Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 924.617
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Biomaterials ; 313: 122757, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39178558

RESUMO

Recent progress in stem cell therapy has demonstrated the therapeutic potential of intravenous stem cell infusions for treating the life-threatening lung disease of pulmonary fibrosis (PF). However, it is confronted with limitations, such as a lack of control over cellular function and rapid clearance by the host after implantation. In this study, we developed an innovative PF therapy through tracheal administration of microfluidic-templated stem cell-laden microcapsules, which effectively reversed the progression of inflammation and fibrotic injury. Our findings highlight that hydrogel microencapsulation can enhance the persistence of donor mesenchymal stem cells (MSCs) in the host while driving MSCs to substantially augment their therapeutic functions, including immunoregulation and matrix metalloproteinase (MMP)-mediated extracellular matrix (ECM) remodeling. We revealed that microencapsulation activates the MAPK signaling pathway in MSCs to increase MMP expression, thereby degrading overexpressed collagen accumulated in fibrotic lungs. Our research demonstrates the potential of hydrogel microcapsules to enhance the therapeutic efficacy of MSCs through cell-material interactions, presenting a promising yet straightforward strategy for designing advanced stem cell therapies for fibrotic diseases.


Assuntos
Cápsulas , Matriz Extracelular , Imunomodulação , Células-Tronco Mesenquimais , Fibrose Pulmonar , Animais , Matriz Extracelular/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Fibrose Pulmonar/terapia , Fibrose Pulmonar/patologia , Transplante de Células-Tronco Mesenquimais/métodos , Camundongos Endogâmicos C57BL , Hidrogéis/química , Camundongos , Masculino
2.
Biomaterials ; 313: 122763, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39180917

RESUMO

Cuproptosis is a new kind of cell death that depends on delivering copper ions into mitochondria to trigger the aggradation of tricarboxylic acid (TCA) cycle proteins and has been observed in various cancer cells. However, whether cuproptosis occurs in cancer stem cells (CSCs) is unexplored thus far, and CSCs often reside in a hypoxic tumor microenvironment (TME) of triple negative breast cancers (TNBC), which suppresses the expression of the cuproptosis protein FDX1, thereby diminishing anticancer efficacy of cuproptosis. Herein, a ROS-responsive active targeting cuproptosis-based nanomedicine CuET@PHF is developed by stabilizing copper ionophores CuET nanocrystals with polydopamine and hydroxyethyl starch to eradicate CSCs. By taking advantage of the photothermal effects of CuET@PHF, tumor hypoxia is overcome via tumor mechanics normalization, thereby leading to enhanced cuproptosis and immunogenic cell death in 4T1 CSCs. As a result, the integration of CuET@PHF and mild photothermal therapy not only significantly suppresses tumor growth but also effectively inhibits tumor recurrence and distant metastasis by eliminating CSCs and augmenting antitumor immune responses. This study presents the first evidence of cuproptosis in CSCs, reveals that disrupting hypoxia augments cuproptosis cancer therapy, and establishes a paradigm for potent cancer therapy by simultaneously eliminating CSCs and boosting antitumor immunity.


Assuntos
Cobre , Nanomedicina , Células-Tronco Neoplásicas , Neoplasias de Mama Triplo Negativas , Microambiente Tumoral , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/terapia , Microambiente Tumoral/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Animais , Feminino , Nanomedicina/métodos , Cobre/química , Cobre/farmacologia , Linhagem Celular Tumoral , Camundongos , Nanopartículas/química , Camundongos Endogâmicos BALB C , Terapia Fototérmica/métodos , Humanos , Polímeros/química , Indóis/farmacologia
3.
Biomaterials ; 313: 122771, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39190940

RESUMO

The notorious tumor microenvironment (TME) usually becomes more deteriorative during phototherapeutic progress that hampers the antitumor efficacy. To overcome this issue, we herein report the ameliorative and adaptive nanoparticles (TPASIC-PFH@PLGA NPs) that simultaneously reverse hypoxia TME and switch photoactivities from photothermal-dominated state to photodynamic-dominated state to maximize phototherapeutic effect. TPASIC-PFH@PLGA NPs are designed by incorporating oxygen-rich liquid perfluorohexane (PFH) into the intraparticle microenvironment to regulate the intramolecular motions of AIE photosensitizer TPASIC. TPASIC exhibits a unique aggregation-enhanced reactive oxygen species (ROS) generation feature. PFH incorporation affords TPASIC the initially dispersed state, thus promoting active intramolecular motions and photothermal conversion efficiency. While PFH volatilization leads to nanoparticle collapse and the formation of tight TPASIC aggregates with largely enhanced ROS generation efficiency. As a consequence, PFH incorporation not only currently promotes both photothermal and photodynamic efficacies of TPASIC and increases the intratumoral oxygen level, but also enables the smart photothermal-to-photodynamic switch to maximize the phototherapeutic performance. The integration of PFH and AIE photosensitizer eventually delivers more excellent antitumor effect over conventional phototherapeutic agents with fixed photothermal and photodynamic efficacies. This study proposes a new nanoengineering strategy to ameliorate TME and adapt the treatment modality to fit the changed TME for advanced antitumor applications.


Assuntos
Fluorocarbonos , Nanopartículas , Fotoquimioterapia , Fármacos Fotossensibilizantes , Espécies Reativas de Oxigênio , Microambiente Tumoral , Nanopartículas/química , Microambiente Tumoral/efeitos dos fármacos , Animais , Fotoquimioterapia/métodos , Espécies Reativas de Oxigênio/metabolismo , Fluorocarbonos/química , Fluorocarbonos/farmacologia , Linhagem Celular Tumoral , Fármacos Fotossensibilizantes/uso terapêutico , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/química , Humanos , Camundongos , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Camundongos Endogâmicos BALB C , Terapia Fototérmica/métodos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Fototerapia/métodos , Feminino
4.
Biomaterials ; 313: 122778, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39213978

RESUMO

Thyroid cancer is increasing globally, with anaplastic thyroid carcinoma (ATC) being the most aggressive type and having a poor prognosis. Current clinical treatments for thyroid cancer present numerous challenges, including invasiveness and the necessity of lifelong medication. Furthermore, a significant portion of patients with ATC experience cancer recurrence and metastasis. To overcome this dilemma, we developed a pH-responsive biomimetic nanocarrier (CLP@HP-A) through the incorporation of Chlorin e6 (Ce6) and Lenvatinib (Len) within hollow polydopamine nanoparticles (HP) that were further modified with platinum nanoparticles (Pt), enabling synergistic chemotherapy and sonodynamic therapy. The CLP@HP-A nanocarriers exhibited specific binding with galectin-3 receptors, facilitating their internalization through receptor-mediated endocytosis for targeted drug delivery. Upon exposure to ultrasound (US) irradiation, Ce6 rapidly generated reactive oxygen species (ROS) to induce significant oxidative stress and trigger apoptosis in tumor cells. Additionally, Pt not only alleviated tumor hypoxia by catalyzing the conversion of H2O2 to oxygen (O2) but also augmented intracellular ROS levels through the production of hydroxyl radicals (•OH), thereby enhancing the efficacy of sonodynamic therapy. Moreover, Len demonstrated a potent cytotoxic effect on thyroid cancer cells through the induction of apoptosis. Transcriptomics analysis findings additionally corroborated that CLP@HP-A effectively triggered cancer cell apoptosis, thereby serving as a crucial mechanism for its cytotoxic effects. In conclusion, the integration of sonodynamic/chemo combination therapy with targeted drug delivery systems offers a novel approach to the management of malignant tumors.


Assuntos
Clorofilídeos , Indóis , Platina , Polímeros , Porfirinas , Neoplasias da Glândula Tireoide , Microambiente Tumoral , Terapia por Ultrassom , Neoplasias da Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/terapia , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/metabolismo , Humanos , Linhagem Celular Tumoral , Microambiente Tumoral/efeitos dos fármacos , Indóis/química , Terapia por Ultrassom/métodos , Porfirinas/química , Porfirinas/farmacologia , Polímeros/química , Animais , Platina/química , Platina/uso terapêutico , Platina/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Compostos de Fenilureia/farmacologia , Compostos de Fenilureia/uso terapêutico , Apoptose/efeitos dos fármacos , Nanopartículas/química , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Camundongos , Quinolinas/farmacologia , Quinolinas/química , Camundongos Nus , Portadores de Fármacos/química
5.
Biomaterials ; 313: 122796, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39226654

RESUMO

Chemotherapy-induced cellular senescence leads to an increased proportion of cancer stem cells (CSCs) in breast cancer (BC), contributing to recurrence and metastasis, while effective means to clear them are currently lacking. Herein, we aim to develop new approaches for selectively killing senescent-escape CSCs. High CD276 (95.60%) expression in multidrug-resistant BC cells, facilitates immune evasion by low-immunogenic senescent escape CSCs. CALD1, upregulated in ADR-resistant BC, promoting senescent-escape of CSCs with an anti-apoptosis state and upregulating CD276, PD-L1 to promote chemoresistance and immune escape. We have developed a controlled-released thermosensitive hydrogel containing pH- responsive anti-CD276 scFV engineered biomimetic nanovesicles to overcome BC in primary, recurrent, metastatic and abscopal humanized mice models. Nanovesicles coated anti-CD276 scFV selectively fuses with cell membrane of senescent-escape CSCs, then sequentially delivers siCALD1 and ADR due to pH-responsive MnP shell. siCALD1 together with ADR effectively induce apoptosis of CSCs, decrease expression of CD276 and PD-L1, and upregulate MHC I combined with Mn2+ to overcome chemoresistance and promote CD8+T cells infiltration. This combined therapeutic approach reveals insights into immune surveillance evasion by senescent-escape CSCs, offering a promising strategy to immunotherapy effectiveness in cancer therapy.


Assuntos
Neoplasias da Mama , Senescência Celular , Resistencia a Medicamentos Antineoplásicos , Células-Tronco Neoplásicas , Humanos , Animais , Neoplasias da Mama/patologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/terapia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Senescência Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Camundongos , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Engenharia Genética/métodos , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Nanopartículas/química , Anticorpos de Cadeia Única/química , Evasão Tumoral/efeitos dos fármacos , Antígeno B7-H1/metabolismo , Apoptose/efeitos dos fármacos , Biomimética/métodos , Antígenos B7
6.
Biomaterials ; 313: 122795, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39232333

RESUMO

Ferroptosis is an iron-dependent form of programmed cell death with the potential to reverse traditional cancer therapy resistance. The combination of ferroptosis with chemotherapy, photodynamic therapy and X-ray therapy has demonstrated remarkably improved therapeutic efficiency. Radiopharmaceutical therapy (RPT) is an emerging approach that achieves precise radiation to diseased tissues via radionuclide delivery. However, insufficient accumulation and retention of therapeutic radiopharmaceuticals in tumor region as well as cancer radioresistance impact treatment efficacy. Here, a nanoassembly of renal clearable ultrasmall iron nanoparticles (USINPs) and 131I-aPD-L1 is prepared via the affinity of fluorophenylboronic acid modified on the USINPs with 131I-aPD-L1. The 150 nm USINAs(131I-aPD-L1) nanoassembly is stable in blood circulation, effectively targets to the tumor and disassembles in the presence of ATP in the tumor microenvironment. Both in vitro and in vivo experiments prove that USINPs-induced ferroptosis boosted the tumor radiosensitization to 131I while 131I-mediated RPT further enhanced ferroptosis. Meanwhile, the immunogenic cell death caused by RPT and ferroptosis combined with PD-L1 immune checkpoint blockade therapy exhibits a strong antitumor immunity. This study provides a novel way to improve the tumor accumulation of ferroptosis inducer and radiopharmaceuticals, insights into the interaction between RPT and ferroptosis and an effective SPECT-guided ferroptosis-enhanced radio-immunotherapy.


Assuntos
Ferroptose , Radioisótopos do Iodo , Compostos Radiofarmacêuticos , Ferroptose/efeitos dos fármacos , Animais , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/uso terapêutico , Camundongos , Radioisótopos do Iodo/uso terapêutico , Radioisótopos do Iodo/química , Linhagem Celular Tumoral , Humanos , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Ferro/química , Camundongos Endogâmicos BALB C , Imunoterapia/métodos , Radioimunoterapia/métodos , Feminino , Neoplasias/terapia
7.
Methods Mol Biol ; 2848: 59-71, 2025.
Artigo em Inglês | MEDLINE | ID: mdl-39240516

RESUMO

Glaucoma is one of the leading causes of irreversible blindness. Stem cell therapy has shown promise in the treatment of primary open-angle glaucoma in animal models. Stem cell-free therapy using stem cell-derived trophic factors might be in demand in patients with high-risk conditions or religious restrictions. In this chapter, we describe methods for trabecular meshwork stem cell (TMSC) cultivation, secretome harvesting, and protein isolation, as well as assays to ensure the health of TMSC post-secretome harvesting and for secretome periocular injection into mice for therapeutic purposes.


Assuntos
Células-Tronco , Malha Trabecular , Malha Trabecular/metabolismo , Malha Trabecular/citologia , Animais , Camundongos , Humanos , Células-Tronco/citologia , Células-Tronco/metabolismo , Regeneração , Glaucoma/terapia , Transplante de Células-Tronco/métodos , Secretoma , Modelos Animais de Doenças , Glaucoma de Ângulo Aberto/terapia , Células Cultivadas , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Técnicas de Cultura de Células/métodos
8.
Biomaterials ; 312: 122714, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39079462

RESUMO

Osteosarcoma, a malignant bone tumor often characterized by high hedgehog signaling activity, residual tumor cells, and substantial bone defects, poses significant challenges to both treatment response and postsurgical recovery. Here, we developed a nanocomposite hydrogel for the sustained co-delivery of bioactive magnesium ions, anti-PD-L1 antibody (αPD-L1), and hedgehog pathway antagonist vismodegib, to eradicate residual tumor cells while promoting bone regeneration post-surgery. In a mouse model of tibia osteosarcoma, this hydrogel-mediated combination therapy led to remarkable tumor growth inhibition and hence increased animal survival by enhancing the activity of tumor-suppressed CD8+ T cells. Meanwhile, the implanted hydrogel improved the microenvironment of osteogenesis through long-term sustained release of Mg2+, facilitating bone defect repair by upregulating the expression of osteogenic genes. After 21 days, the expression levels of ALP, COL1, RUNX2, and BGLAP in the Vis-αPD-L1-Gel group were approximately 4.1, 5.1, 5.5, and 3.4 times higher than those of the control, respectively. We believe that this hydrogel-based combination therapy offers a potentially valuable strategy for treating osteosarcoma and addressing the tumor-related complex bone diseases.


Assuntos
Neoplasias Ósseas , Hidrogéis , Imunoterapia , Nanocompostos , Osteossarcoma , Osteossarcoma/patologia , Osteossarcoma/tratamento farmacológico , Osteossarcoma/terapia , Animais , Hidrogéis/química , Nanocompostos/química , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Neoplasias Ósseas/terapia , Camundongos , Imunoterapia/métodos , Linhagem Celular Tumoral , Regeneração Óssea/efeitos dos fármacos , Humanos , Osteogênese/efeitos dos fármacos , Antígeno B7-H1/metabolismo , Camundongos Endogâmicos BALB C , Magnésio/química
9.
Biomaterials ; 312: 122720, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39084098

RESUMO

Mesenchymal stem cells (MSCs) are expected to be useful therapeutics in osteoarthritis (OA), the most common joint disorder characterized by cartilage degradation. However, evidence is limited with regard to cartilage repair in clinical trials because of the uncontrolled differentiation and weak cartilage-targeting ability of MSCs after injection. To overcome these drawbacks, here we synthesized CuO@MSN nanoparticles (NPs) to deliver Sox9 plasmid DNA (favoring chondrogenesis) and recombinant protein Bmp7 (inhibiting hypertrophy). After taking up CuO@MSN/Sox9/Bmp7 (CSB NPs), the expressions of chondrogenic markers were enhanced while hypertrophic markers were decreased in response to these CSB-engineered MSCs. Moreover, a cartilage-targeted peptide (designated as peptide W) was conjugated onto the surface of MSCs via a click chemistry reaction, thereby prolonging the residence time of MSCs in both the knee joint cavity of mice and human-derived cartilage. In a surgery-induced OA mouse model, the NP and peptide dual-modified W-CSB-MSCs showed an enhancing therapeutic effect on cartilage repair in knee joints compared with other engineered MSCs after intra-articular injection. Most importantly, W-CSB-MSCs accelerated cartilage regeneration in damaged cartilage explants derived from OA patients. Thus, this new peptide and NPs dual engineering strategy shows potential for clinical applications to boost cartilage repair in OA using MSC therapy.


Assuntos
Diferenciação Celular , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Nanopartículas , Osteoartrite , Peptídeos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Animais , Osteoartrite/terapia , Osteoartrite/patologia , Nanopartículas/química , Humanos , Diferenciação Celular/efeitos dos fármacos , Peptídeos/química , Transplante de Células-Tronco Mesenquimais/métodos , Condrogênese/efeitos dos fármacos , Camundongos , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Cartilagem Articular/patologia , Cartilagem Articular/efeitos dos fármacos , Proteína Morfogenética Óssea 7/química , Proteína Morfogenética Óssea 7/farmacologia , Engenharia Tecidual/métodos , Regeneração/efeitos dos fármacos
10.
Biomaterials ; 312: 122709, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39094521

RESUMO

Sonodynamic therapy (SDT) relies heavily on the presence of oxygen to induce cell death. Its effectiveness is thus diminished in the hypoxic regions of tumor tissue. To address this issue, the exploration of ultrasound-based synergistic treatment modalities has become a significant research focus. Here, we report an ultrasonic cavitation effect enhanced sonodynamic and 1208 nm photo-induced cancer treatment strategy based on thermoelectric/piezoelectric oxygen-defect bismuth oxychloride nanosheets (BNs) to realize the high-performance eradication of tumors. Upon ultrasonic irradiation, the local high temperature and high pressure generated by the ultrasonic cavitation effect combined with the thermoelectric and piezoelectric effects of BNs create a built-in electric field. This facilitates the separation of carriers, increasing their mobility and extending their lifetimes, thereby greatly improving the effectiveness of SDT and NIR-Ⅱ phototherapy on hypoxia. The Tween-20 modified BNs (TBNs) demonstrate ∼88.6 % elimination rate against deep-seated tumor cells under hypoxic conditions. In vivo experiments confirm the excellent antitumor efficacy of TBNs, achieving complete tumor elimination within 10 days with no recurrences. Furthermore, due to the high X-ray attenuation of Bi and excellent NIR-Ⅱ absorption, TBNs enable precise cancer diagnosis through photoacoustic (PA) imaging and computed tomography (CT).


Assuntos
Bismuto , Neoplasias da Mama , Oxigênio , Terapia por Ultrassom , Bismuto/química , Feminino , Animais , Neoplasias da Mama/terapia , Terapia por Ultrassom/métodos , Oxigênio/química , Camundongos , Camundongos Endogâmicos BALB C , Humanos , Linhagem Celular Tumoral , Raios Infravermelhos , Nanoestruturas/química , Fototerapia/métodos
11.
Biomaterials ; 312: 122712, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39098305

RESUMO

Immunosuppressive tumor microenvironment (ITM) severely limited the efficacy of immunotherapy against triple-negative breast cancer (TNBC). Herein, Apt-LPR, a light-activatable photodynamic therapy (PDT)/RNAi immune synergy-enhancer was constructed by co-loading miR-34a and photosensitizers in cationic liposomes (in phase III clinical trial). Interestingly, the introduction of tumor-specific aptamers creates a special "Liposome-Aptamer-Target" interface, where the aptamers are initially in a "lying down" state but transform to "standing up" after target binding. The interfacing mechanism was elaborately revealed by computational and practical experiments. This unique interface endowed Apt-LPR with neutralized surface potential of cationic liposomes to reduce non-specific cytotoxicity, enhanced DNase resistance to protect aptamers, and preserved target-binding ability for selective drug delivery. Upon near-infrared irradiation, the generated reactive oxygen species would oxidize unsaturated phospholipids to destabilize both liposomes and lysosomes, realizing stepwise lysosomal escape of miR-34a for tumor cell apoptosis and downregulation of PD-L1 to suppress immune escape. Together, tumor-associated antigens released from PDT-damaged mitochondria and endoplasmic reticulum could activate the suppressive immune cells to establish an "immune hot" milieu. The collaborative immune-enhancing strategy effectively aroused systemic antitumor immunity and inhibited primary and distal tumor progression as well as lung metastasis in 4T1 xenografted mouse models. The photo-controlled drug release and specific tumor-targeting capabilities of Apt-LPR were also visualized in MDA-MB-231 xenografted zebrafish models. Therefore, this photoswitchable PDT/RNAi immune stimulator offered a powerful approach to reprogramming ITM and reinforcing cancer immunotherapy efficacy.


Assuntos
Lipossomos , MicroRNAs , Fotoquimioterapia , Fármacos Fotossensibilizantes , Neoplasias de Mama Triplo Negativas , Microambiente Tumoral , Animais , Humanos , Lipossomos/química , MicroRNAs/genética , MicroRNAs/metabolismo , Fotoquimioterapia/métodos , Microambiente Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Feminino , Neoplasias de Mama Triplo Negativas/terapia , Neoplasias de Mama Triplo Negativas/patologia , Camundongos , Aptâmeros de Nucleotídeos/química , Preparações de Ação Retardada/química , Interferência de RNA , Peixe-Zebra
12.
Biomaterials ; 312: 122745, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39098306

RESUMO

Stimulator of interferon genes (STING) agonists have shown promise in cancer treatment by stimulating the innate immune response, yet their clinical potential has been limited by inefficient cytosolic entry and unsatisfactory pharmacological activities. Moreover, aggressive tumors with "cold" and immunosuppressive microenvironments may not be effectively suppressed solely through innate immunotherapy. Herein, we propose a multifaceted immunostimulating nanoparticle (Mn-MC NP), which integrates manganese II (Mn2+) coordinated photosensitizers (chlorin e6, Ce6) and STING agonists (MSA-2) within a PEGylated nanostructure. In Mn-MC NPs, Ce6 exerts potent phototherapeutic effects, facilitating tumor ablation and inducing immunogenic cell death to elicit robust adaptive antitumor immunity. MSA-2 activates the STING pathway powered by Mn2+, thereby promoting innate antitumor immunity. The Mn-MC NPs feature a high drug-loading capacity (63.42 %) and directly ablate tumor tissue while synergistically boosting both adaptive and innate immune responses. In subsutaneous tumor mouse models, the Mn-MC NPs exhibit remarkable efficacy in not only eradicating primary tumors but also impeding the progression of distal and metastatic tumors through synergistic immunotherapy. Additionally, they contribute to preventing tumor recurrence by fostering long-term immunological memory. Our multifaceted immunostimulating nanoparticle holds significant potential for overcoming limitations associated with insufficient antitumor immunity and ineffective cancer treatment.


Assuntos
Imunoterapia , Manganês , Nanopartículas , Animais , Imunoterapia/métodos , Manganês/química , Nanopartículas/química , Camundongos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Linhagem Celular Tumoral , Humanos , Porfirinas/química , Porfirinas/farmacologia , Clorofilídeos , Neoplasias/terapia , Neoplasias/imunologia , Fotoquimioterapia/métodos , Imunidade Inata/efeitos dos fármacos , Feminino , Camundongos Endogâmicos C57BL , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/química
13.
Biomaterials ; 312: 122724, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39106818

RESUMO

The residual bone tumor and defects which is caused by surgical therapy of bone tumor is a major and important problem in clinicals. And the sequential treatment for irradiating residual tumor and repairing bone defects has wildly prospects. In this study, we developed a general modification strategy by gallic acid (GA)-assisted coordination chemistry to prepare black calcium-based materials, which combines the sequential photothermal therapy of bone tumor and bone defects. The GA modification endows the materials remarkable photothermal properties. Under the near-infrared (NIR) irradiation with different power densities, the black GA-modified bone matrix (GBM) did not merely display an excellent performance in eliminating bone tumor with high temperature, but showed a facile effect of the mild-heat stimulation to accelerate bone regeneration. GBM can efficiently regulate the microenvironments of bone regeneration in a spatial-temporal manner, including inflammation/immune response, vascularization and osteogenic differentiation. Meanwhile, the integrin/PI3K/Akt signaling pathway of bone marrow mesenchymal stem cells (BMSCs) was revealed to be involved in the effect of osteogenesis induced by the mild-heat stimulation. The outcome of this study not only provides a serial of new multifunctional biomaterials, but also demonstrates a general strategy for designing novel blacked calcium-based biomaterials with great potential for clinical use.


Assuntos
Neoplasias Ósseas , Regeneração Óssea , Cálcio , Ácido Gálico , Células-Tronco Mesenquimais , Ácido Gálico/química , Regeneração Óssea/efeitos dos fármacos , Animais , Cálcio/metabolismo , Neoplasias Ósseas/terapia , Neoplasias Ósseas/tratamento farmacológico , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Terapia Fototérmica/métodos , Osteogênese/efeitos dos fármacos , Camundongos , Humanos , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Linhagem Celular Tumoral
14.
Biomaterials ; 312: 122743, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39111233

RESUMO

Photodynamic therapy (PDT) is an appealing modality for cancer treatments. However, the limited tissue penetration depth of external-excitation light makes PDT impossible in treating deep-seated tumors. Meanwhile, tumor hypoxia and intracellular reductive microenvironment restrain the generation of reactive oxygen species (ROS). To overcome these limitations, a tumor-targeted self-illuminating supramolecular nanoparticle T-NPCe6-L-N is proposed by integrating photosensitizer Ce6 with luminol and nitric oxide (NO) for chemiluminescence resonance energy transfer (CRET)-activated PDT. The high H2O2 level in tumor can trigger chemiluminescence of luminol to realize CRET-activated PDT without exposure of external light. Meanwhile, the released NO significantly relieves tumor hypoxia via vascular normalization and reduces intracellular reductive GSH level, further enhancing ROS abundance. Importantly, due to the different ROS levels between cancer cells and normal cells, T-NPCe6-L-N can selectively trigger PDT in cancer cells while sparing normal cells, which ensured low side effect. The combination of CRET-based photosensitizer-activation and tumor microenvironment modulation overcomes the innate challenges of conventional PDT, demonstrating efficient inhibition of orthotopic and metastatic tumors on mice. It also provoked potent immunogenic cell death to ensure long-term suppression effects. The proof-of-concept research proved as a new strategy to solve the dilemma of PDT in treatment of deep-seated tumors.


Assuntos
Nanopartículas , Fotoquimioterapia , Fármacos Fotossensibilizantes , Microambiente Tumoral , Fotoquimioterapia/métodos , Microambiente Tumoral/efeitos dos fármacos , Animais , Nanopartículas/química , Fármacos Fotossensibilizantes/uso terapêutico , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Humanos , Camundongos , Linhagem Celular Tumoral , Espécies Reativas de Oxigênio/metabolismo , Transferência de Energia , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Camundongos Endogâmicos BALB C , Luz , Camundongos Nus , Óxido Nítrico/metabolismo
15.
Biomaterials ; 312: 122751, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39121726

RESUMO

Tumor immunotherapies have emerged as a promising frontier in the realm of cancer treatment. However, challenges persist in achieving localized, durable immunostimulation while counteracting the tumor's immunosuppressive environment. Here, we develop a natural mussel foot protein-based nanomedicine with spatiotemporal control for tumor immunotherapy. In this nanomedicine, an immunoadjuvant prodrug and a photosensitizer are integrated, which is driven by their dynamic bonding and non-covalent assembling with the protein carrier. Harnessing the protein carrier's bioadhesion, this nanomedicine achieves a drug co-delivery with spatiotemporal precision, by which it not only promotes tumor photothermal ablation but also broadens tumor antigen repertoire, facilitating in situ immunotherapy with durability and maintenance. This nanomedicine also modulates the tumor microenvironment to overcome immunosuppression, thereby amplifying antitumor responses against tumor progression. Our strategy underscores a mussel foot protein-derived design philosophy of drug delivery aimed at refining combinatorial immunotherapy, offering insights into leveraging natural proteins for cancer treatment.


Assuntos
Imunoterapia , Nanomedicina , Animais , Imunoterapia/métodos , Nanomedicina/métodos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/uso terapêutico , Fármacos Fotossensibilizantes/farmacologia , Terapia Fototérmica/métodos , Camundongos , Humanos , Microambiente Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral , Proteínas/química , Feminino , Neoplasias/terapia , Neoplasias/imunologia , Adesivos/química , Camundongos Endogâmicos C57BL , Adjuvantes Imunológicos/farmacologia
16.
Biomaterials ; 312: 122755, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39151270

RESUMO

Copper-catalyzed click chemistry offers creative strategies for activation of therapeutics without disrupting biological processes. Despite tremendous efforts, current copper catalysts face fundamental challenges in achieving high efficiency, atom economy, and tissue-specific selectivity. Herein, we develop a facile "mix-and-match synthetic strategy" to fabricate a biomimetic single-site copper-bipyridine-based cerium metal-organic framework (Cu/Ce-MOF@M) for efficient and tumor cell-specific bioorthogonal catalysis. This elegant methodology achieves isolated single-Cu-site within the MOF architecture, resulting in exceptionally high catalytic performance. Cu/Ce-MOF@M favors a 32.1-fold higher catalytic activity than the widely used MOF-supported copper nanoparticles at single-particle level, as first evidenced by single-molecule fluorescence microscopy. Furthermore, with cancer cell-membrane camouflage, Cu/Ce-MOF@M demonstrates preferential tropism for its parent cells. Simultaneously, the single-site CuII species within Cu/Ce-MOF@M are reduced by upregulated glutathione in cancerous cells to CuI for catalyzing the click reaction, enabling homotypic cancer cell-activated in situ drug synthesis. Additionally, Cu/Ce-MOF@M exhibits oxidase and peroxidase mimicking activities, further enhancing catalytic cancer therapy. This study guides the reasonable design of highly active heterogeneous transition-metal catalysts for targeted bioorthogonal reactions.


Assuntos
Materiais Biomiméticos , Cobre , Humanos , Cobre/química , Materiais Biomiméticos/química , Catálise , Estruturas Metalorgânicas/química , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Cério/química , Linhagem Celular Tumoral , Animais , Química Click/métodos , Biomimética/métodos , Camundongos
17.
Biomaterials ; 313: 122776, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39236629

RESUMO

Surgical resection, the mainstay for melanoma treatment, faces challenges due to high tumor recurrence rates and complex postoperative wound healing. Chronic inflammation from residual disease and the risk of secondary infections impede healing. We introduce an innovative, injectable hydrogel system that integrates a multifaceted therapeutic approach. The hydrogel, crosslinked by calcium ions with sodium alginate, encapsulates a blood clot rich in dendritic cells (DCs) chemoattractants and melanoma cell-derived nanovesicles (NVs), functioning as a potent immunostimulant. This in situ recruitment strategy overcomes the limitations of subcutaneous tumor vaccine injections and more effectively achieves antitumor immunity. Additionally, the hydrogel incorporates Chlorella extracts, enhancing its antimicrobial properties to prevent wound infections and promote healing. One of the key findings of our research is the dual functionality of Chlorella extracts; they not only expedite the healing process of infected wounds but also increase the hydrogel's ability to stimulate an antitumor immune response. Given the patient-specific nature of the blood clot and NVs, our hydrogel system offers customizable solutions for individual postoperative requirements. This personalized approach is highlighted by our study, which demonstrates the synergistic impact of the composite hydrogel on preventing melanoma recurrence and hastening wound healing, potentially transforming postsurgical melanoma management.


Assuntos
Células Dendríticas , Hidrogéis , Melanoma , Cicatrização , Hidrogéis/química , Animais , Células Dendríticas/imunologia , Células Dendríticas/efeitos dos fármacos , Melanoma/terapia , Melanoma/patologia , Cicatrização/efeitos dos fármacos , Humanos , Recidiva Local de Neoplasia/prevenção & controle , Camundongos Endogâmicos C57BL , Anti-Infecciosos/uso terapêutico , Anti-Infecciosos/farmacologia , Camundongos , Linhagem Celular Tumoral , Feminino
18.
Biomaterials ; 313: 122788, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39236628

RESUMO

Drug resistance is a significant challenge in cancer chemotherapy and is a primary factor contributing to poor recovery for cancer patients. Although drug-loaded nanoparticles have shown promise in overcoming chemotherapy resistance, they often carry a combination of drugs and require advanced design and manufacturing processes. Furthermore, they seldom approach chemotherapy-resistant tumors from an immunotherapy perspective. In this study, we developed a therapeutic nanovaccine composed solely of chemotherapy-induced resistant tumor antigens (CIRTAs) and the immune adjuvant Toll-like receptor (TLR) 7/8 agonist R848 (CIRTAs@R848). This nanovaccine does not require additional carriers and has a simple production process. It efficiently delivers antigens and immune stimulants to dendritic cells (DCs) simultaneously, promoting DCs maturation. CIRTAs@R848 demonstrated significant tumor suppression, particularly when used in combination with the immune checkpoint blockade (ICB) anti-PD-1 (αPD-1). The combined therapy increased the infiltration of T cells into the tumor while decreasing the proportion of regulatory T cells (Tregs) and modulating the tumor microenvironment, resulting in long-term immune memory. Overall, this study introduces an innovative strategy for treating chemotherapy-resistant tumors from a novel perspective, with potential applications in personalized immunotherapy and precision medicine.


Assuntos
Vacinas Anticâncer , Desoxicitidina , Resistencia a Medicamentos Antineoplásicos , Gencitabina , Imunoterapia , Nanopartículas , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapêutico , Desoxicitidina/farmacologia , Animais , Imunoterapia/métodos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Nanopartículas/química , Camundongos , Humanos , Células Dendríticas/imunologia , Células Dendríticas/efeitos dos fármacos , Linhagem Celular Tumoral , Camundongos Endogâmicos C57BL , Feminino , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos , Antígenos de Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/tratamento farmacológico , Nanovacinas
19.
Biomaterials ; 313: 122801, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39236630

RESUMO

Chemoimmunotherapy is an emerging paradigm in the clinic for treating several malignant diseases, such as non-small cell lung cancer, breast cancer, and large B-cell lymphoma. However, the efficacy of this strategy is still restricted by serious adverse events and a high therapeutic termination rate, presumably due to the lack of tumor-targeted distribution of both chemotherapeutic and immunotherapeutic agents. Targeted drug delivery has the potential to address this issue. Among the most promising nanocarriers in clinical translation, liposomes have drawn great attention in cancer chemoimmunotherapy in recent years. Liposomes-enabled cancer chemoimmunotherapy has made significant progress in clinics, with impressive therapeutic outcomes. This review summarizes the latest preclinical and clinical progress in liposome-enabled cancer chemoimmunotherapy and discusses the challenges and future directions of this field.


Assuntos
Imunoterapia , Lipossomos , Neoplasias , Lipossomos/química , Humanos , Imunoterapia/métodos , Animais , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Antineoplásicos/uso terapêutico , Antineoplásicos/administração & dosagem
20.
Biomaterials ; 313: 122775, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39241549

RESUMO

Acute Myocardial Infarction (AMI) has seen rising cases, particularly in younger people, leading to public health concerns. Standard treatments, like coronary artery recanalization, often don't fully repair the heart's microvasculature, risking heart failure. Advances show that Mesenchymal Stromal Cells (MSCs) transplantation improves cardiac function after AMI, but the harsh microenvironment post-AMI impacts cell survival and therapeutic results. MSCs aid heart repair via their membrane proteins and paracrine extracellular vesicles that carry microRNA-125b, which regulates multiple targets, preventing cardiomyocyte death, limiting fibroblast growth, and combating myocardial remodeling after AMI. This study introduces ultrasound-responsive phase-change bionic nanoparticles, leveraging MSCs' natural properties. These particles contain MSC membrane and microRNA-125b, with added macrophage membrane for stability. Using Ultrasound Targeted Microbubble Destruction (UTMD), this method targets the delivery of MSC membrane proteins and microRNA-125b to AMI's inflamed areas. This aims to enhance cardiac function recovery and provide precise, targeted AMI therapy.


Assuntos
Células-Tronco Mesenquimais , MicroRNAs , Infarto do Miocárdio , Nanopartículas , Infarto do Miocárdio/terapia , Animais , Nanopartículas/química , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , MicroRNAs/metabolismo , MicroRNAs/genética , Masculino , Recuperação de Função Fisiológica , Transplante de Células-Tronco Mesenquimais/métodos , Humanos , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Camundongos , Microbolhas , Ondas Ultrassônicas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA