied to image fibroadenoma and fibrocystic lesion, which are two of the most common breast benign diseases. The results show that MPM has the capability to identify the microstructure of lobule and stroma in normal breast tissue, the interaction of compressed ducts with surrounding collagen fiber in fibroadenoma, and the architecture of cysts filled with cystic fluid in fibrocystic disease. These findings indicate that, with integration of MPM into currently accepted clinical imaging system, it has the potential to make a real-time diagnosis of breast benign diseases in vivo, as well as breast cancer.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Mama/patología , Fibroadenoma/diagnóstico , Enfermedad Fibroquística de la Mama/diagnóstico , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Neoplasias de la Mama/patología , Femenino , Fibroadenoma/patología , Enfermedad Fibroquística de la Mama/patología , Humanos
13.
Pituitary ; 21(4): 362-370, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29594837

RESUMEN

PURPOSE: If we can find a new method that can achieve rapid diagnosis of adenoma during operation, it will help surgeon shorten the operation time and enhance the treatment efficacy. This study discusses the feasibility of multiphoton microscopy (MPM) in diagnosing pituitary adenoma. METHOD: MPM, based on two-photon excited fluorescence (TPEF) and second harmonic generation (SHG) is performed for the diagnosis of pituitary adenoma in unstained sections. RESULTS: Our results show that MPM can reveal the variation of reticulin fiber by SHG signals of collagen, combined with the measurement of area of acinus, thickness of collagen fiber and collagen percentage. MPM can further reflect the change of meshwork in normal pituitary and hyperplasia quantitatively. And the characteristics of typical growth patterns of pituitary adenoma are demonstrated by the overlay of SHG and TPEF images. What's more, we can identify the boundary of normal pituitary, hyperplasia and adenoma from MPM images. And the experiment also results verify the feasibility of this method in frozen sections. CONCLUSION: These results indicated that MPM can make a diagnosis of pituitary adenoma by the morphological changes without routine pathological processing including hematoxylin-eosin (H&E) staining and other special staining. Therefore, this technique is expected to help diagnosis of pituitary adenoma during operation.


Asunto(s)
Adenoma/diagnóstico , Neoplasias Hipofisarias/diagnóstico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Femenino , Humanos , Técnicas In Vitro , Masculino , Microscopía , Persona de Mediana Edad , Adulto Joven
14.
Oncotarget ; 6(14): 12481-92, 2015 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-25980579

RESUMEN

Here we found loss of c-Cbl, an E3 ligase, expression in non-small cell lung cancer (NSCLC) compared with its adjacent normal tissue in patient specimens. HDAC inhibition by WJ or knockdown of HDAC 1, HDAC2, HDAC3 or HDAC6 all induced c-Cbl. Ectopic expression of c-Cbl induced decreased EGFR, inhibited growth in NSCLC cells. Knockdown of EGFR inhibited NSCLC growth. Mutation of EGFR at Y1045 decreased WJ-induced growth inhibition as well as in vivo anti-cancer effect and EGFR degradation mediated by WJ. Time-lapse confocal analysis showed co-localization of c-Cbl and EGFR after WJ treatment. Furthermore, WJ inhibited lung tumor growth through c-Cbl induction in orthotopic and tail vein injected models. C-Cbl up-regulation induced by HDACi is a potential strategy for NSCLC treatment.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogénicas c-cbl/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Microscopía Confocal , ARN Interferente Pequeño , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA