Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Colloids Surf B Biointerfaces ; 234: 113707, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38181689

RESUMEN

Activated M1-type macrophages, which produce inflammatory factors that exacerbate rheumatoid arthritis (RA), represent crucial target cells for inhibiting the disease process. In this study, we developed a novel photoresponsive targeted drug delivery system (TPNPs-HA) that can effectively deliver the hypoxia-activated prodrug tirapazamine (TPZ) specifically to activated macrophages. After administration, this metal-organic framework, PCN-224, constructed uing the photosensitizer porphyrin, exhibits the ability to generate excessive toxic reactive oxygen species (ROS) when exposed to near-infrared light. Additionally, the oxygen-consumed hypoxic environment further activates the chemotherapeutic effect of TPZ, thus creating a synergistic combination of photodynamic therapy (PDT) and hypoxia-activated chemotherapy (HaCT) to promote the elimination of activated M1-type macrophages. The results highlight the significantly potential of this photoresponsive nano-delivery system in providing substantial relief for RA. Furthermore, these findings support its effectiveness in inhibiting the disease process of RA, thereby offering new possibilities for the development of precise and accurate strategies for RA.


Asunto(s)
Artritis Reumatoide , Estructuras Metalorgánicas , Nanopartículas , Neoplasias , Fotoquimioterapia , Humanos , Tirapazamina/farmacología , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Hipoxia , Artritis Reumatoide/tratamiento farmacológico , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico
2.
J Colloid Interface Sci ; 659: 178-190, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38163404

RESUMEN

Microwave hyperthermia (MH) is an emerging treatment for solid tumors, such as breast cancer, due to its advantages of minimally invasive and deep tissue penetration. However, MH induced tumor hypoxia is still an obstacle to breast tumor treatment failure. Therefore, an original nanoengineering strategy was proposed to exacerbate hypoxia in two stages, thereby amplifying the efficiency of activating tirapazamine (TPZ). And a novel microwave-sensitized nanomaterial (GdEuMOF@TPZ, GEMT) is designed. GdEuMOF (GEM) nanoparticles are certified excellent microwave (MW) sensitization performance, thus improving tumor selectivity to achieve MH. Meanwhile MW can aggravate the generation of thrombus and caused local circulatory disturbance of tumor, resulting in the Stage I exacerbated hypoxia environment passively. Due to tumor heterogeneity and uneven hypoxia, GEMT nanoparticles under microwave could actively deplete residual oxygen through the chemical reaction, exacerbating hypoxia level more evenly, thus forming the Stage II of exacerbated hypoxia environment. Consequently, a two-stage exacerbated hypoxia GEMT nanoparticles realize amplifying activation of TPZ, significantly enhance the efficacy of microwave hyperthermia and chemotherapy, and effectively inhibit breast cancer. This research provides insights into the development of progressive nanoengineering strategies for effective breast tumor therapy.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Hipertermia Inducida , Neoplasias , Humanos , Femenino , Tirapazamina/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Microondas , Neoplasias/terapia , Hipoxia/terapia , Línea Celular Tumoral
3.
Microbiol Spectr ; 11(4): e0035223, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37306577

RESUMEN

Pseudomonas aeruginosa is the most common pathogen infecting cystic fibrosis (CF) lungs, causing acute and chronic infections. Intrinsic and acquired antibiotic resistance allow P. aeruginosa to colonize and persist despite antibiotic treatment, making new therapeutic approaches necessary. Combining high-throughput screening and drug repurposing is an effective way to develop new therapeutic uses for drugs. This study screened a drug library of 3,386 drugs, mostly FDA approved, to identify antimicrobials against P. aeruginosa under physicochemical conditions relevant to CF-infected lungs. Based on the antibacterial activity, assessed spectrophotometrically against the prototype RP73 strain and 10 other CF virulent strains, and the toxic potential evaluated toward CF IB3-1 bronchial epithelial cells, five potential hits were selected for further analysis: the anti-inflammatory and antioxidant ebselen, the anticancer drugs tirapazamine, carmofur, and 5-fluorouracil, and the antifungal tavaborole. A time-kill assay showed that ebselen has the potential to cause rapid and dose-dependent bactericidal activity. The antibiofilm activity was evaluated by viable cell count and crystal violet assays, revealing carmofur and 5-fluorouracil as the most active drugs in preventing biofilm formation regardless of the concentration. In contrast, tirapazamine and tavaborole were the only drugs actively dispersing preformed biofilms. Tavaborole was the most active drug against CF pathogens other than P. aeruginosa, especially against Burkholderia cepacia and Acinetobacter baumannii, while carmofur, ebselen, and tirapazamine were particularly active against Staphylococcus aureus and B. cepacia. Electron microscopy and propidium iodide uptake assay revealed that ebselen, carmofur, and tirapazamine significantly damage cell membranes, with leakage and cytoplasm loss, by increasing membrane permeability. IMPORTANCE Antibiotic resistance makes it urgent to design new strategies for treating pulmonary infections in CF patients. The repurposing approach accelerates drug discovery and development, as the drugs' general pharmacological, pharmacokinetic, and toxicological properties are already well known. In the present study, for the first time, a high-throughput compound library screening was performed under experimental conditions relevant to CF-infected lungs. Among 3,386 drugs screened, the clinically used drugs from outside infection treatment ebselen, tirapazamine, carmofur, 5-fluorouracil, and tavaborole showed, although to different extents, anti-P. aeruginosa activity against planktonic and biofilm cells and broad-spectrum activity against other CF pathogens at concentrations not toxic to bronchial epithelial cells. The mode-of-action studies revealed ebselen, carmofur, and tirapazamine targeted the cell membrane, increasing its permeability with subsequent cell lysis. These drugs are strong candidates for repurposing for treating CF lung P. aeruginosa infections.


Asunto(s)
Fibrosis Quística , Infecciones por Pseudomonas , Humanos , Pseudomonas aeruginosa , Fibrosis Quística/microbiología , Ensayos Analíticos de Alto Rendimiento , Reposicionamiento de Medicamentos , Tirapazamina/farmacología , Tirapazamina/uso terapéutico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Fluorouracilo , Biopelículas , Infecciones por Pseudomonas/microbiología
4.
Adv Sci (Weinh) ; 10(22): e2300899, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37156756

RESUMEN

As a currently common strategy to treat cancer, surgical resection may cause tumor recurrence and metastasis due to residual postoperative tumors. Herein, an implantable sandwich-structured dual-drug depot is developed to trigger a self-intensified starvation therapy and hypoxia-induced chemotherapy sequentially. The two outer layers are 3D-printed using a calcium-crosslinked mixture ink containing soy protein isolate, polyvinyl alcohol, sodium alginate, and combretastatin A4 phosphate (CA4P). The inner layer is one patch of poly (lactic-co-glycolic acid)-based electrospun fibers loaded with tirapazamine (TPZ). The preferentially released CA4P destroys the preexisting blood vessels and prevents neovascularization, which obstructs the external energy supply to cancer cells but aggravates hypoxic condition. The subsequently released TPZ is bioreduced to cytotoxic benzotriazinyl under hypoxia, further damaging DNA, generating reactive oxygen species, disrupting mitochondria, and downregulating hypoxia-inducible factor 1α, vascular endothelial growth factor, and matrix metalloproteinase 9. Together these processes induce apoptosis, block the intracellular energy supply, counteract the disadvantage of CA4P in favoring intratumor angiogenesis, and suppress tumor metastasis. The in vivo and in vitro results and the transcriptome analysis demonstrate that the postsurgical adjuvant treatment with the dual-drug-loaded sandwich-like implants efficiently inhibits tumor recurrence and metastasis, showing great potential for clinical translation.


Asunto(s)
Antineoplásicos , Recurrencia Local de Neoplasia , Humanos , Recurrencia Local de Neoplasia/prevención & control , Factor A de Crecimiento Endotelial Vascular , Línea Celular Tumoral , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Tirapazamina/farmacología , Hipoxia
5.
Int J Nanomedicine ; 17: 6257-6273, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36531117

RESUMEN

Purpose: Effective therapy for rheumatoid arthritis (RA) keeps a challenge due to the complex pathogenesis of RA. It is not enough to completely inhibit the process of RA with any single therapy method. The purpose of the research is to compensate for the insufficiency of monotherapy using multiple treatment regimens with different mechanisms. Material and Methods: In this study, we developed a new method to synthesize mesoporous silica nanoparticles hybridized with photosensitizer PCPDTBT (HNs). Branched polyethyleneimine-folic acid (PEI-FA) could be coated on the surface of HNs through electrostatic interactions. It simultaneously blocked the hypoxia-activated prodrug tirapazamine loaded into the mesopores and binded with Mcl-1 siRNA (siMcl-1) that interfered with the expression of the anti-apoptotic protein Mcl-1. Released from the co-delivery nanoparticles (PFHNs/TM) Tirapazamine and siMcl-1 upon exposure to acidic conditions of endosomes/lysosomes in activated macrophages. Under NIR irradiation, photothermal therapy and photodynamic therapy derived from PCPDTBT, hypoxia-activated chemotherapy derived from tirapazamine, and RNAi derived from siMcl-1 were used for the combined treatment for RA by killing activated macrophages. PEI-FA-coated PFHNs/TM exhibited activated macrophage-targeting characteristics, thereby enhancing the in vitro and in vivo NIR-induced combined treatment of RA. Results: The prepared PFHNs/TM have high blood compatibility (far below 5% of hemolysis) and ideal in vitro phototherapy effect while controlling the TPZ release and binding siMcl-1. We prove that PEI-FA-coated PFHNs/TM not only protect the bound siRNA but also are selectively uptaked by activated macrophages through FA receptor-ligand-mediated endocytosis, and effectively silence the target anti-apoptotic protein by siMcl-1 transfection. In vivo, PFHNs/TM have also been revealed to be selectively enriched at the inflammatory site of RA, exhibiting NIR-induced anti-RA efficacy. Conclusion: Overall, these FA-functionalized, pH-responsive PFHNs/TM represent a promising platform for the co-delivery of chemical drugs and nucleic acids for the treatment of RA cooperating with NIR-induced phototherapy.


Asunto(s)
Artritis Reumatoide , Nanopartículas , Humanos , Tirapazamina/farmacología , Interferencia de ARN , Sistema de Administración de Fármacos con Nanopartículas , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Fototerapia/métodos , Artritis Reumatoide/tratamiento farmacológico , ARN Interferente Pequeño , Ácido Fólico , Hipoxia
6.
J Photochem Photobiol B ; 234: 112535, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35930948

RESUMEN

The treatment efficacy of anticancer drugs in complex physiological environments is still restricted by multi-drug resistance. To overcome this issue, a nanodrug system of HA-SS@CuS@ZIF-8@TPZ&TBMACN (HSCZTT) that breaks through the detoxification barrier for tirapazamine (TPZ) delivery was developed in this manuscript. In addition to the photothermal effect aroused by CuS in HSCZTT, which can damage tumour cells, TBMACN with photostable fluorescence in the aggregate state can also generate sufficient reactive oxygen species (ROS) to destroy tumour cells. The continuous consumption of oxygen in PDT aggravates the hypoxic environment of tumours, which further activates the TPZ released in the acidic microenvironment of the tumour to achieve apoptosis of the tumour cells. The HSCZTT can not only target the CD44 receptor overexpressed on the surface of the cancer cell, but can also effectively consume a large amount of glutathione (GSH) through the disulphide bond-modified hyaluronic acid, which serves as a targeted disulphide bond, interfering with the detoxification barrier. Our finding presents a rational strategy to overcome multidrug resistance for the improved efficacy of anticancer drugs by the targeting of Hyaluronic acid (HA), release of the drug by the acid response of ZIF-8, breakthrough of the detoxification barrier, precise positioning of the drug release and combined treatment with phototherapy and hypoxia-activated chemotherapy.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Fotoquimioterapia , Antineoplásicos/química , Línea Celular Tumoral , Disulfuros , Humanos , Ácido Hialurónico/química , Hipoxia , Nanopartículas/química , Neoplasias/patología , Fármacos Fotosensibilizantes/química , Tirapazamina/química , Tirapazamina/metabolismo , Tirapazamina/farmacología , Microambiente Tumoral
7.
Chem Commun (Camb) ; 58(42): 6251-6254, 2022 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-35510707

RESUMEN

Nanosheet carriers loaded with drugs and phototherapeutics are used for effective cancer therapy, but the process remains challenging. Here, we prepared sulfur nanosheets (S-NSs) and then loaded tirapazamine (TPZ) and indocyanine green (ICG) with a loading efficiency of 6.3% and 94%, respectively. The obtained S-NSs-TPZ-ICG exhibits near-infrared (NIR) fluorescence, high 1O2 generation and photothermal conversion capabilities, good biocompatibility, and tumor microenvironment responsiveness. In vivo and in vitro experiments reveal that S-NSs-TPZ-ICG can be selectively decomposed under acidic and H2O2 conditions to release TPZ and ICG, and significantly inhibit tumor growth under laser irradiation without obvious toxic side effects.


Asunto(s)
Nanopartículas , Neoplasias , Línea Celular Tumoral , Humanos , Peróxido de Hidrógeno/farmacología , Verde de Indocianina/farmacología , Neoplasias/tratamiento farmacológico , Fototerapia , Azufre , Tirapazamina/farmacología , Tirapazamina/uso terapéutico , Microambiente Tumoral
8.
J Vasc Interv Radiol ; 33(8): 926-933.e1, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35504436

RESUMEN

PURPOSE: To investigate the safety of replacing doxorubicin with tirapazamine in conventional transarterial chemoembolization (TACE) in an Asian population with hepatocellular carcinoma (HCC), and to determine the optimal tirapazamine dose for phase II studies. MATERIALS AND METHODS: This was a phase I, 3 + 3 dose-escalation study for patients with unresectable early- and intermediate-stage HCC who received 5, 10, or 20 mg/m2 of intra-arterial (IA) tirapazamine followed by ethiodized oil/gelatin sponge-based embolization. Key eligibilities included HCCs no more than 10 cm in diameter, prior embolization allowed, Eastern Cooperative Oncology Group performance status of 0 or 1, Child-Pugh score of 5-7, and platelet count of ≥60,000 µL. Dose-limiting toxicity (DLT) was defined as any grade 3 nonhematological or grade 4 hematological toxicity, with the exception of transient elevation of aminotransferase levels after the procedure. RESULTS: Seventeen patients were enrolled, 59% of whom had progression from a prior HCC therapy and 35% of whom had progression or recurrence after TACE. All patients tolerated the tirapazamine TACE well without any DLT or serious adverse event. Using the modified Response Evaluation Criteria in Solid Tumors, the complete response (CR) rate was 47%, and the CR + partial response rate was 65%. The median duration of response was not reached. The median time to progression was 12.6 months (95% confidence interval, 5.1-not reached). The median overall survival was 29.3 months. The selected phase II dose was set at a fixed dose of 35 mg of IA tirapazamine. CONCLUSIONS: IA tirapazamine with transarterial embolization was well tolerated and showed promising efficacy signals in intermediate-stage HCC, justifying pursuit of a phase II study.


Asunto(s)
Carcinoma Hepatocelular , Quimioembolización Terapéutica , Neoplasias Hepáticas , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/terapia , Quimioembolización Terapéutica/efectos adversos , Quimioembolización Terapéutica/métodos , Aceite Etiodizado , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/terapia , Tirapazamina/efectos adversos , Resultado del Tratamiento
9.
Drug Deliv ; 29(1): 238-253, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35001784

RESUMEN

Photodynamic therapy (PDT) has been applied in cancer treatment by utilizing reactive oxygen species (ROS) to kill cancer cells. However, the effectiveness of PDT is greatly reduced due to local hypoxia. Hypoxic activated chemotherapy combined with PDT is expected to be a novel strategy to enhance anti-cancer therapy. Herein, a novel liposome (LCT) incorporated with photosensitizer (PS) and bioreductive prodrugs was developed for PDT-activated chemotherapy. In the design, CyI, an iodinated cyanine dye, which could simultaneously generate enhanced ROS and heat than other commonly used cyanine dyes, was loaded into the lipid bilayer; while tirapazamine (TPZ), a hypoxia-activated prodrug was encapsulated in the hydrophilic nucleus. Upon appropriate near-infrared (NIR) irradiation, CyI could simultaneously produce ROS and heat for synergistic PDT and photothermal therapy (PTT), as well as provide fluorescence signals for precise real-time imaging. Meanwhile, the continuous consumption of oxygen would result in a hypoxia microenvironment, further activating TPZ free radicals for chemotherapy, which could induce DNA double-strand breakage and chromosome aberration. Moreover, the prepared LCT could stimulate acute immune response through PDT activation, leading to synergistic PDT/PTT/chemo/immunotherapy to kill cancer cells and reduce tumor metastasis. Both in vitro and in vivo results demonstrated improved anticancer efficacy of LCT compared with traditional PDT or chemotherapy. It is expected that these iodinated cyanine dyes-based liposomes will provide a powerful and versatile theranostic strategy for tumor target phototherapy and PDT-induced chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Hipoxia/patología , Sistema de Administración de Fármacos con Nanopartículas/química , Fármacos Fotosensibilizantes/farmacología , Fototerapia/métodos , Tirapazamina/farmacología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Supervivencia Celular/efectos de los fármacos , Química Farmacéutica , Aberraciones Cromosómicas/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Portadores de Fármacos/química , Liberación de Fármacos , Liposomas/química , Ratones , Ratones Endogámicos BALB C , Tamaño de la Partícula , Fármacos Fotosensibilizantes/administración & dosificación , Fármacos Fotosensibilizantes/farmacocinética , Especies Reactivas de Oxígeno/metabolismo , Propiedades de Superficie , Tirapazamina/administración & dosificación , Tirapazamina/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Mater Chem B ; 9(44): 9142-9152, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34693960

RESUMEN

Multimodal synergistic therapy has gained increasing attention in cancer treatment to overcome the limitations of monotherapy and achieve high anticancer efficacy. In this study, a synergistic phototherapy and hypoxia-activated chemotherapy nanoplatform based on natural melanin nanoparticles (MPs) loaded with the bioreduction prodrug tirapazamine (TPZ) and decorated with hyaluronic acid (HA) was developed. A self-reporting aggregation-induced emission (AIE)-active photosensitizer (PS) (BATTMN) was linked to the prepared nanoparticles by boronate ester bonds. The MPs and BATTMN-HA played roles as quenchers for PS and cancer targeting/photodynamic moieties, respectively. As a pH sensitive bond, the borate ester bonds between HA and BATTMN are hydrolysed in the acidic cancer environment, thereby separating BATTMN from the nanoparticles and leading to the induction of fluorescence for imaging-guided synergistic phototherapy/hypoxia-activated chemotherapy under dual irradiation. TPZ can be released upon activation by pH, near-infrared (NIR) and hyaluronidase (Hyal). Particularly, the hypoxia-dependent cytotoxicity of TPZ was amplified by oxygen consumption in the tumor intracellular environment induced by the AIE-active PS in photodynamic therapy (PDT). The nanoparticles developed in our research showed favorable photothermal conversion efficiency (η = 37%), desired cytocompatibility, and excellent synergistic therapeutic efficacy. The proposed nanoplatform not only extends the application scope of melanin materials with AIE-active PSs, but also offers useful insights into developing multistimulus as well as multimodal synergistic tumor treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Portadores de Fármacos/química , Melaninas/uso terapéutico , Nanopartículas/uso terapéutico , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/uso terapéutico , Animales , Antineoplásicos/química , Ácidos Borónicos/química , Ácidos Borónicos/efectos de la radiación , Ácidos Borónicos/uso terapéutico , Terapia Combinada , Quimioterapia , Femenino , Humanos , Células MCF-7 , Melaninas/química , Melaninas/efectos de la radiación , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Nanopartículas/efectos de la radiación , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/efectos de la radiación , Terapia Fototérmica , Profármacos/química , Profármacos/uso terapéutico , Tirapazamina/química , Tirapazamina/uso terapéutico , Hipoxia Tumoral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Mater Chem B ; 9(26): 5318-5328, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34231629

RESUMEN

For cancer treatment, the traditional monotherapy has the problems of low drug utilization rate, poor efficacy and easy recurrence of the cancer. Herein, nanoparticles (NPs) based on a novel semiconducting molecule (ITTC) are developed with excellent photostability, high photothermal conversion efficiency and good 1O2 generation ability. The chemotherapy of the hypoxia-activated prodrug tirapazamine (TPZ) was improved accordingly after oxygen consumption by the photodynamic therapy of ITTC NPs. Additionally, the metabolic process of ITTC NPs in vivo could be monitored in real time for fluorescence imaging guided phototherapy, which presented great passive targeting ability to the tumor site. Remarkably, both in vitro and in vivo experiments demonstrated that the combination of ITTC NPs and TPZ presented excellent synergistic tumor ablation through photothermal therapy, photodynamic therapy and hypoxia-activated chemotherapy with great potential for clinical applications in the future.


Asunto(s)
Antineoplásicos/farmacología , Hipoxia/diagnóstico por imagen , Hipoxia/tratamiento farmacológico , Nanopartículas/química , Imagen Óptica , Fármacos Fotosensibilizantes/farmacología , Tirapazamina/farmacología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estructura Molecular , Nanopartículas/administración & dosificación , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/tratamiento farmacológico , Fármacos Fotosensibilizantes/administración & dosificación , Fármacos Fotosensibilizantes/química , Semiconductores , Tirapazamina/administración & dosificación , Tirapazamina/química
12.
J Nanobiotechnology ; 19(1): 89, 2021 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-33781277

RESUMEN

BACKGROUND: Areas of hypoxia are often found in triple-negative breast cancer (TNBC), it is thus more difficult to treat than other types of breast cancer, and may require combination therapies. A new strategy that combined bioreductive therapy with photodynamic therapy (PDT) was developed herein to improve the efficacy of cancer treatment. Our design utilized the characteristics of protoporphyrin IX (PpIX) molecules that reacted and consumed O2 at the tumor site, which led to the production of cytotoxic reactive oxygen species (ROS). The low microenvironmental oxygen levels enabled activation of a bioreductive prodrug, tirapazamine (TPZ), to become a toxic radical. The TPZ radical not only eradicated hypoxic tumor cells, but it also promoted therapeutic efficacy of PDT. RESULTS: To achieve the co-delivery of PpIX and TPZ for advanced breast cancer therapy, thin-shell hollow mesoporous Ia3d silica nanoparticles, designated as MMT-2, was employed herein. This nanocarrier designed to target the human breast cancer cell MDA-MB-231 was functionalized with PpIX and DNA aptamer (LXL-1), and loaded with TPZ, resulting in the formation of TPZ@LXL-1-PpIX-MMT-2 nanoVector. A series of studies confirmed that our nanoVectors (TPZ@LXL-1-PpIX-MMT-2) facilitated in vitro and in vivo targeting, and significantly reduced tumor volume in a xenograft mouse model. Histological analysis also revealed that this nanoVector killed tumor cells in hypoxic regions efficiently. CONCLUSIONS: Taken together, the synergism and efficacy of this new therapeutic design was confirmed. Therefore, we concluded that this new therapeutic strategy, which exploited a complementary combination of PpIX and TPZ, functioned well in both normoxia and hypoxia, and is a promising medical procedure for effective treatment of TNBC.


Asunto(s)
Antineoplásicos/farmacología , Nanopartículas/uso terapéutico , Fotoquimioterapia/métodos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Aptámeros de Nucleótidos , Línea Celular Tumoral , Terapia Combinada , Femenino , Humanos , Ratones , Oxígeno , Profármacos , Especies Reactivas de Oxígeno , Dióxido de Silicio , Tirapazamina , Carga Tumoral , Hipoxia Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Colloids Surf B Biointerfaces ; 200: 111591, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33548893

RESUMEN

Radiotherapy (RT) is becoming a pervasive therapeutic pattern in clinical cancer therapy. However, the hypoxic microenvironment of tumors has a strong resistance to radiotherapy and could lead to a potential recurrence and metastasis after the treatment. Therefore, the use of synergistic strategies for improving and supplementing the RT efficiency is important. Herein, a novel Bi2S3/alginate (ALG) hydrogel containing tirapazamine (TPZ) was designed for the effective suppression of tumor recurrence, by introducing Bi3+ into the ALG, Na2S and TPZ solution. In this formulation, Bi3+ was used to crosslink with the ALG to form the hydrogel and react with S2- to simultaneously form Bi2S3 nanoparticles in the hydrogel matrix. The resulting Bi2S3 nanoparticles not only exhibit the superb radiosensitization effect to boost the effective eradication of tumors during RT but also manifest an excellent photothermal transforming performance for tumor hyperthermia and computed tomography (CT) imaging capacity for tumor monitoring. Furthermore, the RT caused hypoxia could activate the reductive prodrug TPZ and enhance its therapeutic efficiency. The reported hydrogel system provides an efficient and safe therapeutic strategy for current local tumor therapy.


Asunto(s)
Hipertermia Inducida , Neoplasias , Profármacos , Línea Celular Tumoral , Humanos , Hidrogeles , Neoplasias/tratamiento farmacológico , Tirapazamina , Microambiente Tumoral
14.
Chem Commun (Camb) ; 56(69): 9978-9981, 2020 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-32851998

RESUMEN

A tumor redox-activatable micellar nanoplatform based on the naturally occurring biomacromolecule hyaluronic acid (HA) was developed for complementary photodynamic/chemotherapy against CD44-positive tumors. Here HA was first conjugated with l-carnitine (Lc)-modified zinc phthalocyanine (ZnPc) via disulfide linkage and then co-assembled with tirapazamine (TPZ) to afford the physiologically stable micellar nanostructure. The mitochondria-targeted photodynamic activity of ZnPc-Lc could efficiently activate the mitochondrial apoptosis cascade and deplete the oxygen in the tumor intracellular environment to amplify the hypoxia-dependent cytotoxic effect of TPZ.


Asunto(s)
Biopolímeros/química , Micelas , Mitocondrias/metabolismo , Nanoestructuras/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Carnitina/química , Línea Celular Tumoral , Humanos , Ácido Hialurónico/química , Indoles/química , Rayos Infrarrojos , Isoindoles , Ratones , Mitocondrias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Compuestos Organometálicos/química , Oxidación-Reducción , Fotoquimioterapia , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Tirapazamina/química , Tirapazamina/farmacología , Tirapazamina/uso terapéutico , Trasplante Heterólogo , Compuestos de Zinc
15.
Biomater Sci ; 8(11): 3116-3129, 2020 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-32352102

RESUMEN

To enhance the specificity and efficiency of anti-tumor therapies, we have designed a multifunctional nanoparticle platform for photochemotherapy using fluorescence (FL) and photoacoustic (PA) imaging guidance. Nanoparticles (NPs) composed of a eutectic mixture of natural fatty acids that undergo a solid-liquid phase transition at 39 °C were used to encapsulate materials for the rapid and uniform release of the hypoxia-activated prodrug tirapazamine (TPZ) and the photosensitizer IR780, which targets the mitochondria of tumor cells and can be used to induce hypoxic cell death via photodynamic therapy and photothermal therapy. In vitro, the NPs containing TPZ and IR7890 exhibited appreciable cell uptake and triggered drug release when irradiated with a NIR laser. In vivo, photochemotherapy of the NPs achieved the best anti-tumor efficacy under PA and FL imaging guidance and monitoring. By combining IR780 mitochondria-targeting phototherapy with TPZ, we observed improved anti-tumor effectiveness and this has the potential to reduce the side effects of traditional chemotherapy. Herein, we demonstrate a new intracellular photochemotherapy nanosystem that co-encapsulates photosensitizers and hypoxia-activated drugs to enhance the overall anti-tumor effect precisely and efficiently.


Asunto(s)
Antineoplásicos/administración & dosificación , Indoles/administración & dosificación , Nanopartículas/administración & dosificación , Neoplasias/tratamiento farmacológico , Fotoquimioterapia , Fármacos Fotosensibilizantes/administración & dosificación , Profármacos/administración & dosificación , Tirapazamina/administración & dosificación , Animales , Antineoplásicos/química , Antineoplásicos/efectos de la radiación , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Femenino , Indoles/química , Indoles/efectos de la radiación , Rayos Láser , Ratones Endogámicos BALB C , Nanopartículas/química , Nanopartículas/efectos de la radiación , Neoplasias/metabolismo , Neoplasias/patología , Imagen Óptica , Técnicas Fotoacústicas , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/efectos de la radiación , Profármacos/química , Profármacos/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , Tirapazamina/química , Tirapazamina/efectos de la radiación
16.
Int J Radiat Biol ; 95(12): 1708-1717, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31545117

RESUMEN

Purpose: To evaluate the usefulness of combined treatment with both continuous administration of a hypoxic cytotoxin, tirapazamine (TPZ) and mild temperature hyperthermia (MTH) in boron neutron capture therapy (BNCT) in terms of local tumor response and lung metastatic potential, referring to the response of intratumor quiescent (Q) cells.Materials and methods: B16-BL6 melanoma tumor-bearing C57BL/6 mice were continuously given 5-bromo-2'-deoxyuridine (BrdU) to label all proliferating (P) cells. The tumors received reactor thermal neutron beam irradiation following the administration of a 10B-carrier (L-para-boronophenylalanine-10B (BPA) or sodium mercaptoundecahydrododecaborate-10B (BSH)) after single intraperitoneal injection of an acute hypoxia-releasing agent (nicotinamide), MTH (40 °C for 60 min), and 24-h continuous subcutaneous infusion of TPZ or combined treatment with both TPZ and MTH. Immediately after irradiation, cells from some tumors were isolated and incubated with a cytokinesis blocker. The responses of the Q and total (=P + Q) tumor cell populations were assessed based on the frequency of micronuclei using immunofluorescence staining for BrdU. In other tumor-bearing mice, 17 days after irradiation, macroscopic lung metastases were enumerated.Results: BPA-BNCT increased the sensitivity of the total tumor cell population more than BSH-BNCT. However, the sensitivity of Q cells treated with BPA was lower than that of BSH-treated Q cells. With or without a 10B-carrier, combination with continuously administered TPZ with or without MTH enhanced the sensitivity of the both total and Q cells, especially Q cells. Even without irradiation, nicotinamide treatment decreased the number of lung metastases. With irradiation, BPA-BNCT, especially in combination with combined treatment with both TPZ and MTH as well as nicotinamide treatment, showed the potential to reduce the number more than BSH-BNCT.Conclusion: BSH-BNCT combined with TPZ with or without MTH improved local tumor control, while BPA-BNCT in combination with both TPZ and MTH as well as nicotinamide is thought to reduce the number of lung metastases. It was elucidated that control of the chronic hypoxia-rich Q cell population in the primary solid tumor has the potential to impact the control of local tumors as a whole and that control of the acute hypoxia-rich total tumor cell population in the primary solid tumor has the potential to impact the control of lung metastases.


Asunto(s)
Terapia por Captura de Neutrón de Boro , Hipertermia Inducida , Neoplasias Pulmonares/secundario , Melanoma/patología , Tirapazamina/farmacología , Hipoxia Tumoral/efectos de los fármacos , Hipoxia Tumoral/efectos de la radiación , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Terapia Combinada , Melanoma/tratamiento farmacológico , Melanoma/radioterapia , Ratones , Tirapazamina/administración & dosificación , Tirapazamina/uso terapéutico , Resultado del Tratamiento
17.
ACS Appl Mater Interfaces ; 11(38): 34755-34765, 2019 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-31474108

RESUMEN

Radiation dosage constraints and hypoxia-associated resistance lead to the failure of radiotherapy (RT), especially in hypoxic liver cancer. Therefore, the intricate use of combined strategies for potentiating and complementing RT is especially important. In this work, we fabricated multifunctional Janus-structured gold triangle-mesoporous silica nanoparticles (NPs) as multifunctional platforms to deliver the hypoxia-activated prodrug tirapazamine (TPZ) for extrinsic radiosensitization, local photothermal therapy, and hypoxia-specific chemotherapy. The subsequent conjugation of folic acid-linked poly(ethylene glycol) provided the Janus nanoplatforms with liver cancer targeting and minimized opsonization properties. In vitro and in vivo experiments revealed the combined radiosensitive and photothermal antitumor effects of the Janus nanoplatforms. Importantly, the TPZ-loaded Janus nanoplatforms exhibited pH-responsive release behavior, which effectively improved the cellular internalization and therapeutic efficiency in hypoxic rather than normoxic liver cancer cells. Hypoxia-specific chemotherapy supplemented the ineffectiveness of radio-photothermal therapy in hypoxic tumor tissues, resulting in remarkable tumor growth inhibition without systematic toxicity. Therefore, our Janus nanoplatforms integrated radio-chemo-photothermal therapy in a hypoxia-activated manner, providing an efficient and safe strategy for treating liver cancer.


Asunto(s)
Quimioradioterapia , Sistemas de Liberación de Medicamentos , Oro , Hipertermia Inducida , Neoplasias Hepáticas Experimentales , Fototerapia , Profármacos , Dióxido de Silicio , Tirapazamina , Animales , Hipoxia de la Célula , Línea Celular Tumoral , Oro/química , Oro/farmacología , Humanos , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Neoplasias Hepáticas Experimentales/terapia , Ratones , Ratones Desnudos , Nanopartículas/química , Nanopartículas/uso terapéutico , Porosidad , Profármacos/química , Profármacos/farmacología , Dióxido de Silicio/química , Dióxido de Silicio/farmacología , Tirapazamina/química , Tirapazamina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Int J Nanomedicine ; 14: 4541-4558, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31417257

RESUMEN

Background: Tumor metastasis is responsible for most cancer death worldwide, which lacks curative treatment. Purpose: The objective of this study was to eliminate tumor and control the development of tumor metastasis. Methods: Herein, we demonstrated a smart nano-enabled platform, in which 2-[2-[2-chloro-3-[(1,3-dihydro-3,3-dimethyl-1-propyl-2h-indol-2-ylidene)ethylidene]-1-cyclohexen-1-yl]ethenyl]-3,3-dimethyl-1-propylindolium iodide (IR780) and tirapazamine (TPZ) were co-loaded in poly(ε-caprolactone)-poly(ethylene glycol) (PEG-PCL) to form versatile nanoparticles (PEG-PCL-IR780-TPZ NPs). Results: The intelligence of the system was reflected in the triggered and controlled engineering. Specially, PEG-PCL not only prolonged the circulation time of IR780 and TPZ but also promoted tumor accumulation of nanodrugs through enhanced permeability and retention (EPR) effect. Moreover, reactive oxygen species (ROS) generated by IR780 armed by an 808 nm laser irradiation evoked a cargo release. Meanwhile, IR780, as a mitochondria-targeting phototherapy agent exacerbated tumor hypoxic microenvironment and activated TPZ for accomplishing hypoxia-activated chemotherapy. Most significantly, IR780 was capable of triggering immunogenic cell death (ICD) during the synergic treatment. ICD biomarkers as a "danger signal" accelerated dendritic cells (DCs) maturation, and subsequently activated toxic T lymphocytes. Conclusion: Eventually, antitumor immune responses stimulated by combinational phototherapy and hypoxia-activated chemotherapy revolutionized the current landscape of cancer treatment, strikingly inhibiting tumor metastasis and providing a promising prospect in the clinical application.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Fototerapia , Animales , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Inmunoterapia , Indoles/uso terapéutico , Liposomas , Ratones Endogámicos BALB C , Nanopartículas/química , Nanopartículas/ultraestructura , Fotoquimioterapia , Fototerapia/métodos , Polietilenglicoles/química , Especies Reactivas de Oxígeno/metabolismo , Temperatura , Tirapazamina/farmacología , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
19.
Biomaterials ; 221: 119422, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31437723

RESUMEN

Highly specific and effective cancer phototherapy remains as a great challenge. Herein, a smart nanoplatform (TENAB NP) sequentially responsive to light, low pH and hypoxia is demonstrated for multi-mode imaging guided synergistic cancer therapy with negligible skin phototoxicity. Upon 808-nm laser irradiation, TENAB NPs can generate hyperthermia to melt the phase change material (PCM-LASA) coat and thereafter release chemo-drug tirapazamine (TPZ). Meanwhile, under acidic pH, photosensitizer ENAB would turn "off" its charge-transfer state, generating prominent 1O2 for photodynamic therapy (PDT) and heat for photothermal therapy (PTT), respectively. Accompanied with PDT-induced hypoxia, the released TPZ can be activated into its cytotoxic form for tumor cells killing. Notably, owing to phase change material LASA coat and ENAB's pH sensitivity, TENAB NPs show negligible photosensitization to skin and normal tissues. As the multi-stimuli responsive mechanism, TENAB NPs demonstrate a promising future in cancer photo-chemo theranostics with excellent skin protection.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Fotoquimioterapia/métodos , Animales , Alcoholes Grasos/química , Femenino , Células HeLa , Humanos , Ácido Linoleico/química , Ratones , Ratones Desnudos , Microscopía Confocal , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/uso terapéutico , Piel/efectos de los fármacos , Piel/metabolismo , Piel/efectos de la radiación , Tirapazamina/uso terapéutico
20.
Mol Pharm ; 16(5): 2172-2183, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-30978027

RESUMEN

Although photodynamic therapy (PDT) has been an attractive strategy for several cancer treatments in the clinical setting, PDT efficacy is attenuated by consumption of oxygen. To address this photodynamic issue, we adopted a phototherapy-chemotherapy combination strategy based on targeted delivery of the near-infrared photosensitizer indocyanine green (ICG), photothermal conversion agent polydopamine (PDA), and tirapazamine (TPZ), a hypoxia-activated prodrug. Under laser irradiation, ICG consumption of oxygen and aggravated hypoxia in tumor sites can activate TPZ to damage DNA. In parallel, ICG produces reactive oxygen species which work in synergy with PDA to enhance phototherapeutic efficiency. Herein, hybrid CaCO3/TPGS nanoparticles delivering ICG, PDA, and TPZ (ICG-PDA-TPZ NPs) were designed for effective and safe cancer therapy. ICG-PDA-TPZ NPs showed significantly improved cellular uptake and accumulation in tumors. Furthermore, we demonstrated that ICG-PDA-TPZ NPs showed intensive photodynamic and photothermal effects in vitro and in vivo, which synergized with TPZ in subcutaneous U87 malignant glioma growth and orthotopic B16F10 tumor inhibition, with negligible side effects. Thus, ICG-PDA-TPZ NPs could be an effective strategy for improvement of PDT.


Asunto(s)
Hipertermia Inducida , Verde de Indocianina , Indoles , Nanopartículas , Neoplasias , Fotoquimioterapia , Profármacos , Fármacos Sensibilizantes a Radiaciones , Tirapazamina , Animales , Humanos , Ratones , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Hipertermia Inducida/métodos , Verde de Indocianina/metabolismo , Verde de Indocianina/uso terapéutico , Indoles/metabolismo , Indoles/uso terapéutico , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Fotoquimioterapia/efectos adversos , Fotoquimioterapia/métodos , Polímeros/metabolismo , Polímeros/uso terapéutico , Profármacos/metabolismo , Profármacos/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/metabolismo , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Especies Reactivas de Oxígeno/efectos de la radiación , Tirapazamina/metabolismo , Tirapazamina/uso terapéutico , Distribución Tisular , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA