Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Nanobiotechnology ; 22(1): 615, 2024 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-39385196

RESUMEN

Focused ultrasound ablation surgery (FUAS) is a minimally invasive treatment option that has been utilized in various tumors. However, its clinical advancement has been hindered by issues such as low safety and efficiency, single image guidance mode, and postoperative tumor residue. To address these limitations, this study aimed to develop a novel multi-functional gas-producing engineering bacteria biological targeting cooperative system. Pulse-focused ultrasound (PFUS) could adjust the ratio of thermal effect to non-thermal effect by adjusting the duty cycle, and improve the safety and effectiveness of treatment.The genetic modification of Escherichia coli (E.coli) involved the insertion of an acoustic reporter gene to encode gas vesicles (GVs), resulting in gas-producing E.coli (GVs-E.coli) capable of targeting tumor anoxia. GVs-E.coli colonized and proliferated within the tumor while the GVs facilitated ultrasound imaging and cooperative PFUS. Additionally, multifunctional cationic polyethyleneimine (PEI)-poly (lactic-co-glycolic acid) (PLGA) nanoparticles (PEI-PLGA/EPI/PFH@Fe3O4) containing superparamagnetic iron oxide (SPIO, Fe3O4), perfluorohexane (PFH), and epirubicin (EPI) were developed. These nanoparticles offered synergistic PFUS, supplementary chemotherapy, and multimodal imaging capabilities.GVs-E.coli effectively directed the PEI-PLGA/EPI/PFH@Fe3O4 to accumulate within the tumor target area by means of electrostatic adsorption, resulting in a synergistic therapeutic impact on tumor eradication.In conclusion, GVs-E.coli-mediated multi-functional nanoparticles can synergize with PFUS and chemotherapy to effectively treat tumors, overcoming the limitations of current FUAS therapy and improving safety and efficacy. This approach presents a promising new strategy for tumor therapy.


Asunto(s)
Escherichia coli , Imagen Multimodal , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Animales , Escherichia coli/efectos de los fármacos , Ratones , Imagen Multimodal/métodos , Línea Celular Tumoral , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Imagen por Resonancia Magnética/métodos , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Fluorocarburos/química , Polietileneimina/química , Humanos , Ingeniería Genética/métodos , Ratones Endogámicos BALB C , Técnicas Fotoacústicas/métodos , Femenino , Nanopartículas/química , Epirrubicina/farmacología , Epirrubicina/uso terapéutico , Epirrubicina/química , Ácido Poliglicólico/química , Ácido Láctico/química , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos
2.
RSC Adv ; 13(28): 19065-19078, 2023 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-37362333

RESUMEN

Objective: To make up for the insufficient ultrasound ablation of tumors, the energy output or synergist is increased but faces the big challenge of normal tissue damage. In this study, we report a tumor-homing bacterium, Bifidobacterium bifidum (B. bifidum), adsorbing liposomes that encapsulate perfluorohexane (PFH) and doxorubicin (DOX) to enhance the pulsed-focused ultrasound (PFUS) for tumor therapy, so as to improve the efficacy, safety and controllability of ultrasound treatment. Methods: The PFH and DOX co-loaded cationic liposomal nanoparticles (CL-PFH-DOX-NPs) were prepared for ultrasound (US) imaging, cell-killing, and B. bifidum adsorption for the reactive oxygen species (ROS) testing. The aggregation of B. bifidum and CL-PFH-DOX-NPs is called tumor-homing aggregation (B. bifidum@CL-PFH-DOX-NPs) in this study, and the synergistic effects of B. bifidum@CL-PFH-DOX-NPs were analyzed in vivo. Results: Comprehensive studies validated that CL-PFH-DOX-NPs can enhance US imaging and cell-killing and B. bifidum can promote ROS, and B. bifidum@CL-PFH-DOX-NPs achieve PFUS synergism in vivo. Importantly, active homing of B. bifidum facilitated the delivery and retention of CL-PFH-DOX-NPs in tumors, reducing dispersion in normal tissues, achieving the targeting ability of B. bifidum@CL-PFH-DOX-NPs. The best sonication time was chosen according to the distribution of CL-PFH-DOX-NPs in vivo to achieve efficient therapy. Especially, B. bifidum@CL-PFH-DOX-NPs amplified cavitation and the immune-boosting effects. Conclusion: Multifunctional B. bifidum@CL-PFH-DOX-NPs were successfully constructed with well targeting, which not only realized US imaging monitoring, strong cavitation and complementary killing during PFUS, but also achieved immunity enhancement after PFUS. The combination of PFUS, B. bifidum and CL-PFH-DOX-NPs provides a new idea for the potential application of ultrasound therapy in solid tumors.

3.
J Nanobiotechnology ; 21(1): 142, 2023 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-37120558

RESUMEN

Focused ultrasound ablation surgery (FUAS) is a novel therapy with a wide range of potential applications. However, synergists are crucial to the therapy process due to the ultrasonic energy's attenuation properties. As a result of the complex hypoxic environment in the tumor area and many factors, the existing synergists have limitations such as weak targeting, single imaging mode, and easy tumor recurrence after treatment. Because of the above deficiencies, this study intends to construct bio-targeted oxygen production probes consisting of Bifidobacterium that naturally target the hypoxia region of the tumor and multi-functional oxygen-producing nanoparticles equipped with IR780, perfluorohexane (PFH), CBP (carboplatin), and oxygen. The probes are expected to achieve targeted and synergistic FUAS therapy and dual-mode imaging to mediate tumor diagnosis and treatment. The oxygen and drugs carried in it are accurately released after FUAS stimulation, which is expected to alleviate tumor hypoxia, avoid tumor drug resistance, improve the effect of chemotherapy, and realize FUAS combined with chemotherapy antitumor therapy. This strategy is expected to make up for the deficiencies of existing synergists, improve the effectiveness and safety of treatment, and provide the foundation for future tumor therapy progress.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Diagnóstico por Imagen , Nanopartículas/uso terapéutico , Oxígeno , Línea Celular Tumoral
4.
Acta Biomater ; 150: 337-352, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35931281

RESUMEN

Focused ultrasonic ablation surgery (FUAS) for tumor treatment has emerged as an effective non-invasive therapeutic approach, but its widespread clinical utilization is limited by its low therapeutic efficiency caused by inadequate tumor targeting, single imaging modality, and possible tumor recurrence following surgery. Therefore, this study aimed to develop a biological targeting synergistic system consisting of genetically engineered bacteria and multi-functional nanoparticles to overcome these limitations. Escherichia coli was genetically modified to carry an acoustic reporter gene encoding the formation of gas vesicles (GVs) and then target the tumor hypoxic environment in mice. After E. coli producing GVs (GVs-E. coli) colonized the tumor target area, ultrasound imaging and collaborative FUAS were performed; multi-functional nanoparticles were then enriched in the tumor target area through electrostatic adsorption. Multi-functional cationic lipid nanoparticles containing IR780, perfluorohexane, and banoxantrone dihydrochloride (AQ4N) were coloaded in the tumor to realize targeted multimodal imaging and enhance the curative effect of FUAS. AQ4N was stimulated by the tumor hypoxic environment and synergistically cooperated with FUAS to kill tumor cells. In sum, synergistic tumor therapy involving multi-functional nanoparticles mediated by genetically engineered bacteria overcomes the limitations and improves the curative effect of existing FUAS. STATEMENT OF SIGNIFICANCE: Inadequate tumor targeting, single image monitoring mode, and prone tumor recurrence following surgery remain significant challenges yet critical for tumor therapy. This study proposes a strategy for genetically engineered bacteria-mediated multifunctional nanoparticles for synergistic tumor therapy. The multifunctional genetically engineered biological targeting synergistic agent can accomplish tumor-targeting therapy, synergistic FUAS ablation, hypoxia-activated chemotherapy combined with FUAS ablation, and multiple-imaging guidance and monitoring all at the same time, thereby compensating for the shortcomings of FUAS treatment. This strategy could pave the way for the progress of tumor therapy.


Asunto(s)
Nanopartículas , Neoplasias , Animales , Línea Celular Tumoral , Escherichia coli , Liposomas , Ratones , Nanopartículas/uso terapéutico , Recurrencia Local de Neoplasia , Neoplasias/terapia
5.
Int J Nanomedicine ; 17: 2943-2960, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35814614

RESUMEN

Purpose: Breast cancer is detrimental to the health of women due to the difficulty of early diagnosis and unsatisfactory therapeutic efficacy of available breast cancer therapies. High intensity focused ultrasound (HIFU) ablation is a new method for the treatment of breast tumors, but there is a problem of low ablation efficiency. Therefore, the improvement of HIFU efficiency to combat breast cancer is immediately needed. This study aimed to describe a novel anaerobic bacteria-mediated nanoplatform, comprising synergistic HIFU therapy for breast cancer under guidance of ultrasound (US) and magnetic resonance (MR) bimodal imaging. Methods: The PFH@CL/Fe3O4 nanoparticles (NPs) (Perfluorohexane (PFH) and superparamagnetic iron oxides (SPIO, Fe3O4) with cationic lipid (CL) NPs) were synthesized using the thin membrane hydration method. The novel nanoplatform Bifidobacterium bifidum-mediated PFH@CL/Fe3O4 NPs were constructed by electrostatic adsorption. Thereafter, US and MR bimodal imaging ability of B. bifidum-mediated PFH@CL/Fe3O4 NPs was evaluated in vitro and in vivo. Finally, the efficacy of HIFU ablation based on B. bifidum-PFH@CL/Fe3O4 NPs was studied. Results: B. bifidum combined with PFH@CL/Fe3O4 NPs by electrostatic adsorption and enhanced the tumor targeting ability of PFH@CL/Fe3O4 NPs. US and MR bimodal imaging clearly displayed the distribution of the bio-targeting nanoplatform in vivo. It was conducive for accurate and effective guidance of HIFU synergistic treatment of tumors. Furthermore, PFH@CL/Fe3O4 NPs could form microbubbles by acoustic droplet evaporation and promote efficiency of HIFU ablation under guidance of bimodal imaging. Conclusion: A bio-targeting nanoplatform with high stability and good physicochemical properties was constructed. The HIFU synergistic agent achieved early precision imaging of tumors and promoted therapeutic effect, monitored by US and MR bimodal imaging during the treatment process.


Asunto(s)
Neoplasias de la Mama , Ultrasonido Enfocado de Alta Intensidad de Ablación , Nanopartículas , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Humanos , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Nanopartículas/química , Ultrasonografía
6.
Biochem Biophys Res Commun ; 606: 29-34, 2022 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-35338856

RESUMEN

Drug-loaded nanoparticles have been widely used as synergists in high-intensity focused ultrasound (HIFU) tumor ablation therapy. However, these synergists have certain limitations, such as poor tumor targeting and low accumulation at the tumor site, that restrict the therapeutic efficacy of HIFU. In this study, we utilized drug-loaded nanoparticles conjugated with genetically engineered bacteria which can selectively colonize the hypoxic areas of tumor to facilitate HIFU ablation. Genetically modified Escherichia coli carrying gas vesicles (GVs-E. coli), which were gas-filled protein nanostructures, had a negatively charged surface and could specifically target into the tumor. In contrast, paclitaxel (PTX) and perfluorohexane (PFH) co-loaded cationic lipid nanoparticles (PTX-CLs) had a positively charged surface, hence, GVs-E. coli was used as a vehicle by conjugating with PTX-CLs via electrostatic adsorption and subsequently attracting more PTX-CLs to the tumor site. To improve the therapeutic efficiency of HIFU, the GVs in GVs-E. coli and PFH encapsulated in PTX-CLs could act as cavitation nuclei to enhance the HIFU cavitation effect, while PTX entrapped in PTX-CLs was released at the tumor site under HIFU irradiation, enhancing the therapeutic efficacy of HIFU and chemo-synergistic therapy. This novel combination strategy has great potential for cancer treatment.


Asunto(s)
Nanopartículas , Neoplasias , Línea Celular Tumoral , Escherichia coli/genética , Humanos , Liposomas , Nanopartículas/química , Neoplasias/terapia , Paclitaxel
7.
J Ultrasound Med ; 41(1): 107-121, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33724514

RESUMEN

PURPOSE: To investigate whether phase-shift perfluoropetane (PFP) nanoemulsions can enhance pulsed high-intensity focused ultrasound (HIFU) ablation. METHODS: PFP was encapsulated by poly(lactic-co-glycolic acid) (PLGA) to form a nanometer-sized droplet (PLGA-PFP), which was added to an isolated perfused liver system. Meanwhile, phosphate-buffered saline (PBS) was used as a control. The perfused liver was exposed to HIFU (150 W, t = 3/5/10 s) at various duty cycles (DCs). The ultrasound images, cavitation emissions, and temperature were recorded. Rabbits with subcutaneous VX2 tumors were exposed to HIFU (150 W) at various DCs with or without PLGA-PFP. After ablation, necrosis volume and energy efficiency factor were calculated. Pathologic characteristics were observed. RESULTS: Compared to the PBS control, PLGA-PFP nanoemulsions markedly enhanced HIFU-induced necrosis volume in both perfused livers and subcutaneous VX2 tumor-bearing rabbits (P <.05). Inertial cavitation was much stronger in the pulsed-HIFU exposure at 10% than that in the continuous-wave HIFU exposure (P <.01). Peak temperature at 100% DC was significantly higher than that at 10% (P <.05). Compared to 100% DC HIFU exposure, the mean necrosis volume induced by 10 s exposure at 50% DC was significantly larger (P <.005) but lower at 10% DC in the isolated perfused livers (P <.05). In addition, the mean necrosis volume in subcutaneous VX2 tumor-bearing rabbits was significantly increased after HIFU exposure at 10% DC when compared to those at 100% DC (P <.05). Histopathologic analysis showed liquefaction necrosis in pulsed HIFU. CONCLUSION: PLGA-PFP nanoemulsions can enhance HIFU ablation in the isolated perfused livers and promote tumor ablation in the subcutaneous xenograft rabbit model. Appropriate pulsed HIFU exposure may increase the necrosis volume and reduce total ultrasound energy required for HIFU ablation.


Asunto(s)
Ultrasonido Enfocado de Alta Intensidad de Ablación , Neoplasias , Animales , Fluorocarburos , Hígado/diagnóstico por imagen , Hígado/cirugía , Conejos
8.
Int J Nanomedicine ; 16: 4643-4659, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34267516

RESUMEN

PURPOSE: Hypoxia is considered to be obstructive to tumor treatment, but the reduced oxygen surroundings provide a suitable habitat for Bifidobacterium bifidum (BF) to colonize. The anaerobe BF selectively colonizes into tumors following systemic injection due to its preference for the hypoxia in the tumor cores. Therefore, BF may be a potential targeting agent which could be used effectively in tumor treatment. We aimed to determine whether a novel BF-mediated strategy, that was designed to deliver AP-PFH/PLGA NPs (aptamers CCFM641-5-functionalized Perfluorohexane (PFH) loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles) by aptamer-directed approach into solid tumor based on the tumor-targeting ability of BF, could improve efficiency of high intensity focused ultrasound (HIFU) treatment of breast cancer. METHODS: We synthesized AP-PFH/PLGA NPs using double emulsion method and carbodiimide method. Then, we evaluated targeting ability of AP-PFH/PLGA NPs to BF in vivo. Finally, we studied the efficacy of HIFU ablation based on BF plus AP-PFH/PLGA NPs (BF-mediated HIFU ablation) in tumor. RESULTS: The elaborately designed AP-PFH/PLGA NPs can target BF colonized in tumor to achieve high tumor accumulation, which can significantly enhance HIFU therapeutic efficiency. We also found that, compared with traditional chemotherapy, this therapy not only inhibits tumor growth, but also significantly prolongs the survival time of mice. More importantly, this treatment strategy has no obvious side effects. CONCLUSION: We successfully established a novel therapy method, BF-mediated HIFU ablation, which provides an excellent platform for highly efficient and non-invasive therapy of tumor.


Asunto(s)
Bifidobacterium bifidum/metabolismo , Portadores de Fármacos/metabolismo , Nanopartículas/administración & dosificación , Animales , Línea Celular Tumoral , Fluorocarburos/química , Humanos , Ratones , Nanopartículas/uso terapéutico , Poliésteres/química
9.
Biochem Biophys Res Commun ; 571: 104-109, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34314995

RESUMEN

The acoustic propagation characteristic of ultrasound determines that the energy of ultrasound beam will decrease with the increase of its propagation depth in the body. Similarly, the energy of High Intensity Focused Ultrasound (HIFU) will be attenuated with the increase of HIFU propagation depth in the body. Ensuring sufficient ultrasound energy deposition in the HIFU ablation region for tumor ablation is usually achieved by increasing the ultrasound irradiation power or prolonging the ultrasound ablation time. However, these two methods may damage the normal tissue adjacent to the HIFU ablation region. Herein, we constructed the nanoparticles conjugated with tumor-homing bacteria as the biological tumor-homing synergist to facilitate HIFU-mediated tumor ablation avoiding the potential safety risk. In our strategy, Bifidobacterium bifidum (B.bifidum) was selectively colonized in the hypoxic region of solid tumors after been injected into 4T1 breast cancer bearing-BALB/c mice via the tail vein due to its anaerobic growth characteristic. The amount of B. bifidum with negative surface potential in the hypoxic region of solid tumors was increased by its anaerobic proliferation. Polyethylenimine (PEI) -modified Poly (lactic-co-glycolic) acid nanoparticles loaded sodium bicarbonate (PEI-PLGA-NaHCO3-NPs) with positive surface potential injected into 4T1 breast cancer bearing-BALB/c mice via the tail vein displayed the tumor-homing ability by the electrostatic adsorption with B. bifidum colonized solid tumors. PEI-PLGA-NaHCO3-NPs could release NaHCO3 to produce carbon dioxide (CO2) as cavitation nuclei inside the acidic microenvironment of solid tumors. When HIFU irradiated solid tumors contained with more cavitation nuclei, the ultrasound energy deposition at the tumor region was increased to destroy the tumors more effectively. Meanwhile, the improved efficiency of HIFU-mediated tumor ablation reduced the dependence of the tumor ablation on the ultrasound energy dose, which improved the safety of HIFU-mediated tumor ablation to the non-targeted ablation tissue. This tumor-homing synergist shows the potential application value on the HIFU-mediated tumor ablation in the clinical.


Asunto(s)
Antineoplásicos/farmacología , Bifidobacterium bifidum/aislamiento & purificación , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/microbiología , Nanopartículas/química , Polietileneimina/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacología , Animales , Antineoplásicos/química , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Ratones , Ratones Endogámicos BALB C , Polietileneimina/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ondas Ultrasónicas
10.
Biomater Sci ; 9(6): 2230-2243, 2021 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-33507174

RESUMEN

Tumor therapy is facing the big challenge of insufficient treatment. Here, we report high-intensity focused ultrasound (HIFU)-responsive magnetic nanoparticles based on superparamagnetic iron oxide (SPIO, Fe3O4 NPs) as the shell and l-arginine (LA) as the core entrapped by poly-lactide-co-glycolide (PLGA) nanoparticles (Fe3O4@PLGA/LA NPs) for synergistic breast cancer therapy. These NPs can significantly enhance therapeutic performance due to their enhanced accumulation and prolonged retention at the tumor site under magnetic guidance. The Fe3O4@PLGA/LA NPs exhibited synergistic therapeutic effects by the rational combination of HIFU-based tumor ablation and nitric oxide (NO) assisted antitumor gas therapy. Both Fe3O4 NPs and LA could be released rapidly under HIFU irradiation, where Fe3O4 NPs can promote HIFU-based tumor ablation by changing the acoustic properties of the tumor tissues and LA can spontaneously react with hydrogen peroxide (H2O2) in the tumor microenvironment to generate NO for gas therapy. Moreover, Fe3O4 NPs can react with H2O2 to produce highly reactive oxygen-containing species (ROS) to accelerate the oxidation of LA and the release of NO. This novel strategy showed synergistic tumor growth suppression as compared with individual HIFU therapy or gas therapy. This can be attributed to the rational design of multifunctional NPs with magnetic targeting and multi-modality imaging properties.


Asunto(s)
Ultrasonido Enfocado de Alta Intensidad de Ablación , Nanopartículas de Magnetita , Nanopartículas , Neoplasias , Arginina , Línea Celular Tumoral , Humanos , Peróxido de Hidrógeno , Microambiente Tumoral
11.
Int J Nanomedicine ; 16: 105-117, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33447030

RESUMEN

PURPOSE: Cancer treatment still faces big challenges in the clinic, which is raising concerns over the world. In this study, we report the novel strategy of combing bacteriotherapy with high-intensity focused ultrasound (HIFU) therapy for more efficient breast cancer treatment. METHODS: The acoustic reporter gene (ARG) was genetically engineered to be expressed successfully in Escherichia coli (E. coli) to produce the protein nanoparticles-gas vesicles (GVs). Ultrasound was utilized to visualize the GVs in E. coli. In addition, it was injected intravenously for targeted breast cancer therapy by combing the bacteriotherapy with HIFU therapy. RESULTS: ARG expressed in E. coli can be visualized in vitro and in vivo by ultrasound. After intravenous injection, E. coli containing GVs could specifically target the tumor site, colonize consecutively in the tumor microenvironment, and it could obviously inhibit tumor growth. Meanwhile, E. coli which contained GVs could synergize HIFU therapy efficiently both in vitro and in vivo as the cavitation nuclei. Furthermore, the tumor inhibition rate in the combination therapy group could be high up to 87% compared with that in the control group. CONCLUSION: Our novel strategy of combing bacteriotherapy with HIFU therapy can treat breast cancers more effectively than the monotherapies, so it can be seen as a promising strategy.


Asunto(s)
Neoplasias de la Mama/terapia , Proteínas de Escherichia coli/genética , Ingeniería Genética , Nanopartículas/química , Acústica , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Terapia Combinada , Escherichia coli/metabolismo , Escherichia coli/ultraestructura , Femenino , Genes Reporteros , Ultrasonido Enfocado de Alta Intensidad de Ablación , Humanos , Liposomas , Ratones Endogámicos BALB C , Ratones Desnudos , Microambiente Tumoral
12.
Cancer Lett ; 498: 121-129, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33129956

RESUMEN

Cancer immunotherapy holds tremendous promise as a strategy for eradicating solid tumors, and its therapeutic effect highly relies on sufficient CD8+ T cells infiltration. Here, we demonstrate that ultrasound stimulated microbubble cavitation (USMC) promotes tumor perfusion, thereby increasing CD8+ T cells infiltration and anti-PD-L1 antibody delivery, then further enhancing the PD-L1 blockade of MC38 colon cancer in mice. Firstly, we optimized the mechanic index (MI) of ultrasound, and found that USMC with MI of 0.4 (equal to peak negative pressure of 0.8 MPa) significantly improved the peak intensity and area under curve of tumor contrast-enhanced ultrasound. Also, flow cytometry exhibited higher percentage of infiltrating CD8+ T cells in the USMC (MI = 0.4)-treated tumors than that of the control. We further explored the combination therapy of optimized USMC with anti-PD-L1 antibody. The combination therapy enhanced tumor perfusion and even led to the tumor vascular normalization. More importantly, flow cytometry showed that the combination not only increased the percentage and absolute number of tumor infiltrating CD8+ T cells, but also promoted the expression of Ki67 as well as the secretions of IFN γ and granzyme B, therefore, the combination therapy achieved greater tumor growth inhibition and longer survival than that of the monotherapies. These suggest that USMC is a promising therapeutic modality for combining immune checkpoint blockade against solid tumors.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/terapia , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Terapia Combinada/métodos , Femenino , Granzimas/metabolismo , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia/métodos , Interferón gamma/metabolismo , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos C57BL , Microburbujas , Perfusión/métodos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
13.
Int J Hyperthermia ; 37(1): 870-878, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32689830

RESUMEN

PURPOSE: This study was conducted to prepare a novel tumor-biotargeting high-intensity focused ultrasound (HIFU) synergist for indirectly delivering lipid nanoparticles based on the targeting ability of Bifidobacterium longum to the hypoxic region of solid tumors. The effects of two different delivery methods on the imaging and treatment of solid tumors enhanced by lipid nanoparticles were compared. METHODS: Biotinylated lipid nanoparticles coated with PFH were prepared, cross-linked with B. longum in vitro using a streptavidin-conjugated B. longum antibody (SBA), and observed and detected by laser confocal microscopy and flow cytometry. Solid tumors were treated with HIFU and PFH/BL-NPs. The effects of different delivery methods on the tumor targeting and efficiency of retention of PFH/BL-NPs were observed using Small animal live imaging and frozen sections from small animals. RESULTS: The PFH/BL-NPs prepared in this study showed good biocompatibility and safety. PFH/BL-NPs and B. longum were cross-linked in a cluster-like manner (confocal laser scanning microscope) in vitro, with a cross-linking rate of 84 ± 6.23% (flow cytometry). The delivery of B. longum followed by that of PFH/BL-NPs not only enhanced the ability of PFH/BL-NPs to target solid tumors (small animal live imaging), but also increased the retention time of PFH/BL-NPs in the tumor (frozen slices), enhancing the effect of the HIFU synergist. CONCLUSION: Delivery of B. longum followed by that of PFH/BL-NPs can enhance the imaging of solid tumors and effectively improve the efficiency of HIFU treatment of solid tumors, providing a basis for further clinical work.


Asunto(s)
Ultrasonido Enfocado de Alta Intensidad de Ablación , Nanopartículas , Neoplasias , Animales , Bifidobacterium , Neoplasias/diagnóstico por imagen , Neoplasias/terapia
14.
Int J Nanomedicine ; 15: 4151-4169, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32606670

RESUMEN

PURPOSE: Focused ultrasound (FUS) is a noninvasive method to produce thermal and mechanical destruction along with an immune-stimulatory effect against cancer. However, FUS ablation alone appears insufficient to generate consistent antitumor immunity. In this study, a multifunctional nanoparticle was designed to boost FUS-induced immune effects and achieve systemic, long-lasting antitumor immunity, along with imaging and thermal enhancement. MATERIALS AND METHODS: PEGylated PLGA nanoparticles encapsulating astragalus polysaccharides (APS) and gold nanorods (AuNRs) were constructed by a simple double emulsion method, characterized, and tested for cytotoxicity. The abilities of PA imaging and thermal-synergetic ablation efficiency were analyzed in vitro and in vivo. The immune-synergistic effect on dendritic cell (DC) differentiation in vitro and the immune response in vivo were also evaluated. RESULTS: The obtained APS/AuNR/PLGA-PEG nanoparticles have an average diameter of 255.00±0.1717 nm and an APS-loading efficiency of 54.89±2.07%, demonstrating their PA imaging capability and high biocompatibility both in vitro and in vivo. In addition, the as-prepared nanoparticles achieved a higher necrosis cell rate and induced apoptosis rate in an in vitro cell suspension assay, greater necrosis area and decreased energy efficiency factor (EEF) in an in vivo rabbit liver assay, and remarkable thermal-synergic performance. In particular, the nanoparticles upregulated the expression of MHC-II, CD80 and CD86 on cocultured DCs in vitro, followed by declining phagocytic function and enhanced interleukin (IL)-12 and interferon (INF)-γ production. Furthermore, they boosted the production of tumor necrosis factor (TNF)-α, IFN-γ, IL-4, IL-10, and IgG1 (P< 0.001) but not IgG2a. Immune promotion peaked on day 3 after FUS in vivo. CONCLUSION: The multifunctional APS/AuNR/PLGA-PEG nanoparticles can serve as an excellent synergistic agent for FUS therapy, facilitating real-time imaging, promoting thermal ablation effects, and boosting FUS-induced immune effects, which have the potential to be used for further clinical FUS treatment.


Asunto(s)
Planta del Astrágalo/química , Neoplasias de la Mama/terapia , Oro/química , Nanopartículas Multifuncionales/química , Nanotubos/química , Polisacáridos/química , Terapia por Ultrasonido , Animales , Antígenos CD/metabolismo , Apoptosis , Muerte Celular , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Citocinas/metabolismo , Células Dendríticas/citología , Femenino , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inmunoglobulina G/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos BALB C , Fagocitosis , Técnicas Fotoacústicas , Poliésteres/síntesis química , Poliésteres/química , Polietilenglicoles/síntesis química , Polietilenglicoles/química , Conejos , Nanomedicina Teranóstica , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Sci Rep ; 10(1): 7772, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32385414

RESUMEN

High intensity focused ultrasound (HIFU) has been recently shown as a rapidly developing new technique for non-invasive ablation of local tumors whose therapeutic efficiency can be significantly improved by changing the tissue acoustic environment (AET). Currently, the method of changing AET is mainly to introduce a medium with high acoustic impedance, but there are some disadvantages such as low retention of the introduced medium in the target area and a short residence time during the process. In our strategy, anaerobic bacterium Bifidobacterium longum (B. longum) which can colonize selectively in hypoxic regions of the animal body was successfully localized and shown to proliferate in the hypoxic zone of tumor tissue, overcoming the above disadvantages. This study aimed to explore the effects of Bifidobacteria on AET (including the structure and acoustic properties of tumor tissues) and HIFU ablation at different time. The results show that the injection of Bifidobacteria increased the collagen fibre number, elastic modulus and sound velocity and decreased neovascularization in tumor tissues. The number of collagen fibres and neovascularization decreased significantly over time. Under the same HIFU irradiation intensity, the B. longum injection increased the coagulative necrosis volume and decreased the energy efficiency factor (EEF). This study confirmed that Bifidobacteria can change the AET and increase the deposition of ultrasonic energy and thereby the efficiency of HIFU. In addition, the time that Bifidobacteria stay in the tumor area after injection is an important factor. This research provides a novel approach for synergistic biologically targeted HIFU therapy.


Asunto(s)
Acústica , Bifidobacterium/metabolismo , Bifidobacterium/efectos de la radiación , Ultrasonido Enfocado de Alta Intensidad de Ablación , Neoplasias/metabolismo , Ondas Ultrasónicas , Biomarcadores , Fenómenos Biomecánicos , Ingeniería Biomédica , Relación Dosis-Respuesta en la Radiación , Módulo de Elasticidad , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Hipoxia/metabolismo , Inmunohistoquímica , Neoplasias/terapia
16.
Int J Nanomedicine ; 15: 1871-1888, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32256065

RESUMEN

PURPOSE: The high-intensity focused ultrasound (HIFU) ablation of tumors is inseparable from synergistic agents and image monitoring, but the existing synergistic agents have the defects of poor targeting and a single imaging mode, which limits the therapeutic effects of HIFU. The construction of a multifunctional biological targeting synergistic agent with high biosafety, multimodal imaging and targeting therapeutic performance has great significance for combating cancer. METHODS: Multifunctional biological targeting synergistic agent consisting of Bifidobacterium longum (B. longum), ICG and PFH coloaded cationic lipid nanoparticles (CL-ICG-PFH-NPs) were constructed for targeting multimode imaging, synergistic effects with HIFU and imaging-guided ablation of tumors, which was evaluated both in vitro and in vivo. RESULTS: Both in vitro and in vivo systematical studies validated that the biological targeting synergistic agent can simultaneously achieve tumor-biotargeted multimodal imaging, HIFU synergism and multimodal image monitoring in HIFU therapy. Importantly, the electrostatic adsorption method and the targeting of B. longum to tumor tissues allow the CL-ICG-PFH-NPs to be retained in the tumor tissue, achieve the targeting ability of synergistic agent. Multimodal imaging chose the best treatment time according to the distribution of nanoparticles in the body to guide the efficient and effective treatment of HIFU. CL-ICG-PFH-NPs could serve as a phase change agent and form microbubbles that can facilitate HIFU ablation by mechanical effects, acoustic streaming and shear stress. This lays a foundation for the imaging and treatment of tumors. CONCLUSION: In this work, a biological targeting synergistic agent was successfully constructed with good stability and physicochemical properties. This biological targeting synergistic agent can not only provide information for early diagnosis of tumors but also realize multimodal imaging monitoring during HIFU ablation simultaneously with HIFU treatment, which improves the shortcomings of HIFU treatment and has broad application prospects.


Asunto(s)
Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Imagen Multimodal/métodos , Nanopartículas/química , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Animales , Bifidobacterium longum , Colorantes/química , Sistemas de Liberación de Medicamentos , Femenino , Fluorocarburos/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Verde de Indocianina/química , Ratones Endogámicos BALB C , Microburbujas , Nanopartículas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Nan Fang Yi Ke Da Xue Xue Bao ; 39(9): 1009-1016, 2019 Sep 30.
Artículo en Chino | MEDLINE | ID: mdl-31640961

RESUMEN

OBJECTIVE: To investigate the effects of Bifidobacterium on the acoustic characteristics of tumor tissue and how such acoustic changes affect the efficacy of high-intensity focused ultrasound (HIFU) ablation in nude mice. METHODS: Forty mice bearing human breast cancer cell (MDA-MB-231) xenograft were randomized into experimental group (n=20) and control group (n=20) for intravenous injection of Bifidobacterium suspension (200 µL, 4 × 108 cfu/mL) and PBS (200 µL) for 3 consecutive days, respectively. Before and at 3 and 7 days after the first injection, shear wave elastography was used to evaluate the hardness of the tumor tissue. On day 7 after the first injection, 10 mice from each group were sacrificed and the sound velocity and sound attenuation of the tumor tissues were measured. The changes in the collagen fibers in the tumors were evaluated using Masson staining, and neovascularization in the tumor was assessed with immunohistochemistry for platelet endothelial cell adhesion molecule-1 (PECAM-1/CD31). The remaining 10 tumor-bearing mice in each group were subjected to HIFU ablation, and the ablation efficiency was evaluated by assessing the changes in irradiation gray values, coagulative necrosis volume, energy efficiency factor (EEF) and irradiation area and by pathological examination with HE staining. RESULTS: In the experimental group, the collagen fibers in the tumor tissues were strong and densely aligned, and the tumors contained fewer new blood vessels showing strip-or spot-like morphologies. In the control group, the collagen fibers in the tumors were thin and loosely arranged, and the tumors showed abundant elongated or round new blood vessels. Bifidobacterium colonized in the tumor 7 days after the injection, and the tumor hardness was significantly greater in the experimental group than in the control group (P=0.01); the acoustic velocity (P=0.001) and the acoustic attenuation (P=0.000) of the tumor tissues were also greater in the experimental group. HIFU irradiation resulted in significantly greater changes in the gray scale of tumor (P=0.0006) and larger coagulative necrosis volume (P=0.0045) in the experimental group than in the control group, and the EEF was significantly smaller in the experimental group (P=0.0134). CONCLUSIONS: Bifidobacterium can cause changes in collagen fiber content, acoustic velocity and attenuation in the tumor tissue and reduce the EEF of HIFU irradiation, thereby improving the efficacy of HIFU irradiation.


Asunto(s)
Bifidobacterium/patogenicidad , Neoplasias de la Mama/patología , Ultrasonido Enfocado de Alta Intensidad de Ablación , Acústica , Animales , Colágeno , Diagnóstico por Imagen de Elasticidad , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Distribución Aleatoria
18.
Biochem Biophys Res Commun ; 514(4): 1147-1153, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31103266

RESUMEN

The hypoxic region microenvironment reduces the susceptibility of the cancer cells to radiotherapy and anticancer drugs of the solid tumors. However, the reduced oxygen surroundings provide an appreciable habitat for anaerobic bacteria to colonize and proliferate. Herein, we present a biocompatible bacteriabased system that can deliver poly(lactic-co-glycolic acid)(PLGA) nanoparticles(PLGA NPs) specifically targeting into solid tumor to achieve precision imaging and treatment. In our strategy, anaerobic bacterium Bifidobacterium longum (B. longum) that colonizes selectively in hypoxic regions of animal body was successfully used as a vehicle to conjugate with PLGA NPs and transported into solid tumors. To improve the efficacy and specificity of tumor therapy, low-boiling point perfluorohexane (PFH) liquid was wrapped in the core of PLGA NPs (PFH/PLGA NPs), which could increase the deposition of energy by affecting the acoustic environment of the tumor and destroy cells after liquid-gas phase transition during High Intensity Focused Ultrasound (HIFU) irradiation. This strategy shows an effective diagnosis and treatment integration for giving stronger imaging, longer retention period and more effective tumor therapy.


Asunto(s)
Antineoplásicos/farmacología , Bifidobacterium longum/química , Fluorocarburos/química , Nanopartículas/química , Imagen Óptica , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Animales , Femenino , Fluorocarburos/administración & dosificación , Humanos , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Neoplasias Mamarias Experimentales/microbiología , Neoplasias Mamarias Experimentales/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/administración & dosificación , Células Tumorales Cultivadas
19.
Sci Rep ; 9(1): 6982, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31061456

RESUMEN

High intensity focused ultrasound (HIFU) is a noninvasive thermal ablation technique for the treatment of benign and malignant solid masses. To improve the efficacy of HIFU ablation, we developed poly (lactide-co-glycolide) (PLGA) nanoparticles encapsulating perfluoropentane (PFP) and hematoporphyrin monomethyl ether (HMME) as synergistic agents (HMME+PFP/PLGA). Two-step biotin-avidin pre-targeting technique was applied for the HIFU ablation. We further modified the nanoparticles with streptavidin (HMME+PFP/PLGA-SA). HMME+PFP/PLGA-SA were highly dispersed with spherical morphology (477.8 ± 81.8 nm in diameter). The encapsulation efficiency of HMME and PFP were 46.6 ± 3.3% and 40.1 ± 2.6%, respectively. The binding efficiency of nanoparticles to streptavidin was 95.5 ± 2.5%. The targeting ability of the HMME+PFP/PLGA-SA nanoparticles was tested by parallel plate flow chamber in vitro. In the pre-targeting group (HMME+PFP/PLGA-SA), a large number of nanoparticles bound to the peripheral and surface of the cell. In the HIFU ablation experiment in vivo, compared with the other groups, the largest gray-scale changes and coagulation necrosis areas were observed in the pre-targeting (HMME+PFP/PLGA-SA) group, with the lowest energy efficiency factor value. Moreover, the microvessel density and proliferation index declined, while the apoptotic index increased, in the tumor tissue surrounding the coagulation necrosis area in the pre-targeting group. Meanwhile, the survival time of the tumor-bearing nude mice in the pre-targeting group was significantly longer than that in the HIFU treatment group. These results suggest that HMME+PFP/PLGA-SA have high potential to act as synergistic agents in HIFU ablation.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias de la Mama/terapia , Fluorocarburos/química , Hematoporfirinas/química , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Nanopartículas/administración & dosificación , Animales , Apoptosis , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Terapia Combinada , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Células Tumorales Cultivadas , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Sci Rep ; 9(1): 6423, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31015517

RESUMEN

High intensity focused ultrasound (HIFU) has been recently regarded to be a new type of technique for non-invasive ablation of local tumors and HIFU synergists could significantly improve its therapeutic efficiency. The therapeutic efficiency of HIFU is greatly limited by the low retention of HIFU synergists in the target area and short residence time. This study aimed to explore a method to increase the deposition of HIFU synergists in tumors. Cationic lipid nanoparticle can be used to enhance the HIFU ablation effect, but there is still a problem for it that the deposition amount in the tumor tissue is small and the residence time is short. Bifidobacterium is highly biosafe and can be selectively colonized in the hypoxic zone of tumor tissue. Cationic lipid nanoparticles can be observed in vitro by attachment to bifidobacterium by electrostatic adsorption. And the effect of the proliferation of bifidobacterium in tumor tissues on the retention amount and retention time of cationic lipid nanoparticles in vivo was evaluated. Results showed that the cationic lipid nanoparticles were linked to the surface of Bifidobacterium effectively in vitro, while in vivo, the retention amount and retention time of cationic lipid nanoparticles could be increased by Bifidobacterium in tumor tissues, which provided a new method for improving the therapeutic efficiency of HIFU.


Asunto(s)
Bifidobacterium/química , Neoplasias de la Mama/terapia , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Nanopartículas/administración & dosificación , Animales , Adhesión Bacteriana , Bifidobacterium/fisiología , Transporte Biológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cationes , Línea Celular Tumoral , Colesterol/análogos & derivados , Colesterol/química , Femenino , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Nanopartículas/química , Fosfatidiletanolaminas/química , Polietilenglicoles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA