Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
2.
Nanoscale Horiz ; 9(2): 295-304, 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38086653

RESUMEN

Cancer immunotherapy has received increasing attention in tumor therapy. However, insufficient infiltration of T cells and over-expressed PD-L1 checkpoint in tumor cells severely impede cancer immunotherapy. Here, an injectable hydrogel was designed to reinforce T cell infiltration and inactivate PD-L1 for powerful cancer immunotherapy. The hydrogel was created by sodium alginate (SA) as the gelator, where linagliptin particles and BMS-202 particles were present in hydrogel micropores. After gelation in the tumor site, the linagliptin powerfully suppressed chemokine CXCL10 degradation, enabling the introduced CXCL10 to realize sustainable chemotaxis towards strong T cell infiltration. Meanwhile, the BMS-202 inactivated PD-L1 of tumor cells, thereby eliminating the PD-L1-governed immune evasion. Therefore, the hydrogel in combination with CXCL10 demonstrated powerful cancer immunotherapy against primary and distant tumors, along with efficient inhibition of lung metastasis. Our study not only offers a potent platform against tumors, but also provides a conceptually new approach to reinforce cancer immunotherapy.


Asunto(s)
Neoplasias Pulmonares , Linfocitos T , Humanos , Linfocitos T/metabolismo , Antígeno B7-H1/metabolismo , Hidrogeles , Evasión Inmune , Linagliptina , Neoplasias Pulmonares/terapia , Inmunoterapia
3.
Int J Pharm ; 650: 123747, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38151104

RESUMEN

Boron neutron capture therapy (BNCT), as an innovative radiotherapy technology, has demonstrated remarkable outcomes when compared to conventional treatments in the management of recurrent and refractory brain tumors. However, in BNCT of brain tumors, the blood-brain barrier is a main stumbling block for restricting the transport of boron drugs to brain tumors, while the tumor targeting and retention of boron drugs also affect the BNCT effect. This review focuses on the recent development of strategies for delivering boron drugs crossing the blood-brain barrier and targeting brain tumors, providing new insights for the development of efficient boron drugs for the treatment of brain tumors.


Asunto(s)
Terapia por Captura de Neutrón de Boro , Neoplasias Encefálicas , Glioma , Humanos , Barrera Hematoencefálica , Boro , Glioma/tratamiento farmacológico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Compuestos de Boro
4.
Acta Biomater ; 169: 306-316, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37574158

RESUMEN

Prophylactic tumor vaccines hold great promise against tumor occurrence. However, their clinical efficacy remains low due to inadequate activation of strong-sustainable immunity. Herein, a biomembrane hydrogel was designed as a powerful single-shot prophylactic tumor vaccine. Mannose-decorated hybrid biomembrane (MHCM) modified with oxidized sodium alginate (OSA) was designed as a gelator (O-MHCM), where the hybrid biomembrane (HCM) is a hybridization of bacterial outer membrane vesicles (OMV) and tumor cell membranes (TCM). The O-MHCM enables quick gelation subcutaneously where the cysteine protease inhibitor E64 is encapsulated in hydrogel micropores. After a single vaccination of E64@O-MHCM hydrogel, MHCM and E64 are released sustainably due to OSA moiety degradation. The MHCM enables active targeting to dendritic cells (DC) and effective DC maturation. Meanwhile, the E64 enables sufficient antigen availability for subsequent cross presentation. Ultimately, strong and sustainable T lymphocyte-mediated immunity was elicited, demonstrating a strong prophylactic effect against breast tumors. This study provides a long-lasting platform to prevent tumor occurrence, opening an innovative avenue for the design of a single-shot prophylactic tumor vaccine. STATEMENT OF SIGNIFICANCE: Developing a single-shot prophylactic tumor vaccine to elicit strong-sustainable immunity is of great interest clinically. Here, a prophylactic tumor vaccine was designed using an injectable biomembrane hydrogel for achieving strong-sustainable immunity. The mannose-tailored hybrid biomembrane was modified with oxidized sodium alginate to result in a gelator, which enabled the formation of the hydrogel after subcutaneous injection. Cysteine protease inhibitor E64 was incorporated into the micropores of the hydrogel. The hydrogel induced strong-sustainable immunity through the continuous release of active components. This was facilitated by the mannose moiety, which enabled active targeting, as well as the antigen and adjuvant function of biomembrane, and the E64-enabled suppression of antigen degradation. The biomembrane hydrogel demonstrated powerful prevention of 4T1 breast tumors. This study offers an attractive strategy for designing a single-shot prophylactic tumor vaccine.


Asunto(s)
Neoplasias de la Mama , Vacunas contra el Cáncer , Humanos , Femenino , Hidrogeles/farmacología , Manosa , Linfocitos T , Antígenos , Neoplasias de la Mama/tratamiento farmacológico , Células Dendríticas
5.
J Control Release ; 358: 345-357, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37150404

RESUMEN

T cell-based immunotherapy (TCBI) is an emerging approach to combat tumors. However, the outcome of TCBI is still far from satisfaction clinically, owing to stumbling blocks from insufficient immunogenicity, T cell exhaustion and immune evasion from programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) pathway. Herein, an injectable tumor lysates-constructed hydrogel is reported to address these issues. Chemically modified tumor lysates are, for the first time, designed as the gelator to intratumorally construct hydrogel, achieving a robust antigen reservoir to induce strong immunogenicity. Meanwhile, hydrogel-encapsulated nicotinamide riboside and SB415286 enable strong mitophagy in T cells to prevent their exhaustion as well as powerfully genetical suppression of PD-1 expression to regulate immune evasion. Thus, our injectable hydrogel creates a robust immune niche within tumor, enabling to significantly potentiate TCBI. Our strategy pharmacologically regulates body's own T cells in situ, demonstrating potent immunotherapeutic effects and offering a conceptually new approach for TCBI.


Asunto(s)
Hidrogeles , Neoplasias , Humanos , Receptor de Muerte Celular Programada 1 , Linfocitos T/metabolismo , Inmunoterapia , Microambiente Tumoral
6.
ACS Nano ; 17(11): 10376-10392, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37194951

RESUMEN

Owing to the insidious onset of ovarian cancer, most patients are in the advanced stage with extensive peritoneal metastasis when they are diagnosed. Treatment of peritoneal metastasis from advanced ovarian cancer remains a significant challenge. Inspired by the massive macrophages in the peritoneal environment, here, we reported an artificial exosome-based peritoneal-localized hydrogel to domesticate peritoneal macrophages as the therapeutic target for realizing potent ovarian cancer therapy, where artificial exosomes derived from genetically sialic-acid-binding Ig-like lectin 10 (Siglec-10)-engineered M1-type macrophages were chemically designed as gelator. Upon triggering immunogenicity with X-ray radiation, our hydrogel encapsulating efferocytosis inhibitor MRX-2843 enabled a cascade regulation to orchestrate polarization, efferocytosis, and phagocytosis of peritoneal macrophages for realizing robust phagocytosis of tumor cells and powerful antigen presentation, offering a potent approach for ovarian cancer therapy via bridging the innate effector function of macrophages with their adaptive immune response. Moreover, our hydrogel is also applicable for potent treatment of inherent CD24-overexpressed triple-negative breast cancer, providing an emerging therapeutic regimen for the most lethal malignancies in women.


Asunto(s)
Exosomas , Neoplasias Ováricas , Neoplasias Peritoneales , Humanos , Femenino , Exosomas/patología , Hidrogeles , Macrófagos , Neoplasias Ováricas/tratamiento farmacológico
7.
Mater Today Bio ; 19: 100577, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36846308

RESUMEN

"Glutamine addiction" is a unique feature of triple negative breast cancer (TNBC), which has a higher demand for glutamine and is more susceptible to glutamine depletion. Glutamine can be hydrolyzed to glutamate by glutaminase (GLS) for synthesis of glutathione (GSH), which is an important downstream of glutamine metabolic pathways in accelerating TNBC proliferation. Consequently, glutamine metabolic intervention suggests potential therapeutic effects against TNBC. However, the effects of GLS inhibitors are hindered by glutamine resistance and their own instability and insolubility. Therefore, it is of great interest to harmonize glutamine metabolic intervention for an amplified TNBC therapy. Unfortunately, such nanoplatform has not been realized. Herein, we reported a self-assembly nanoplatform (BCH NPs) with a core of the GLS inhibitor Bis-2-(5-phenylacetamido-1,3,4-thiadiazol-2-yl) ethyl sulfide (BPTES) and photosensitizer Chlorin e6 (Ce6) and a shell of human serum albumin (HSA), enabling effective harmonization of glutamine metabolic intervention for TNBC therapy. BPTES inhibited the activity of GLS to block the glutamine metabolic pathways, thereby inhibiting the production of GSH to amplify the photodynamic effect of Ce6. While Ce6 not only directly killed tumor cells by producing excessive reactive oxygen species (ROS), but also deplete GSH to destroy redox balance, thus enhancing the effects of BPTES when glutamine resistance occurred. BCH NPs effectively eradicated TNBC tumor and suppressed tumor metastasis with favorable biocompatibility. Our work provides a new insight for photodynamic-mediated glutamine metabolic intervention against TNBC.

8.
Acta Biomater ; 159: 300-311, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36642338

RESUMEN

Ferroptosis has received increasing attentions in cancer therapy owing to its unique advantages over apoptosis. However, ferroptosis is governed by the efficiency of reactive oxygen species (ROS) production and the tumor cell antioxidant microenvironment that compromises therapeutic efficacy of ferroptosis. It is of great significance to develop a strategy that can both achieve high-efficiency ROS production and modulate tumor cell antioxidant microenvironment to amplify ferroptosis. However, until now, such a strategy has rarely been realized. Here, we, for the first time, reported a radiotherapy -mediated redox homeostasis-controllable nanomedicine for amplifying ferroptosis sensitivity in tumor therapy. The nanomedicine is constructed by co-assembling a ferroptosis inducer hemin and a thioredoxin 1 (Trx-1) inhibitor 1-methylpropyl 2-imidazolyl disulfide (PX-12) with human serum albumin. For our nanomedicine, hemin converts H2O2 to ROS via Fenton reaction to induce ferroptosis while PX-12 effectively inhibits the activity of antioxidant Trx-1 to suppress ROS depletion, resulting in amplified ferroptosis. Particularly, combining radiotherapy with the nanomedicine, radiotherapy depletes the other key antioxidant glutathione and generates additional radiotherapy-induced ROS, further boosting the ferroptosis effect. Therefore, our strategy can simultaneously ensure efficient ROS production and regulation of tumor cell antioxidant microenvironment, thereby enhancing efficacy of ferroptosis in tumor therapy. Our work offers an innovative approach to amplify ferroptosis sensitivity against tumors by simultaneously promoting ROS production and regulating redox homeostasis. STATEMENT OF SIGNIFICANCE: The antioxidants such as thioredoxin 1 (Trx-1) and glutathione (GSH) in tumor cells, are significantly upregulated by the innate cancer cellular redox homeostasis, severely restricting the reactive oxygen species (ROS)-based therapy and compromising the effect of Fenton reaction-induced ferroptosis against tumors. It is urgent to develop a strategy to simultaneously achieve Fenton reaction-induced ferroptosis and regulate the cancer cellular redox homeostasis against upregulated levels of Trx-1 and GSH. A radiotherapy-mediated redox homeostasis-regulatable nanomedicine was designed for amplifying ferroptosis sensitivity in tumor therapy, where the therapeutic efficacy of ferroptosis against tumors can be significantly amplified by integrating Fenton reaction-induced and radiotherapy-induced ferroptosis as well as PX-12-enabled inhibition of antioxidant Trx-1 and radiotherapy-induced downregulation of antioxidant GSH levels.


Asunto(s)
Ferroptosis , Neoplasias , Humanos , Antioxidantes/farmacología , Especies Reactivas de Oxígeno , Nanomedicina , Hemina/farmacología , Peróxido de Hidrógeno/farmacología , Oxidación-Reducción , Glutatión/metabolismo , Homeostasis , Tiorredoxinas/metabolismo , Tiorredoxinas/farmacología , Línea Celular Tumoral , Microambiente Tumoral
9.
Small ; 18(32): e2202663, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35843879

RESUMEN

T cell exhaustion caused by mitochondrial dysfunction is the major obstacle of T cells-based cancer immunotherapy. Besides exhausted T cells, the insufficient major histocompatibility complex class I (MHC I) on tumor cells leads to inefficient T cell recognition of tumor cells, compromising therapeutic efficacy. Therapeutic platform to regulate T cell exhaustion and MHC I expression for boosting T cells-based cancer immunotherapy has not been realized up to date. Herein, an injectable hydrogel is designed to simultaneously tune T cell exhaustion and MHC I expression for amplified cancer immunotherapy. The hydrogel is in situ constructed in tumor site by utilizing oxidized sodium alginate-modified tumor cell membrane vesicle (O-TMV) as a gelator, where axitinib is encapsulated in the lipid bilayer of O-TMV while 4-1BB antibody and proprotein convertase subtilisin/kexin type 9 inhibitor PF-06446846 nanoparticles are present in the cavities of hydrogel. After immune response trigged by O-TMV antigen, the 4-1BB antibody-promoted T cell mitochondrial biogenesis and the axitinib-lowered hypoxia synergistically reverse T cell exhaustion while the PF-06446846-amplified MHC I expression facilitates T cell recognition of tumor cells, demonstrating a powerful immunotherapeutic efficacy. This strategy on reprograming T cell exhaustion and improving T cell potency offers new concept for T cells-based cancer immunotherapy.


Asunto(s)
Neoplasias , Linfocitos T , Anticuerpos , Axitinib , Antígenos de Histocompatibilidad Clase I , Humanos , Hidrogeles , Inmunoterapia , Neoplasias/terapia
10.
J Mol Graph Model ; 116: 108239, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35696774

RESUMEN

The carbamate molecule rivastigmine was found to possess promising anti-acetylcholinesterase activity, enabling to target and occupy choline binding sites, and as a result, widely used to improve the treatment of Alzheimer's disease (AD). Higher dose of rivastigmine indicates rapid onset but more adverse effects, such as the large fluctuations in plasma concentration level and frequent incidence of gastrointestinal side effect. To solve the dilemma, we developed a three-dimensional quantitative structure-activity relationship (3D-QSAR), docking and molecular dynamics (MD) simulation strategy to construct a dismountable nanoplatform of inhibitor engineering, verification and application for improving the inhibitory activity per unit concentration. With the aid of 3D-QSAR method, we constructed a model by using 25 molecules reported, and verified well the rationality of these QSAR models by non-cross validation coefficient (r2 = 0.902). Docking and MD results show that rivastigmine, as a control, does target exactly the binding sites of acetylcholinesterase, those already observed experimentally, in turn, confirming the reliability of the present 3D-QSAR results. The method suggests that groups with electron-donating chemical property can improve the inhibitory activity, and screens out two novel inhibitors L-1 and L-2 with more activity from database (about 8000 compounds). Moreover, L-1 and L-2 not only target exactly the same binding sites of acetylcholinesterase as the rivastigmine does, but also hold stronger binding energy, showing a more powerful inhibitory ability. More broadly, this work showcases an approach in the engineering of carbamate inhibitors to enhance their inhibitory activity using electron-donating groups, which simplifies the design process of complex bioactive molecules.


Asunto(s)
Inhibidores de la Colinesterasa , Relación Estructura-Actividad Cuantitativa , Acetilcolinesterasa/química , Inhibidores de la Colinesterasa/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Reproducibilidad de los Resultados , Rivastigmina/farmacología
11.
Acta Biomater ; 147: 366-376, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35588995

RESUMEN

Immune response is initiated by dendritic cells (DCs), where the cross-presentation of antigens by DCs determines the activating of cytotoxic T cells. However, the efficacy of DCs-initiated immune response is governed by multiple (cascade) steps of immunogenic cell death (ICD), recruitment of DCs, and cross-presentation of DCs. It is urgent but challenging to achieve a platform for simultaneously regulating these multiple steps, amplifying the immune response against tumors. Herein, we reported a photodynamic nanodrug enabling simultaneous regulation of these multiple steps for realizing powerful immune response. The nanodrug was designed by the co-assembling of chlorin e6 (Ce6), celecoxib and 6-thio-2'-deoxyguanosine (6-thio-dG). In our nanodrug, Ce6 enables induction of ICD, while celecoxib down-regulates the prostaglandin E2 (PGE2) for promoting recruitment of DCs enabled by chemokine CCL5 produced from natural killer (NK) cells. Moreover, 6-thio-dG triggers DNA damages in the tumor cells, which in turn activates STING/interferon I pathway for enhancing the cross-presentation ability of DCs. Therefore, an amplified immune therapeutic effect against tumors is achieved, thanks to the simultaneous regulation of these multiple steps. The nanodrug effectively inhibits tumor growth and postoperative recurrence, demonstrating a new approach for boosting immune response initiated by DCs in cancer therapy. STATEMENT OF SIGNIFICANCE: The dendritic cells (DCs)-initiated immune response against tumors is dominated by multiple (cascade) steps including the process of (I) immunogenic cell death (ICD), (II) recruitment of DCs, and (III) cross-presentation of antigens by DCs. Based on this, it is urgent to design a nanoplatform enabling simultaneous regulation of these multiple steps for achieving a potent therapeutic efficacy. A carrier-free photodynamic nanodrug, engineered by a co-assembling approach, was designed to regulate DCs for realizing a powerful DCs-initiated immune response against tumors, thanks to the simultaneous regulation of the above multiple steps. Our nanodrug demonstrated a boosted immune response against tumors, powerfully suppressing primary/abscopal tumor growth and postoperative recurrence, which offers a conceptually innovative strategy for amplifying immunity against tumors.


Asunto(s)
Nanopartículas , Neoplasias , Celecoxib , Línea Celular Tumoral , Células Dendríticas , Humanos , Inmunoterapia , Nanopartículas/uso terapéutico , Neoplasias/metabolismo
12.
Nanoscale Horiz ; 7(2): 198-210, 2022 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-35023537

RESUMEN

Ferroptosis, an unusual non-apoptotic cell death caused by the iron-dependent accumulation of lipid peroxide, enables the flexible design of an antitumor platform. Specifically, as a positive role, ferroptosis can induce an immune response accompanied with the interferon-γ (IFN-γ)-triggered disruption of the glutathione peroxidase 4 pathway for cascade enhancement of ferroptotic cell death and ferroptosis-induced immunotherapeutic efficacy. However, as a negative role, ferroptosis also triggers inflammation-associated immunosuppression by up-regulation of the cyclooxygenase-2/prostaglandin E2 pathway and IFN-γ-associated adaptive immune resistance by up-regulation of programmed death ligand-1 (PD-L1), impeding the antitumor efficacy of multiple immune cells by immune escape. Negative and positive roles endow ferroptosis with a Janus-faced nature. It is urgent to manipulate the Janus-faced nature of ferroptosis for eliciting the maximized ferroptotic therapeutic efficacy. Herein, a self-amplifying nanodrug (RCH NPs) was designed by co-assembling hemin (ferric porphyrin), celecoxib (anti-inflammatory drug) and roscovitine (cyclin-dependent kinase 5 inhibitor) with the assistance of human serum albumin for reprograming the Janus-faced nature of ferroptosis. During hemin-triggered ferroptosis, celecoxib disrupted the inflammation-related immunosuppression while roscovitine destroyed the IFN-γ-induced up-regulation of PD-L1 via the genetic blockade effect. The RCH NPs thus demonstrated superior therapeutic effects on tumors, thanks to self-amplifying ferroptotic immunotherapy. Our work offers a conceptually innovative strategy for harnessing ferroptosis against tumors.


Asunto(s)
Ferroptosis , Nanopartículas , Neoplasias , Muerte Celular , Humanos , Inmunoterapia , Nanopartículas/uso terapéutico , Neoplasias/tratamiento farmacológico
13.
J Colloid Interface Sci ; 610: 698-708, 2022 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-34863556

RESUMEN

Non-small cell lung cancer (NSCLC) is a type of cancer dominated by metastasis-induced death. The transcription factor BTB and CNC homology 1 (Bach1) regulates almost all metastasis steps by activating the transcription of critical metastatic genes. It is urgent to engineer a nanodrug enabling regulation of Bach1 against tumor metastasis. Herein, a minimalist nanodrug integrating chemodynamic therapy (CDT) and Bach1 degradation was reported to prevent metastasis of NSCLC. The nanodrug was achieved by self-assembly of ferrocene (Fc) and Tin protoporphyrin IX (TinPPIX). In our nanodrug, Fc not only triggers the production of highly cytotoxic ∙OH for tumor ablation via Fenton reaction, but also induces heme release from heme-containing proteins to stimulate Bach 1 degradation. Moreover, TinPPIX further augments the free heme level along with amplifies the CDT efficacy by disabling heme oxygenase-1 (HO-1)-mediated heme conversion into antioxidative bilirubin. The results showed that, compared with control group, TinPPIX/Fc nanodrug caused a four-fold increase in heme level, which triggered remarkable Bach1 degradation in Fbxo22-mediated manner and successfully inhibited Bach1-dominated metastasis. Therefore, this nanodrug could powerfully impeded NSCLC progression and metastasis, offering an innovative heme-regulatable chemodynamic therapeutic approach for lung cancer with strong metastasis capability.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Proteínas F-Box , Neoplasias Pulmonares , Células A549 , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Hemo/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Ratones SCID , Nanomedicina , Metástasis de la Neoplasia , Receptores Citoplasmáticos y Nucleares
14.
J Control Release ; 340: 282-291, 2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34740722

RESUMEN

Targeting breast cancer stem cells (BCSCs) therapy is a prospective strategy to eliminate tumors owing to the BCSCs-governed drug resistance, tumor progression and metastasis. BCSCs are intrinsically in a disequilibrium state with favorable ability of self-renewal rather than differentiation, resulting in inability of complete tumor eradication. Besides the original BCSCs, epithelial-mesenchymal transition (EMT) process can further facilitate BCSCs regeneration, accompanied by tumor progression and metastasis. Herein, we, for the first time, engineered a photodynamic nanoplatform to manipulate BCSCs against tumor progression and metastasis by not only remolding the disequilibrium state but also blocking the EMT process. The HP@PP was constructed by haloperidol (HP)-incorporated polyethyleneimine-polyhistidine (PP) micelles, which was further integrated with low molecular weight heparin (LMWH)-chlorin e6 (Ce6) conjugate (LC) to form HP@PP/LC nanoparticles (NPs). For HP@PP/LC NPs, the protonation of PP in tumor tissues precisely targeted HP to BCSCs for remolding the disequilibrium state via promoting BCSCs differentiation into tumor cells. Simultaneously, LC conjugate targeted to tumors for exerting EMT blocking ability with LMWH, as well as exerting photodynamic clearance of tumor cells with Ce6 component. Therefore, our nanoplatform provides an emerging strategy for manipulating BCSCs against tumor progression and metastasis, demonstrating a promising photodynamic platform against tumors.


Asunto(s)
Neoplasias de la Mama , Células Madre Neoplásicas , Fototerapia , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Heparina de Bajo-Peso-Molecular , Humanos , Nanopartículas , Estudios Prospectivos
15.
ACS Appl Mater Interfaces ; 13(40): 47407-47417, 2021 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-34597015

RESUMEN

Pyroptosis is a programmed cell death to enhance immunogenicity of tumor cells, but pyroptosis-based immunotherapy is limited due to the immune escape involving myeloid-derived suppressor cells (MDSCs). Therefore, designing a nanoplatform to not only trigger apoptosis-pyroptosis transformation but also combat the MDSC-based immune escape is of great significance. As a proof-of-concept study, here, we designed a metal organic framework (MOF)-based nanoplatform to tailor the pyroptosis immunotherapy through disrupting the MDSC-mediated immunosuppression. By pH-responsive zeolitic imidazolate framework-8 (ZIF-8) modified with hyaluronic acid (HA), the chemotherapeutic drug mitoxantrone (MIT) and DNA demethylating agent hydralazine (HYD) were successfully co-encapsulated into ZIF-8 for achieving (M+H)@ZIF/HA nanoparticles. This nanoplatform demonstrated a powerful apoptosis-to-pyroptosis transformation with a potent disruption of MDSC-mediated T cell paralysis via reducing immunosuppressive methylglyoxal by HYD. Overall, our two-pronged nanoplatform (M+H)@ZIF/HA can switch the cold tumor into an arsenal of antigens that stimulate robust immunological responses, while suppressing immune escape, collectively triggering vigorous cytotoxic T cell responses with remarkable tumor elimination and building a long-term immune memory response against metastasis.


Asunto(s)
Antineoplásicos/uso terapéutico , Portadores de Fármacos/química , Factores Inmunológicos/uso terapéutico , Células Supresoras de Origen Mieloide/efectos de los fármacos , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Liberación de Fármacos , Femenino , Ácido Hialurónico/química , Hidralazina/química , Hidralazina/uso terapéutico , Imidazoles/química , Factores Inmunológicos/química , Inmunomodulación/efectos de los fármacos , Inmunoterapia/métodos , Estructuras Metalorgánicas/química , Ratones Endogámicos BALB C , Mitoxantrona/química , Mitoxantrona/uso terapéutico , Metástasis de la Neoplasia/prevención & control , Prueba de Estudio Conceptual , Piroptosis/efectos de los fármacos
16.
Chem Commun (Camb) ; 57(71): 8993-8996, 2021 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-34486606

RESUMEN

The oxidative phosphorylation inhibitor atovaquone (ATO) and the photosensitizer new indocyanine green (IR820) were self-assembled into carrier-free nanodrugs (IR820/ATO NPs) to achieve superior photothermal therapy (PTT), offering an attractive mitochondrial metabolism-regulatable approach for breast cancer treatment, where adenosine triphosphate (ATP) was downregulated along with downregulating the expression of heat shock proteins (HSPs) to amplify the sensitivity of PTT.


Asunto(s)
Antineoplásicos/farmacología , Atovacuona/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Verde de Indocianina/análogos & derivados , Nanopartículas/uso terapéutico , Fármacos Fotosensibilizantes/farmacología , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Atovacuona/química , Atovacuona/toxicidad , Línea Celular Tumoral , Femenino , Verde de Indocianina/química , Verde de Indocianina/farmacología , Verde de Indocianina/toxicidad , Ratones Endogámicos BALB C , Mitocondrias/efectos de los fármacos , Nanopartículas/química , Nanopartículas/toxicidad , Fosforilación Oxidativa/efectos de los fármacos , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/toxicidad , Terapia Fototérmica
17.
J Control Release ; 338: 33-45, 2021 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-34391837

RESUMEN

Interferon-γ (IFN-γ) plays contradictory roles in tumor immunology: (I) to activate positive host's immunity for eliminating tumor; (II) to induce negative adaptive immune resistance via up-regulating programmed death ligand-1 (PD-L1) expression for tumors to evade immune surveillance. The negative feedback loop between the IFN-γ recovery and the IFN-γ-induced PD-L1 up-regulation puts postoperative adjuvant chemotherapy into a dilemma. It is of great significance but challenging to manipulate the double-edge effects of IFN-γ against postoperative tumor progression. Herein, a platelet-engineered nanoplatform (PMF@DR NPs) capable of harmonizing janus-faced nature of IFN-γ was designed via uniquely co-assembling doxorubicin (Dox) and cyclin-dependent kinase 5 inhibitor roscovitine (Rosco) with platelet membrane fragment (PMF) as the particulate stabilizer. With PMF@DR NPs navigated by PMF to residual tumor, the Dox-activated immune response recovered IFN-γ secretion for positive host's immunity, while the IFN-γ-induced negative adaptive immune resistance was potently overcome by Rosco via disabling PD-L1 expression without dependence of IFN-γ stimulation. The negative feedback loop between IFN-γ recovery and PD-L1 up-regulation was thus potently disrupted in postoperative adjuvant chemotherapy. Our PMF@DR NPs not only harmonized janus-faced nature of IFN-γ to effectively regulate postoperative tumor progression, but also illustrated an innovative strategy for high-drug-loading biomimic nanoplatform.


Asunto(s)
Interferón gamma , Neoplasias Pulmonares , Línea Celular Tumoral , Humanos , Recurrencia Local de Neoplasia
18.
Nanoscale ; 13(27): 11953-11962, 2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-34212166

RESUMEN

Chlorin e6 (Ce6) is a widely utilized photosensitizer in photodynamic therapy (PDT) against tumor growth, but its hydrophobic feature and the hypoxia in the tumor microenvironment greatly compromise its therapeutic efficacy. To address the issues, here we designed a new Ce6 derivative (TCe6) by coupling Ce6 with amphiphilic d-α-tocopherol polyethylene glycol 1000 succinate (TPGS), endowing Ce6 with an excellent amphiphilic feature. In particular, the overall reactive oxygen species (ROS) generation by TCe6 was significantly enhanced because TPGS could interact with mitochondrial complex II to induce extra ROS production, amplifying the total ROS production under PDT. Inspired by the unique property of α-cyano-4-hydroxycinnamate (CHC) in regulating lactate metabolism to spare more intracellular oxygen for PDT, TCe6 was further co-assembled with CHC to construct TCe6/CHC nanoparticles (NPs) for addressing the insufficient oxygen issue in PDT. The as-prepared TCe6/CHC NPs not only increased the efficiency of cell internalization but also improved the solubility and stability of Ce6 and CHC. Thanks to the extra ROS production by the TPGS unit, the amphiphilic feature of TCe6 and the CHC-mediated hypoxia microenvironment, the TCe6/CHC NPs demonstrated excellent PDT against tumor growth. This work provided a versatile strategy to solve the current bottleneck in photosensitizer-based PDT, holding great promise for the design of advanced photodynamic nanoplatforms.


Asunto(s)
Nanopartículas , Fotoquimioterapia , Porfirinas , Línea Celular Tumoral , Clorofilidas , Lactatos , Fármacos Fotosensibilizantes/farmacología , Porfirinas/farmacología
19.
J Colloid Interface Sci ; 594: 493-501, 2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-33774405

RESUMEN

Theranostic nanoplatforms integrating simultaneously photodynamic therapy (PDT) and photothermal therapy (PTT) exhibit intrinsic advantages in tumor therapy due to distinct mechanisms of action. However, it is challenging to achieve PDT and PTT under single near-infrared (NIR) laser irradiation with a nanoplatform utilizing conventional organic photodynamic agent and inorganic photothermal agent owing to the difference in inherent excitation wavelengths. Particularly, the single NIR light (660 nm)-triggered PTT and PDT nanoplatform, constructed from chlorin e6 (Ce6) and copper sulfide (CuS) nanoparticles (NPs), has never been reported. Herein, we, for the first time, designed and established a dual-modal phototherapeutic nanoplatform that achieved both PTT and PDT under single NIR laser (660 nm) irradiation for Ce6 and CuS NPs with the strategy of core-shell structured CuS@Carbon integrated with Ce6. Introducing of carbon shell not only endows small CuS NPs with excellent tumor accumulation, but also significantly strengthens the photothermal performance of CuS NPs, realizing efficient photothermal performance under 660 nm laser irradiation. Moreover, Ce6 in carbon shell endowed the nanoplatform with photodynamic effect under 660 nm laser irradiation. The as-prepared Ce6/CuS@Carbon nanoplatform thus achieved dual-modal phototherapy under single NIR laser irradiation, significantly inhibiting tumor growth with minimal adverse effects and superior biosafety.


Asunto(s)
Nanopartículas , Neoplasias , Fotoquimioterapia , Línea Celular Tumoral , Humanos , Rayos Láser , Luz , Neoplasias/tratamiento farmacológico , Fototerapia
20.
Chem Commun (Camb) ; 57(18): 2305-2308, 2021 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-33533351

RESUMEN

A two-pronged concept combining photodynamic therapy (PDT) and epithelial-mesenchymal transition (EMT) blockade in a minimalist nanoplatform was proposed to combat basal-like breast cancer (BLBC) metastasis. Based on PDT-mediated tumor killing and epalrestat (Epa)-mediated EMT blockade, as-prepared Ce6/Epa nanoparticles prevented BLBC metastasis effectively in vivo, providing a very promising two-pronged strategy against BLBC metastasis.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Nanopartículas/administración & dosificación , Fotoquimioterapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Rodanina/análogos & derivados , Tiazolidinas/farmacología , Animales , Apoptosis , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Clorofilidas , Inhibidores Enzimáticos/farmacología , Transición Epitelial-Mesenquimal , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones SCID , Nanopartículas/química , Porfirinas/farmacología , Rodanina/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...