1
|
Hu HY, Sun YJ, Yuan XF, Han JF, Liao TT, Zhang FY, Mao JD, Zhang L, Ye WL. Ultrasound-controllable dexamethasone-loaded nanobubbles for highly effective rheumatoid arthritis therapy. J Mater Chem B 2025; 13:2052-2066. [PMID: 39757977 DOI: 10.1039/d4tb01120a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that seriously threatens human health and affects the quality of life of patients. At present, pharmacotherapy is still the mainstream treatment for RA, but most methods have shortcomings, such as poor drug targeting, a low effective drug dosage at the inflammatory site, and high systemic toxicity. The combined application of drug-loaded nanobubbles and ultrasound technology provides a new technique for the treatment of RA. Low-intensity focused ultrasound (LIFU) traces the transmission of drug-loaded nanobubbles in the body, and high-intensity focused ultrasound (HIFU) causes the nanobubbles to rupture to release drugs at the inflammatory site, thereby reducing their toxicity to normal tissues. In this study, a drug-loaded nanobubble delivery system (DEXsp@Liposomes/C3F8) with ultrasonic response characteristics was successfully constructed, and its therapeutic effect was evaluated for the treatment of RA in vitro and in vivo. DEXsp@Liposomes/C3F8 + LIFU had good biocompatibility and excellent ultrasound imaging ability. DEXsp@Liposomes/C3F8 +HIFU distinctly increased the cellular uptake of DEXsp and significantly reduced the secretion of related inflammatory factors in RAW264.7 cells. Moreover, DEXsp@Liposomes/C3F8 + HIFU effectively alleviated the symptoms of RA in model rats and significantly improved their exercise capacity. In conclusion, the prepared ultrasound-mediated DEXsp@Liposomes/C3F8 system exhibits good imaging, monitoring and therapeutic effects, and the results of this study provide a new direction for the diagnosis and treatment of RA.
Collapse
Affiliation(s)
- Hang-Yi Hu
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.
- Department of Materials Science, Shenzhen MSU-BIT University, Shenzhen, 518172, China
| | - Ying-Jian Sun
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.
| | - Xiao-Feng Yuan
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.
- Department of Pharmacy, Chinese People's Liberation Army Logistics Support Force No. 967 Hospital, Dalian, 116021, China
| | - Jiang-Fan Han
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.
| | - Tian-Tian Liao
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.
| | - Fei-Yue Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jin-Dong Mao
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.
| | - Lin Zhang
- Department of Outpatient Service, 986th Hospital Affilliated to Air Force Medical University, Xi'an, China.
| | - Wei-Liang Ye
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
2
|
Tang Y, Shen Q, Lin P, Chen Z, Fan D, Zhuo M, Gan Y, Su Y, Qian Q, Lin L, Xue E, Chen Z. aPD-L1-facilitated theranostic and tumor microenvironment remodeling of pancreatic cancer via docetaxel-loaded phase-transformation nanoparticles triggered by low-intensity pulsed ultrasound. J Nanobiotechnology 2025; 23:48. [PMID: 39871305 PMCID: PMC11773723 DOI: 10.1186/s12951-025-03105-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025] Open
Abstract
Early diagnosis of pancreatic ductal adenocarcinoma (PDAC) is challenging because of its depth, which often leads to misdiagnosis during ultrasound examinations. The unique PDAC tumor microenvironment (TME) is characterized by significant fibrous tissue growth, and high interstitial pressure hinders drug penetration into tumors. Additionally, hypoxia and immune suppression within the tumor contribute to poor responses to radiotherapy and chemotherapy, ultimately leading to an unfavorable prognosis. In this study, aPD-L1-modified docetaxel and perfluoropentane-loaded liquid‒vapor phase-transformation lipid nanoparticles (aPDL1-DTX/PFP@Lipid) were synthesized and had an average diameter of 61.63 nm with 84.3% antibody modification. We demonstrated that the nanoparticles (NPs) exhibited excellent PDAC-targeting capabilities both in vitro and in vivo. Upon exposure to low-intensity pulsed ultrasound (LIPUS) stimulation, the NPs underwent a phase transformation to form microbubbles with substantial molecular ultrasound diagnostic effects, and combined treatment resulted in a tumor growth inhibition rate of 88.91%. This treatment strategy also led to the infiltration of CD8+ T cells, the downregulation of Treg cells, the promotion of M1 macrophage polarization, the inhibition of fibrosis to reduce tumor stromal pressure, and the facilitation of perfluoropentane (PFP) gasification to release O2 and improve tumor hypoxia. In conclusion, aPD-L1-modified liquid‒vapor phase-transformation nanoparticles loaded with docetaxel (DTX) and PFP were successfully combined with ultrasound for the molecular diagnosis and targeted treatment of PDAC. aPDL1-DTX/PFP@Lipid could reshape the PDAC TME, offering a new approach for ultrasound-mediated diagnosis and treatment with promising clinical applications.
Collapse
Affiliation(s)
- Yi Tang
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
- Fujian Institute of Ultrasonic Medicine Laboratory, 29 Xinquan Road, Fuzhou, China
| | - Qingling Shen
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
- Fujian Institute of Ultrasonic Medicine Laboratory, 29 Xinquan Road, Fuzhou, China
| | - Peng Lin
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Zhixin Chen
- Fujian College Association Instrumental Analysis Center, Fuzhou University, 2 Xueyuan Road, Fuzhou, China
| | - Denghui Fan
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, China
| | - Minling Zhuo
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Yajiao Gan
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Yixi Su
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Qingfu Qian
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Liwu Lin
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Ensheng Xue
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Zhikui Chen
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China.
- Fujian Institute of Ultrasonic Medicine Laboratory, 29 Xinquan Road, Fuzhou, China.
| |
Collapse
|
3
|
Sardari M, Ghanati F, Mobasheri H, Hajnorouzi A. Short-term airborne ultrasound induced cell death in tobacco cells and changed their wall components. Sci Rep 2025; 15:3509. [PMID: 39875541 PMCID: PMC11775165 DOI: 10.1038/s41598-025-87762-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025] Open
Abstract
The effects of low-intensity ultrasound on plants such as piezoelectric and ultrasonic water baths, on plants have been extensively studied. However, the specific effect of airborne ultrasound on plant cells has yet to be reported. The present study was conducted to elucidate the physiological responses of plant cells to airborne US. Homogeneous suspension-cultured tobacco cells (Nicotiana tabacum L. cv Burley 21) were subjected to airborne US at 24 kHz in two pulsatile and continuous modes for 10 and 20 s. The study's outcome revealed that airborne US triggered the production of H2O2, elevated internal calcium concentration, and reduced antioxidant capacity upon cavitation. Alteration of covalently bound peroxidase and other wall-modifying enzyme activities was accompanied by reduced cellulose, pectin, and hemicellulose B but increased lignin and hemicellulose A. The biomass and viability of tobacco cells were also significantly decreased by airborne US, which ultimately resulting in PCD and secondary necrosis. The results highlight the potential risks of even short-time exposure to the airborne US on plant physiology and cell wall chemical composition raising significant concerns about its implications.
Collapse
Affiliation(s)
- Mahsa Sardari
- Department of Plant Biology, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Faezeh Ghanati
- Department of Plant Biology, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran.
| | - Hamid Mobasheri
- Laboratory of Membrane Biophysics and Macromolecules, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Abazar Hajnorouzi
- Department of Physics, Faculty of Basic Sciences, Shahed University, Tehran, Iran
| |
Collapse
|
4
|
Yang H, Qu Y, Tian Y, Wang C, Sun Y, Dai Z, Yue X, Cheng W. Ultrasound-Targeted Microbubble Destruction Enhances the Inhibitory Effect of Sonodynamic Therapy against Hepatocellular Carcinoma. ACS OMEGA 2024; 9:51253-51263. [PMID: 39758613 PMCID: PMC11696411 DOI: 10.1021/acsomega.4c07746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/16/2024] [Accepted: 11/28/2024] [Indexed: 01/07/2025]
Abstract
Purpose: To assess the anticancer effect of microbubbles (MBs) in combination with sinoporphyrin sodium (DVDMS)-mediated sonodynamic therapy (SDT) for the in vitro and in vivo treatment of hepatocellular carcinoma (HCC). Methods: HepG2 cells were used for in vitro experiments. Reactive oxygen species (ROS) production was detected using 2',7'-dichlorodihydrofluorescein diacetate and singlet oxygen sensor green in vitro and in solution, respectively. Cytotoxicity was evaluated using a Cell Counting Kit 8 assay and the calcein AM/PI double-staining method. Annexin V-FITC/PI staining was employed to analyze the rate of cell apoptosis. Cell surface calreticulin exposure, high mobility group box 1 release, and adenosine triphosphate secretion were measured to detect immunogenic cell death (ICD). The anticancer effect of the combination therapy was further assessed in Hepa1-6 tumor-bearing mice. Results: Compared with SDT alone, ROS production in the MBs + SDT group was enhanced 1.2-fold (p < 0.0001). The cytotoxic effect of DVDMS-mediated SDT on HepG2 cells was concentration-dependent, and the additional application of MBs increased cytotoxicity. Additionally, MBs augmented the SDT-induced apoptosis rate from 33.26 ± 13.48 to 72.95 ± 7.95% (p < 0.01). Notably, our results demonstrated that MBs can enhance SDT-induced ICD. In in vivo experiments, SDT combined with MBs significantly reduced tumor volume, with negligible differences in mouse body weight. Furthermore, MBs effectively enhanced SDT-induced tumor tissue destruction. Conclusion: The present study indicates that MBs can markedly improve the anticancer effects of SDT in HCC.
Collapse
Affiliation(s)
- Huajing Yang
- Department
of Ultrasound, Harbin Medical University
Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang Province 150081, China
| | - Yunfeng Qu
- Department
of Biomedical Engineering, College of Future Technology, National
Biomedical Imaging Center, Peking University, No.5 Yiheyuan Road, Beijing 100871, China
| | - Yuhang Tian
- Department
of Ultrasound, Harbin Medical University
Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang Province 150081, China
| | - Chunyue Wang
- Department
of Ultrasound, Harbin Medical University
Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang Province 150081, China
| | - Yucao Sun
- Department
of Ultrasound, Harbin Medical University
Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang Province 150081, China
| | - Zhifei Dai
- Department
of Biomedical Engineering, College of Future Technology, National
Biomedical Imaging Center, Peking University, No.5 Yiheyuan Road, Beijing 100871, China
| | - Xiuli Yue
- School
of Environment, Harbin Institute of Technology, No.92 Xidazhi Street, Harbin, Heilongjiang Province 150001, China
| | - Wen Cheng
- Department
of Ultrasound, Harbin Medical University
Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang Province 150081, China
| |
Collapse
|
5
|
Jiang X, Yu W, Chen Z, Li C, Li X, Xu Y, Li F, Gao H, Qian J, Xiong B, Rong S, Chen G, She Q, Huang J. Low-intensity focused ultrasound combined with microbubbles for non-invasive downregulation of rabbit carotid body activity in the treatment of hypertension. Hypertens Res 2024; 47:3182-3192. [PMID: 39300302 DOI: 10.1038/s41440-024-01904-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024]
Abstract
Targeting the carotid body (CB) is a new approach in treating hypertension. This study investigates the efficacy and safety of ultrasound combined with microbubbles in targeting CB to treat hypertension. Twenty-seven hypertensive rabbits were randomly assigned to three groups: microbubbles only (sham group, n = 11), ultrasound plus microbubbles (LIFU group, n = 11), and bilateral carotid sinus nerve denervation (CSND group, n = 5). Four weeks post-intervention, blood pressure, hypoxic ventilatory response (HVR), blood pressure variability (BPV), heart rate variability (HRV), biochemical indicators, neurohormones, and histopathology were assessed in all groups. The results indicated significant reductions in systolic and diastolic blood pressure in the LIFU and CSND groups post-intervention, along with decreases in BPV, HRV, and catecholamines. HVR results showed a 35.10% reduction in CB activity in the LIFU group compared to the sham group, which was significantly lower than the reduction in the CSND group compared to the sham group (73.85%). Histopathology and transmission electron microscopy confirmed CB damage and cell apoptosis, with immunofluorescence showing a reduction in type I and II cells. In conclusion, LIFU combined with microbubbles can reduce blood pressure by lowering CB and sympathetic nerve activity.
Collapse
Affiliation(s)
- Xiujuan Jiang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Ultrasound Imaging, Department of Ultrasound, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Cardiology, People's Hospital of Santai County, Santai County, Sichuan, China
| | - Wei Yu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Ultrasound Imaging, Department of Ultrasound, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zijun Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Cardiology, The Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Chaohong Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xingyu Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Xu
- Department of Cardiology, People's Hospital of Santai County, Santai County, Sichuan, China
| | - Fugui Li
- Department of Cardiology, People's Hospital of Santai County, Santai County, Sichuan, China
| | - Hongli Gao
- Department of Cardiology, The Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Qian
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bo Xiong
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shunkang Rong
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guozhu Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Huang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Institute of Ultrasound Imaging, Department of Ultrasound, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
6
|
Su X, Li J, Xu X, Ye Y, Wang C, Pang G, Liu W, Liu A, Zhao C, Hao X. Strategies to enhance the therapeutic efficacy of anti-PD-1 antibody, anti-PD-L1 antibody and anti-CTLA-4 antibody in cancer therapy. J Transl Med 2024; 22:751. [PMID: 39123227 PMCID: PMC11316358 DOI: 10.1186/s12967-024-05552-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Although immune checkpoint inhibitors (anti-PD-1 antibody, anti-PD-L1 antibody, and anti-CTLA-4 antibody) have displayed considerable success in the treatment of malignant tumors, the therapeutic effect is still unsatisfactory for a portion of patients. Therefore, it is imperative to develop strategies to enhance the effect of these ICIs. Increasing evidence strongly suggests that the key to this issue is to transform the tumor immune microenvironment from a state of no or low immune infiltration to a state of high immune infiltration and enhance the tumor cell-killing effect of T cells. Therefore, some combination strategies have been proposed and this review appraise a summary of 39 strategies aiming at enhancing the effectiveness of ICIs, which comprise combining 10 clinical approaches and 29 foundational research strategies. Moreover, this review improves the comprehensive understanding of combination therapy with ICIs and inspires novel ideas for tumor immunotherapy.
Collapse
Affiliation(s)
- Xin Su
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Jian Li
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Xiao Xu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Youbao Ye
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Cailiu Wang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Guanglong Pang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Wenxiu Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Ang Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Changchun Zhao
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Xiangyong Hao
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China.
| |
Collapse
|
7
|
Bhoopathi P, Mannangatti P, Pradhan AK, Kumar A, Maji S, Lang FF, Klibanov AL, Madan E, Cavenee WK, Keoprasert T, Sun D, Bjerkvig R, Thorsen F, Gogna R, Das SK, Emdad L, Fisher PB. Noninvasive therapy of brain cancer using a unique systemic delivery methodology with a cancer terminator virus. J Cell Physiol 2024; 239:e31302. [PMID: 38775127 DOI: 10.1002/jcp.31302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/03/2024] [Accepted: 04/30/2024] [Indexed: 08/15/2024]
Abstract
Primary, glioblastoma, and secondary brain tumors, from metastases outside the brain, are among the most aggressive and therapeutically resistant cancers. A physiological barrier protecting the brain, the blood-brain barrier (BBB), functions as a deterrent to effective therapies. To enhance cancer therapy, we developed a cancer terminator virus (CTV), a unique tropism-modified adenovirus consisting of serotype 3 fiber knob on an otherwise Ad5 capsid that replicates in a cancer-selective manner and simultaneously produces a potent therapeutic cytokine, melanoma differentiation-associated gene-7/interleukin-24 (MDA-7/IL-24). A limitation of the CTV and most other viruses, including adenoviruses, is an inability to deliver systemically to treat brain tumors because of the BBB, nonspecific virus trapping, and immune clearance. These obstacles to effective viral therapy of brain cancer have now been overcome using focused ultrasound with a dual microbubble treatment, the focused ultrasound-double microbubble (FUS-DMB) approach. Proof-of-principle is now provided indicating that the BBB can be safely and transiently opened, and the CTV can then be administered in a second set of complement-treated microbubbles and released in the brain using focused ultrasound. Moreover, the FUS-DMB can be used to deliver the CTV multiple times in animals with glioblastoma growing in their brain thereby resulting in a further enhancement in survival. This strategy permits efficient therapy of primary and secondary brain tumors enhancing animal survival without promoting harmful toxic or behavioral side effects. Additionally, when combined with a standard of care therapy, Temozolomide, a further increase in survival is achieved. The FUS-DMB approach with the CTV highlights a noninvasive strategy to treat brain cancers without surgery. This innovative delivery scheme combined with the therapeutic efficacy of the CTV provides a novel potential translational therapeutic approach for brain cancers.
Collapse
Affiliation(s)
- Praveen Bhoopathi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Padmanabhan Mannangatti
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Anjan K Pradhan
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Amit Kumar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Santanu Maji
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Frederick F Lang
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Alexander L Klibanov
- Biomedical Engineering, Radiology and Medical Imaging, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Esha Madan
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- Department of Surgery, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Webster K Cavenee
- Ludwig Institute for Cancer Research, University of San Diego, La Jolla, California, USA
| | - Timothy Keoprasert
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Dong Sun
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Rolf Bjerkvig
- Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Centre, University of Bergen, Bergen, Norway
| | - Frits Thorsen
- Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Centre, University of Bergen, Bergen, Norway
| | - Rajan Gogna
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
8
|
Huang H, Zheng Y, Chang M, Song J, Xia L, Wu C, Jia W, Ren H, Feng W, Chen Y. Ultrasound-Based Micro-/Nanosystems for Biomedical Applications. Chem Rev 2024; 124:8307-8472. [PMID: 38924776 DOI: 10.1021/acs.chemrev.4c00009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Due to the intrinsic non-invasive nature, cost-effectiveness, high safety, and real-time capabilities, besides diagnostic imaging, ultrasound as a typical mechanical wave has been extensively developed as a physical tool for versatile biomedical applications. Especially, the prosperity of nanotechnology and nanomedicine invigorates the landscape of ultrasound-based medicine. The unprecedented surge in research enthusiasm and dedicated efforts have led to a mass of multifunctional micro-/nanosystems being applied in ultrasound biomedicine, facilitating precise diagnosis, effective treatment, and personalized theranostics. The effective deployment of versatile ultrasound-based micro-/nanosystems in biomedical applications is rooted in a profound understanding of the relationship among composition, structure, property, bioactivity, application, and performance. In this comprehensive review, we elaborate on the general principles regarding the design, synthesis, functionalization, and optimization of ultrasound-based micro-/nanosystems for abundant biomedical applications. In particular, recent advancements in ultrasound-based micro-/nanosystems for diagnostic imaging are meticulously summarized. Furthermore, we systematically elucidate state-of-the-art studies concerning recent progress in ultrasound-based micro-/nanosystems for therapeutic applications targeting various pathological abnormalities including cancer, bacterial infection, brain diseases, cardiovascular diseases, and metabolic diseases. Finally, we conclude and provide an outlook on this research field with an in-depth discussion of the challenges faced and future developments for further extensive clinical translation and application.
Collapse
Affiliation(s)
- Hui Huang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yi Zheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Jun Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Lili Xia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Chenyao Wu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wei Feng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yu Chen
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
9
|
Ren L, Wang L, Yi X, Tan Y, Yi L, He J, Li D. Ultrasound Microbubble-Stimulated miR-145-5p Inhibits Malignant Behaviors of Breast Cancer Cells by Targeting ACTG1. Ultrasound Q 2024; 40:136-143. [PMID: 38350033 DOI: 10.1097/ruq.0000000000000678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
ABSTRACT Ultrasound-targeted microbubble destruction (UTMD) technology combines ultrasound with a variety of functional microbubble vectors to enhance the transfection and expression of target genes, and has become a promising noninvasive method for localized gene transfer, which is widely used in gene therapy for cancer. This research aimed to explore the role of UTMD-mediated miR-145-5p on breast cancer (BC) tumorigenesis and the underlying mechanisms. To achieve UTMD-mediated miR-145-5p overexpression, BC cells were cotransfected with microbubbles (MBs) and miR-145-5p mimics. The BC cell malignant phenotypes were assessed through CCK-8, wound healing, and transwell assays. MiR-145-5p and actin gamma 1 (ACTG1) binding relationship was verified through luciferase reporter and RNA pull-down assays. MiR-145-5p and ACTG1 levels in BC cells and tissues were detected through RT-qPCR and Western blotting. ACTG1 was upregulated, whereas miR-145-5p was downregulated in BC cells and tissues. MiR-145-5p targeted ACTG1 and negatively regulated its level in BC cells. Overexpressing miR-145-5p restrained BC cell growth, migration, and invasion. Ultrasound-targeted microbubble destruction improved the overexpression efficiency of miR-145-5p and enhanced the suppressive influence on BC cell malignant phenotypes. In addition, ACTG1 overexpression compromises the repression of UTMD-mediated miR-145-5p on cellular behaviors in BC. Ultrasound-targeted microbubble destruction-delivered miR-145-5p hindered malignant behaviors of BC cells through downregulating ACTG1.
Collapse
Affiliation(s)
| | - Li Wang
- Yichang Yiling People's Hospital, Yichang, Hubei, China
| | - Xuelin Yi
- Yichang Yiling People's Hospital, Yichang, Hubei, China
| | - Yang Tan
- Yichang Yiling People's Hospital, Yichang, Hubei, China
| | - Lingxian Yi
- Yichang Yiling People's Hospital, Yichang, Hubei, China
| | - Jinlan He
- Yichang Yiling People's Hospital, Yichang, Hubei, China
| | | |
Collapse
|
10
|
Wu Y, Li J, Shu L, Tian Z, Wu S, Wu Z. Ultrasound combined with microbubble mediated immunotherapy for tumor microenvironment. Front Pharmacol 2024; 15:1304502. [PMID: 38487163 PMCID: PMC10937735 DOI: 10.3389/fphar.2024.1304502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/11/2024] [Indexed: 03/17/2024] Open
Abstract
The tumor microenvironment (TME) plays an important role in dynamically regulating the progress of cancer and influencing the therapeutic results. Targeting the tumor microenvironment is a promising cancer treatment method in recent years. The importance of tumor immune microenvironment regulation by ultrasound combined with microbubbles is now widely recognized. Ultrasound and microbubbles work together to induce antigen release of tumor cell through mechanical or thermal effects, promoting antigen presentation and T cells' recognition and killing of tumor cells, and improve tumor immunosuppression microenvironment, which will be a breakthrough in improving traditional treatment problems such as immune checkpoint blocking (ICB) and himeric antigen receptor (CAR)-T cell therapy. In order to improve the therapeutic effect and immune regulation of TME targeted tumor therapy, it is necessary to develop and optimize the application system of microbubble ultrasound for organs or diseases. Therefore, the combination of ultrasound and microbubbles in the field of TME will continue to focus on developing more effective strategies to regulate the immunosuppression mechanisms, so as to activate anti-tumor immunity and/or improve the efficacy of immune-targeted drugs, At present, the potential value of ultrasound combined with microbubbles in TME targeted therapy tumor microenvironment targeted therapy has great potential, which has been confirmed in the experimental research and application of breast cancer, colon cancer, pancreatic cancer and prostate cancer, which provides a new alternative idea for clinical tumor treatment. This article reviews the research progress of ultrasound combined with microbubbles in the treatment of tumors and their application in the tumor microenvironment.
Collapse
Affiliation(s)
| | | | | | | | | | - Zuohui Wu
- Department of Ultrasound, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
11
|
Zhang G, Zhan M, Zhang C, Wang Z, Sun H, Tao Y, Shi Q, He M, Wang H, Rodrigues J, Shen M, Shi X. Redox-Responsive Dendrimer Nanogels Enable Ultrasound-Enhanced Chemoimmunotherapy of Pancreatic Cancer via Endoplasmic Reticulum Stress Amplification and Macrophage Polarization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301759. [PMID: 37350493 PMCID: PMC10460845 DOI: 10.1002/advs.202301759] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/01/2023] [Indexed: 06/24/2023]
Abstract
Developing a multifunctional nanoplatform to achieve efficient theranostics of tumors through multi-pronged strategies remains to be challenging. Here, the design of the intelligent redox-responsive generation 3 (G3) poly(amidoamine) dendrimer nanogels (NGs) loaded with gold nanoparticles (Au NPs) and chemotherapeutic drug toyocamycin (Au/Toy@G3 NGs) for ultrasound-enhanced cancer theranostics is showcased. The constructed hybrid NGs with a size of 193 nm possess good colloidal stability under physiological conditions, and can be dissociated to release Au NPs and Toy in the reductive glutathione-rich tumor microenvironment (TME). The released Toy can promote the apoptosis of cancer cells through endoplasmic reticulum stress amplification and cause immunogenic cell death to maturate dendritic cells. The loaded Au NPs can induce the conversion of tumor-associated macrophages from M2-type to antitumor M1-type to remodulate the immunosuppressive TME. Combined with antibody-mediated immune checkpoint blockade, effective chemoimmunotherapy of a pancreatic tumor mouse model can be realized, and the chemoimmunotherapy effect can be further ultrasound enhanced due to the sonoporation-improved tumor permeability of NGs. The developed Au/Toy@G3 NGs also enable Au-mediated computed tomography imaging of tumors. The constructed responsive dendrimeric NGs tackle tumors through a multi-pronged chemoimmunotherapy strategy targeting both cancer cells and immune cells, which hold a promising potential for clinical translations.
Collapse
Affiliation(s)
- Guizhi Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsShanghai Engineering Research Center of Nano‐Biomaterials and Regenerative MedicineCollege of Biological Science and Medical EngineeringDonghua UniversityShanghai201620China
| | - Mengsi Zhan
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsShanghai Engineering Research Center of Nano‐Biomaterials and Regenerative MedicineCollege of Biological Science and Medical EngineeringDonghua UniversityShanghai201620China
| | - Changchang Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsShanghai Engineering Research Center of Nano‐Biomaterials and Regenerative MedicineCollege of Biological Science and Medical EngineeringDonghua UniversityShanghai201620China
| | - Zhiqiang Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsShanghai Engineering Research Center of Nano‐Biomaterials and Regenerative MedicineCollege of Biological Science and Medical EngineeringDonghua UniversityShanghai201620China
| | - Huxiao Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsShanghai Engineering Research Center of Nano‐Biomaterials and Regenerative MedicineCollege of Biological Science and Medical EngineeringDonghua UniversityShanghai201620China
| | - Yuchen Tao
- Department of UltrasoundShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Qiusheng Shi
- Department of UltrasoundShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Meijuan He
- Department of RadiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Han Wang
- Department of RadiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - João Rodrigues
- CQM – Centro de Química da MadeiraMMRGUniversidade da MadeiraCampus Universitário da PenteadaFunchal9020‐105Portugal
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsShanghai Engineering Research Center of Nano‐Biomaterials and Regenerative MedicineCollege of Biological Science and Medical EngineeringDonghua UniversityShanghai201620China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsShanghai Engineering Research Center of Nano‐Biomaterials and Regenerative MedicineCollege of Biological Science and Medical EngineeringDonghua UniversityShanghai201620China
- CQM – Centro de Química da MadeiraMMRGUniversidade da MadeiraCampus Universitário da PenteadaFunchal9020‐105Portugal
| |
Collapse
|
12
|
Navarro-Becerra JA, Borden MA. Targeted Microbubbles for Drug, Gene, and Cell Delivery in Therapy and Immunotherapy. Pharmaceutics 2023; 15:1625. [PMID: 37376072 DOI: 10.3390/pharmaceutics15061625] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Microbubbles are 1-10 μm diameter gas-filled acoustically-active particles, typically stabilized by a phospholipid monolayer shell. Microbubbles can be engineered through bioconjugation of a ligand, drug and/or cell. Since their inception a few decades ago, several targeted microbubble (tMB) formulations have been developed as ultrasound imaging probes and ultrasound-responsive carriers to promote the local delivery and uptake of a wide variety of drugs, genes, and cells in different therapeutic applications. The aim of this review is to summarize the state-of-the-art of current tMB formulations and their ultrasound-targeted delivery applications. We provide an overview of different carriers used to increase drug loading capacity and different targeting strategies that can be used to enhance local delivery, potentiate therapeutic efficacy, and minimize side effects. Additionally, future directions are proposed to improve the tMB performance in diagnostic and therapeutic applications.
Collapse
Affiliation(s)
| | - Mark A Borden
- Mechanical Engineering Department, University of Colorado Boulder, Boulder, CO 80309, USA
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
13
|
Wu N, Cao Y, Liu Y, Zhou Y, He H, Tang R, Wan L, Wang C, Xiong X, Zhong L, Li P. Low-intensity focused ultrasound targeted microbubble destruction reduces tumor blood supply and sensitizes anti-PD-L1 immunotherapy. Front Bioeng Biotechnol 2023; 11:1173381. [PMID: 37139047 PMCID: PMC10150078 DOI: 10.3389/fbioe.2023.1173381] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/04/2023] [Indexed: 05/05/2023] Open
Abstract
Immune checkpoint blockade (ICB) typified by anti-PD-1/PD-L1 antibodies as a revolutionary treatment for solid malignancies has been limited to a subset of patients due to poor immunogenicity and inadequate T cell infiltration. Unfortunately, no effective strategies combined with ICB therapy are available to overcome low therapeutic efficiency and severe side effects. Ultrasound-targeted microbubble destruction (UTMD) is an effective and safe technique holding the promise to decrease tumor blood perfusion and activate anti-tumor immune response based on the cavitation effect. Herein, we demonstrated a novel combinatorial therapeutic modality combining low-intensity focused ultrasound-targeted microbubble destruction (LIFU-TMD) with PD-L1 blockade. LIFU-TMD caused the rupture of abnormal blood vessels to deplete tumor blood perfusion and induced the tumor microenvironment (TME) transformation to sensitize anti-PD-L1 immunotherapy, which markedly inhibited 4T1 breast cancer's growth in mice. We discovered immunogenic cell death (ICD) in a portion of cells induced by the cavitation effect from LIFU-TMD, characterized by the increased expression of calreticulin (CRT) on the tumor cell surface. Additionally, flow cytometry revealed substantially higher levels of dendritic cells (DCs) and CD8+ T cells in draining lymph nodes and tumor tissue, as induced by pro-inflammatory molecules like IL-12 and TNF-α. These suggest that LIFU-TMD as a simple, effective, and safe treatment option provides a clinically translatable strategy for enhancing ICB therapy.
Collapse
Affiliation(s)
- Nianhong Wu
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuting Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Liu
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Ultrasound, The Third People’s Hospital of Chengdu City, Chengdu, China
| | - Ying Zhou
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongye He
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Tang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Wan
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Can Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xialin Xiong
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Linhong Zhong
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Pan Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Pan Li,
| |
Collapse
|
14
|
Chen J, Huang F, Fang X, Li S, Liang Y. Silencing TLR4 using an ultrasound-targeted microbubble destruction-based shRNA system reduces ischemia-induced seizures in hyperglycemic rats. Open Life Sci 2022; 17:1689-1697. [PMID: 36619717 PMCID: PMC9795576 DOI: 10.1515/biol-2022-0526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/28/2022] [Accepted: 10/12/2022] [Indexed: 12/28/2022] Open
Abstract
The toll-like receptor 4 (TLR4) pathway is involved in seizures. We investigated whether ultrasound-targeted microbubble destruction (UTMD)-mediated delivery of short hairpin RNA (shRNA) targeting the TLR4 gene (shRNA-TLR4) can reduce ischemia-induced seizures in rats with hyperglycemia. A total of 100 male Wistar rats were randomly assigned to five groups: (1) Sham; (2) normal saline (NS); (3) shRNA-TLR4, where rats were injected with shRNA-TLR4; (4) shRNA-TLR4 + US, where rats were injected with shRNA-TLR4 followed by ultrasound (US) irradiation; and (5) shRNA-TLR4 + microbubbles (MBs) + US, where rats were injected with shRNA-TLR4 mixed with MBs followed by US irradiation. Western blot and immunohistochemical staining were used to measure TLR4-positive cells. Half of the rats in the NS group developed tonic-clonic seizures, and TLR4 expression in the CA3 region of the hippocampus was increased in these rats. In addition, the NS group showed an increased number of TLR4-positive cells compared with the Sham group, while there was a decreased number of TLR4-positive cells in the shRNA, shRNA + US, and shRNA + MBs + US groups. Our findings indicate that the TLR4 pathway is involved in the pathogenesis of ischemia-induced seizures in hyperglycemic rats and that UTMD technology may be a promising strategy to treat brain diseases.
Collapse
Affiliation(s)
- Jia Chen
- Department of Neurology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Fami Huang
- Department of Intensive Care Unit, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511500, China
| | - Xiaobo Fang
- Department of Neurology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Siying Li
- Department of Neurology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Yanling Liang
- Department of Neurology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China,Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, 510150, China
| |
Collapse
|
15
|
Ji J, Qin H, Yang Y, Wu J, Wu J. The targeting imaging and treatment capacity of gelsolin-targeted and paclitaxel-loaded PLGA nanoparticles in vitro and in vivo. Front Bioeng Biotechnol 2022; 10:933856. [PMID: 36338135 PMCID: PMC9632342 DOI: 10.3389/fbioe.2022.933856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
As a vital sign of carcinomas, lymph node metastasis is closely related to poor prognosis due to a lack of identification and effective treatment in the early stage. Nanoscale contrast agents targeting specific tumor antigens are expected to identify tumor metastasis in the early stage and achieve precise treatment. As a biomarker in the early stage of tumor invasion and metastasis, gelsolin (GSN) might be a promising molecular target to identify and screen tumor metastasis through the lymphatic system. Therefore, GSN-targeted paclitaxel-loaded poly(lactic-co-glycolic acid) nanoparticles (GSN-PTX-PLGA NPs) were prepared, and their physicochemical properties, encapsulation efficiency, drug loading, and dissolution were determined. Besides, uptake experiments and the fluorescent imaging system were used to evaluate their targeting capability. The targeting imaging and treatment capacity were also assessed by experiments in vitro and in vivo. The diameter of the GSN-PTX-PLGA NPs was 328.59 ± 3.82 nm. Hca-F cells with GSN-PLGA NPs showed stronger green fluorescence than Hca-P cells. DiI-labeled GSN-PLGA NPs in tumor-bearing mice and isolated organs exhibited more prominent fluorescence aggregation. The imaging of GSN-PLGA NPs was satisfactory in vitro, and the echo intensity gradually increased with increasing concentrations of GSN-PLGA NPs. After treatment with GSN-PTX-PLGA NPs, there was an obvious decrease in tumor volume and lymph node metastasis rate compared to the other groups (p < 0.05). In conclusion, GSN-PTX-PLGA NPs have a remarkable targeting capacity in vivo and in vitro, and they effectively inhibit tumor growth and lymph node metastasis in vivo.
Collapse
Affiliation(s)
- Jiamei Ji
- Department of Ultrasound, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Haocheng Qin
- Department of Ultrasound, Lianyungang First People’s Hospital, Lianyungang, Jiangsu, China
| | - Yan Yang
- Department of Ultrasound, Huainan First People’s Hospital, Huainan, Anhui, China
| | - Jun Wu
- Department of Ultrasound, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- *Correspondence: Jun Wu,
| | - Juan Wu
- Department of Ultrasound, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
16
|
Xu XX, Chen SY, Yi NB, Li X, Chen SL, Lei Z, Cheng DB, Sun T. Research progress on tumor hypoxia-associative nanomedicine. J Control Release 2022; 350:829-840. [PMID: 36100192 DOI: 10.1016/j.jconrel.2022.09.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 12/17/2022]
Abstract
Hypoxia at the solid tumor site is generally related to the unrestricted proliferation and metabolism of cancerous cells, which can cause tumor metastasis and aggravate tumor progression. Besides, hypoxia plays a substantial role in tumor treatment, and it is one of the main reasons that malignant tumors are difficult to cure and have a poor prognosis. On account of the tumor specific hypoxic environment, many hypoxia-associative nanomedicine have been proposed for tumor treatment. Considering the enhanced targeting effect, designing hypoxia-associative nanomedicine can not only minimize the adverse effects of drugs on normal tissues, but also achieve targeted therapy at the lesion site. Mostly, there can be three strategies for the treatment of hypoxic tumor, including improvement of hypoxic environment, hypoxia responsive drug release and hypoxia activated prodrug. The review describes the design principle and applications of tumor hypoxia-associative nanomedicine in recent years, and also explores its development trends in solid tumor treatment. Moreover, this review presents the current limitations of tumor hypoxia-associative nanomedicine in chemotherapy, radiotherapy, photodynamic therapy, sonodynamic therapy and immunotherapy, which may provide a reference for clinic translation of tumor hypoxia-associative nanomedicine.
Collapse
Affiliation(s)
- Xiao-Xue Xu
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Si-Yi Chen
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Ning-Bo Yi
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Xin Li
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Si-Lin Chen
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| | - Taolei Sun
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| |
Collapse
|