1
|
Fan S, Tang Y, Hu X, Qin M, Zhao Y, Chen X, Zou H, Gao H, Li P, Xu H, Yuan R. Efficacy and safety of ultrasound combined with microbubbles for treating retinal artery occlusion in rats. J Control Release 2025; 382:113703. [PMID: 40189055 DOI: 10.1016/j.jconrel.2025.113703] [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: 01/30/2025] [Revised: 03/20/2025] [Accepted: 04/03/2025] [Indexed: 04/16/2025]
Abstract
Retinal artery occlusion (RAO) is an eye emergency that results in severe and permanent visual impairment. The effectiveness of conventional treatment on retinal artery recanalization and vision improvement is uncertain. Ultrasound combined with microbubbles (USMB) technology is a minimally invasive transvascular drug delivery technique that has been used to investigate the treatment of stroke, myocardial infarction ,and obstructive vascular disease. The aim of this study was to investigate the efficacy and safety of USMB in the treatment of RAO. RAO model was induced by photochemical thrombosis. Normal rat eyes were treated with ultrasound at different mechanical index (MI) of 0.2,0.4 and 0.8, to explore its safety. RAO rats were randomly divided into RAO group, RAO + USMB group and RAO + US (Ultrasound) group to explore the effectiveness of the USMB in the treatment of RAO. A set of relevant ophthalmic in vivo imaging techniques was used to explore the natural history of RAO model rats while assessing tolerance and efficacy to USMB treatment. We found that blocked retinal arteries recanalized within 4-24 h in the RAO model. Retinal edema peaks within 4-24 h and resolves within 3-7 days. Blood flow density (BFD) began to recover 4 h after model induction. USMB did not cause irreversible retinal damage when the MI was below 0.4. Treatment with USMB at an MI of 0.2 significantly reduced retinal edema in RAO rats 1 day after model induction and prevented further retinal atrophy. In addition, USMB significantly increased BFD in RAO rats within 4 h and promoted the recovery of visual function. USMB can be a safe and effective treatment for RAO with protective effects on the neuroretina.
Collapse
Affiliation(s)
- Sen Fan
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yonghong Tang
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xinying Hu
- Department of Ophthalmology, Jiangyin People's Hospital, Medical School of Southeast University, Jiangyin, Jiangsu, China
| | - Mingmin Qin
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yuancheng Zhao
- Department of Ophthalmology, The General Hospital of Western Theater Command, Chengdu, China
| | - Xiaofan Chen
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Huan Zou
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Hui Gao
- 953rd Hospital, Shigatse Branch of Xinqiao Hospital, Army Medical University (Third Military Medical University), Shigatse, China
| | - Peijing Li
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Rongdi Yuan
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, China.
| |
Collapse
|
2
|
Li H, Deng Q, Cai Z, Wang Q, Huang L, Gao Y, Dong X, Sun L, Liu Z. Ultrasound Erosion of Rabbit Liver Induced by Locally Injected Phase-Shift Acoustic Droplets and With Lauromacrogol. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2025; 44:903-914. [PMID: 39835387 DOI: 10.1002/jum.16650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVES Our previous studies have found that low-frequency, low-pressure, weakly focused ultrasound (FUS) can induce acoustic droplet vaporization (ADV) of perfluoropentane (PFP) droplets and result in localized liver and prostate tissue controllable cavitation resonance and mechanical damage. To further investigate the mechanical erosion induced by ultrasound and locally injected phase-shift acoustic droplets in rabbit liver. METHODS The liver of each rabbit was treated with perfluoromethylcyclopentane (PFMCP) alone, FUS combined with PFMCP (FUS + PFMCP), and FUS combined with PFP (FUS + PFP). RESULTS Two-dimensional ultrasound images showed that immediately after the completion of FUS + PFP group treatments, a high echogenicity bubble cloud could be observed, while there were no significant differences in the PFMCP and FUS + PFMCP group before and after treatment. The liver necrotic area in the FUS + PFP group was 6.2 times that of the FUS + PFMCP group (P < .05), whereas no liver necrosis was observed in the PFMCP group. At the same time, the number of vacuoles in the liver in the FUS + PFP group was approximately 70 times that of the FUS + PFMCP group (P < .001), whereas no vacuoles were observed in the PFMCP group (P < .001). CONCLUSIONS Both FUS + PFMCP and PFMCP alone have poor mechanical erosion in liver tissue, and may even cause no damage. Only PFP droplets combined with FUS can cause significant mechanical destruction of liver tissue, leading to tissue necrosis in the droplet injection area.
Collapse
Affiliation(s)
- Hui Li
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Qingyue Deng
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Zhiping Cai
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
- Department of Ultrasound, General Hospital of Southern Theater Command, Guangzhou, China
| | - Qin Wang
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Leidan Huang
- Department of Ultrasound, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yuejuan Gao
- Department of Ultrasound, The 5th Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaoxiao Dong
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Zheng Liu
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
3
|
Li X, Tan C, Fu X, Qiu J, Shen W, Xu Z, Wu X, Zhou Y, Li X, Sun L, Qin J. Disrupting Cdc42 activation-driven filopodia formation with low-intensity ultrasound and microbubbles: A novel strategy to block ovarian cancer metastasis. Colloids Surf B Biointerfaces 2025; 253:114724. [PMID: 40300280 DOI: 10.1016/j.colsurfb.2025.114724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/04/2025] [Accepted: 04/18/2025] [Indexed: 05/01/2025]
Abstract
Metastasis is a primary cause of mortality and treatment failure in ovarian cancer, with limited effective therapeutic strategies. Low-intensity ultrasound (LIUS) and microbubbles (MBs) has been demonstrated as an adjunctive technique capable of enhancing drug delivery and suppressing tumor metastasis. However, the underlying mechanisms remain incompletely understood. In this study, we aimed to investigate whether LIUS + MBs alone could suppress tumor metastasis and to explore its mechanism of action through disruption of the cytoskeletal remodeling in filopodia, an essential structure in the early stages of cancer cell dissemination. Based on cell-based experiments to determine the optimal parameters, our results showed LIUS + MBs significantly inhibited the migration and invasion of ovarian cancer cells. In vivo, LIUS + MBs treatment markedly suppressed the overall metastasis in the orthotopic ovarian cancer model, and in both the intraperitoneal and hematogenous metastatic models established by injecting pretreated cells. Morphologically, such treatment led to a notable reduction in the length and number of filopodia, while the number of lamellipodia remained unaffected. At the molecular level, LIUS + MBs disturbed filopodia formation and the metastatic potential of ovarian cancer cells by suppressing the activation of Cdc42, a key regulator of cytoskeletal dynamics. The inhibitory effect was reversed by the overexpression of Cdc42CA. Further proteomic and bioinformatics analysis implied that LIUS + MBs may reduce Cdc42 activity by upregulating the expression of GTPase-activating proteins (GAPs). Our research provides novel insight into the mechanism by which LIUS + MBs can inhibit tumor metastasis, highlighting its role in disturbing the Cdc42-mediated cytoskeletal remodelling of filopodia.
Collapse
Affiliation(s)
- Xiaoying Li
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou 310006, China; Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Chengwei Tan
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiuxiu Fu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Jian Qiu
- Department of Obstetrics and Gynaecology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou 313000, China
| | - Wanting Shen
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Zhikang Xu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiaodong Wu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yiting Zhou
- Department of Orthopaedic Surgery and Department of Biochemistry of the Second Affiliated Hospital, Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Xiao Li
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, 310006, China; Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Hangzhou 310006, China.
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou 310006, China.
| | - Jiale Qin
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, 310006, China; Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Hangzhou 310006, China.
| |
Collapse
|
4
|
Feng Y, Qiu D, He Y, Jin H, Chen L, Xi F, Hu Z, Xie Y, Li Y, Lin M, Sun P, He Y, Liu J. Effect of ultrasound combined with microbubbles therapy on tumor hypoxic microenvironment. Front Pharmacol 2025; 15:1502349. [PMID: 39872052 PMCID: PMC11769831 DOI: 10.3389/fphar.2024.1502349] [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: 09/26/2024] [Accepted: 12/18/2024] [Indexed: 01/29/2025] Open
Abstract
Introduction Tumor tissues exhibit significantly lower oxygen partial pressure compared to normal tissues, leading to hypoxia in the tumor microenvironment and result in resistance to tumor treatments. Strategies to mitigate hypoxia include enhancing blood perfusion and oxygen supply, for example,by decomposing hydrogen peroxide within the tumor. Improving hypoxia in the tumor microenvironment could potentially improve the efficacy of cancer treatments. Previous studies have demonstrated that ultrasound of appropriate intensity when combined with microbubbles, can improve tumor blood perfusion. However, its effects on tumor hypoxia remain unclear. This study aimed to assess the effects of low-frequency non-focused ultrasound combined with microbubbles at different intensities on tumor microenvironment hypoxia and to identify the optimal ultrasound parameters for alleviating tumor hypoxia. Method Rabbits with VX2 tumors received ultrasound and microbubble treatments at different acoustic pressures and pulse repetition frequencies. The changes in tumor tissue blood perfusion before and after treatment were observed by contrast enhanced ultrasound (CEUS). The changes in tumor tissue hypoxia before and after treatment were observed by measuring oxygen partial pressure directly with in tumor tissue and immunohistochemical staining for hypoxia-inducible factor-1α (HIF-1α). Results Results indicated that low frequency, non-focused ultrasound at 0.5 MPa/20 Hz and 0.5 MPa/40 Hz, when combined with microbubbles, could increase tumor tissue blood perfusion and improve the hypoxia in tumor tissues. Discussion This study provides a new method for improving hypoxia in the tumor microenvironment (TME) which could potentially improve the cancer treatments resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Yan He
- Department of Ultrasound, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
| | - Jianhua Liu
- Department of Ultrasound, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
| |
Collapse
|
5
|
Chen J, Wang J, Yan X, Zhang X, Zhang Z, Li H, Wang Y. Enhancing the therapeutic efficacy of gefitinib on subcutaneously transplanted SKOV3 ovarian cancer tumors in nude mice via ultrasound‑stimulated microbubble cavitation. Exp Ther Med 2024; 28:336. [PMID: 39006449 PMCID: PMC11240252 DOI: 10.3892/etm.2024.12625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 05/02/2024] [Indexed: 07/16/2024] Open
Abstract
The present study aimed to explore the effect of ultrasound-stimulated microbubble cavitation (USMC) on drug concentration and therapeutic efficacy of oral gefitinib in treating subcutaneously transplanted SKOV3 ovarian cancer tumors in nude mice. The present study employed the VINNO70 ultrasonic diagnostic and treatment integrated machine for USMC therapy. Firstly, the mechanical index was set at 0.25, and the therapeutic efficacy of USMC treatment was assessed at intervals of 5, 10 and 20 min. Briefly, 72 nude mice were randomized into the following four groups (n=18/group): Control group, USMC5 min group, USMC10 min group and USMC20 min group, and the therapeutic response to USMC treatment was evaluated by comparing pre-and post-intervention effects. Additionally, the combined therapeutic efficacy of USMC and gefitinib was investigated by randomly dividing 96 tumor-bearing mice into the following four groups (n=24/group): Control group, USMC group, gefitinib group and USMC + gefitinib group. Contrast-enhanced ultrasound, hematoxylin and eosin staining, western blotting, immunofluorescence staining, TUNEL staining, ELISA and liquid chromatography-mass spectrometry were performed in the present study. The results showed that USMC combined with gefitinib had the best treatment effect; the tumor inhibition rate was higher than that of gefitinib alone and the overall survival time was prolonged. In addition, the drug concentration in the tumor tissue obtained from the USMC + gefitinib group was revealed to be ~1.4 times higher than that detected in the group treated with gefitinib alone. The experimental results also confirmed that the strongest tumor inhibition rate and longest overall survival time was observed in the USMC + gefitinib group, followed by the gefitinib group and USMC group. STAT3 is an important signaling transducer and transcription factor, which, when phosphorylated, can lead to abnormal cell proliferation and malignant transformation. In addition, the upregulation of phosphorylated (p)-STAT3 is consider a reason for the poor efficacy of gefitinib in treating ovarian cancer. The present study revealed that ultrasound microbubble therapy could overcome this side effect. In conclusion, USMC improved the effects of oral gefitinib on subcutaneously transplanted SKOV3 ovarian cancer tumors in nude mice and increased drug penetration. In addition, USMC overcame the gefitinib-induced side effect of upregulated STAT3 phosphorylation and reduced the expression levels of p-STAT3 in the tumor.
Collapse
Affiliation(s)
- Jianghong Chen
- Department of Ultrasound, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050030, P.R. China
| | - Juan Wang
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xiaonan Yan
- Department of Obstetrics and Gynecology Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xiaolin Zhang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei 050017, P.R. China
| | - Zhengzheng Zhang
- Department of Immunology, Hebei Medical University, Key Laboratory of Immune Mechanism and Intervention for Serious Diseases in Hebei Province, Shijiazhuang, Hebei 050017, P.R. China
| | - Hui Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Yueheng Wang
- Department of Cardiac Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
6
|
Xu H, Liu Z, Du M, Chen Z. Progression in low-intensity ultrasound-induced tumor radiosensitization. Cancer Med 2024; 13:e7332. [PMID: 38967145 PMCID: PMC11224918 DOI: 10.1002/cam4.7332] [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: 01/25/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Radiotherapy (RT) is a widely utilized tumor treatment approach, while a significant obstacle in this treatment modality is the radioresistance exhibited by tumor cells. To enhance the effectiveness of RT, scientists have explored radiosensitization approaches, including the use of radiosensitizers and physical stimuli. Nevertheless, several approaches have exhibited disappointing results including adverse effects and limited efficacy. A safer and more effective method of radiosensitization involves low-intensity ultrasound (LIUS), which selectively targets tumor tissue and enhances the efficacy of radiation therapy. METHODS This review summarized the tumor radioresistance reasons and explored LIUS potential radiosensitization mechanisms. Moreover, it covered diverse LIUS application strategies in radiosensitization, including the use of LIUS alone, ultrasound-targeted intravascular microbubble destruction, ultrasound-mediated targeted radiosensitizers delivery, and sonodynamic therapy. Lastly, the review presented the limitations and prospects of employing LIUS-RT combined therapy in clinical settings, emphasizing the need to connect research findings with practical applications. RESULTS AND CONCLUSION LIUS employs cost-effective equipment to foster tumor radiosensitization, curtail radiation exposure, and elevate the quality of life for patients. This efficacy is attributed to LIUS's ability to utilize thermal, cavitation, and mechanical effects to overcome tumor cell resistance to RT. Multiple experimental analyses have underscored the effectiveness of LIUS in inducing tumor radiosensitization using diverse strategies. While initial studies have shown promising results, conducting more comprehensive clinical trials is crucial to confirm its safety and effectiveness in real-world situations.
Collapse
Affiliation(s)
- Haonan Xu
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, The Affiliated Changsha Central Hospital, Hengyang Medical SchoolUniversity of South ChinaChangshaHunan ProvinceChina
- Institute of Medical Imaging, Hengyang Medical School, University of South ChinaHengyangHunan ProvinceChina
| | - Zichao Liu
- Institute of Medical Imaging, Hengyang Medical School, University of South ChinaHengyangHunan ProvinceChina
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical SchoolUniversity of South ChinaChangshaHunan ProvinceChina
| | - Meng Du
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, The Affiliated Changsha Central Hospital, Hengyang Medical SchoolUniversity of South ChinaChangshaHunan ProvinceChina
- Institute of Medical Imaging, Hengyang Medical School, University of South ChinaHengyangHunan ProvinceChina
| | - Zhiyi Chen
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, The Affiliated Changsha Central Hospital, Hengyang Medical SchoolUniversity of South ChinaChangshaHunan ProvinceChina
- Institute of Medical Imaging, Hengyang Medical School, University of South ChinaHengyangHunan ProvinceChina
| |
Collapse
|
7
|
Han F, Wang Y, Dong X, Lin Q, Wang Y, Gao W, Yun M, Li Y, Gao S, Huang H, Li N, Luo T, Luo X, Qiu M, Zhang D, Yan K, Li A, Liu Z. Clinical sonochemotherapy of inoperable pancreatic cancer using diagnostic ultrasound and microbubbles: a multicentre, open-label, randomised, controlled trial. Eur Radiol 2024; 34:1481-1492. [PMID: 37796294 DOI: 10.1007/s00330-023-10210-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/02/2023] [Accepted: 07/04/2023] [Indexed: 10/06/2023]
Abstract
OBJECTIVES Sonochemotherapy, which uses microbubble (MB)-assisted ultrasound (US) to deliver chemotherapeutic agents, has the potential to enhance tumour chemotherapy. The combination of US and MB has been demonstrated to prolong the survival of patients with pancreatic cancer. This phase 2 clinical trial aimed to determine the clinical efficacy and safety of sonochemotherapy for inoperable pancreatic ductal adenocarcinoma by using US and MB. METHODS Eighty-two patients with stage III or IV pancreatic cancer were recruited from July 2018 to March 2021 and followed up until September 2022. US treatment was performed with a modified diagnostic US scanner for 30 min after chemotherapeutic infusion. The primary endpoint was overall survival (OS), and the secondary endpoints were Eastern Cooperative Oncology Group (ECOG) status < 2, progression-free survival (PFS), disease control rate (DCR), and adverse events. RESULTS Seventy-eight patients were randomly allocated (40 to chemotherapy and 38 to sonochemotherapy). The median OS was longer with sonochemotherapy than with chemotherapy (9.10 vs. 6.10 months; p = 0.037). The median PFS with sonochemotherapy was 5.50 months, compared with 3.50 months (p = 0.080) for chemotherapy. The time of ECOG status < 2 was longer with sonochemotherapy (7.20 months) than with chemotherapy (5.00 months; p = 0.029). The DCR was 73.68% for sonochemotherapy compared with 42.50% for the control (p = 0.005). The incidence of overall adverse events was balanced between the two groups. CONCLUSIONS The use of sonochemotherapy can extend the survival and well-being time of stage III or IV pancreatic cancer patients without any increase in serious adverse events. TRIAL REGISTRATION ChineseClinicalTrials.gov ChiCTR2100044721 CLINICAL RELEVANCE STATEMENT: This multicentre, randomised, controlled trial has proven that sonochemotherapy, namely, the combination of diagnostic ultrasound, microbubbles, and chemotherapy, could extend the overall survival of patients with end-stage pancreatic ductal adenocarcinoma from 6.10 to 9.10 months without increasing any serious adverse events. KEY POINTS • This is the first multicentre, randomised, controlled trial of sonochemotherapy for clinical pancreatic cancer treatment using ultrasound and a commercial ultrasound contrast agent. • Sonochemotherapy extended the median overall survival from 6.10 (chemotherapy alone) to 9.10 months. • The disease control rate increased from 42.50% with chemotherapy to 73.68% with sonochemotherapy.
Collapse
Affiliation(s)
- Feng Han
- Department of Ultrasound, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, 651 Dongfengdong Road, Guangzhou, 510060, China
| | - Yanjie Wang
- Department of Ultrasound, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 of Fucheng Road, Haidian District, Beijing, 100142, China
| | - Xiaoxiao Dong
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Qingguang Lin
- Department of Ultrasound, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, 651 Dongfengdong Road, Guangzhou, 510060, China
| | - Yixi Wang
- Department of Ultrasound, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 of Fucheng Road, Haidian District, Beijing, 100142, China
| | - Wenhong Gao
- Department of Ultrasound, General Hospital of Central Theater, Wuhan, China
| | - Miao Yun
- Department of Ultrasound, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, 651 Dongfengdong Road, Guangzhou, 510060, China
| | - Yan Li
- Department of Gastrointestinal Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Shunji Gao
- Department of Ultrasound, General Hospital of Central Theater, Wuhan, China
| | - Huilong Huang
- Department of Ultrasound, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ningshan Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Tingting Luo
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiao Luo
- Department of Radiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Miaozhen Qiu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Dongsheng Zhang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kun Yan
- Department of Ultrasound, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 of Fucheng Road, Haidian District, Beijing, 100142, China.
| | - Anhua Li
- Department of Ultrasound, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, 651 Dongfengdong Road, Guangzhou, 510060, China.
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
8
|
Li R, Lu L, Huang Z, Gao Y. Downregulation of carbonic anhydrase IX expression in mouse xenograft nasopharyngeal carcinoma model via doxorubicin nanobubble combined with ultrasound. Open Med (Wars) 2024; 19:20240910. [PMID: 38463523 PMCID: PMC10921437 DOI: 10.1515/med-2024-0910] [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: 08/25/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 03/12/2024] Open
Abstract
The purpose of this study was to investigate whether doxorubicin nanobubbles (DOX-NB) combined with diagnostic ultrasound (DUS) irradiation could downregulate the expression of carbonic anhydrase IX (CAIX) in mouse xenograft nasopharyngeal carcinoma (NPC) model. In this study, the prepared DOX-NB was round and well dispersed. The average diameter of DOX-NB was 250.9 ± 50.8 nm, with an average polydispersity of 0.321 ± 0.05. The cumulative release of DOX in the DOX-NB + DUS group was significantly higher compared with that of the DOX-NB group (p < 0.05). DOX-NB combined with DUS irradiation could significantly inhibit cell viability (p < 0.05). The expression of CAIX and microvessel density (MVD) in the xenografted tumors was the lowest in the DOX-NB + DUS group compared with that of other groups (p < 0.05). In conclusion, DOX-NB combined with DUS irradiation could improve DOX-NB drug release and synergistically inhibit NPC cell activity. DOX-NB combined with DUS irradiation can downregulate the expression of CAIX in mouse xenograft NPC model. This may be due to the synergistic effect of DUS combined with DOX-NB in reducing MVD in NPC.
Collapse
Affiliation(s)
- Rong Li
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, 530021Guangxi, China
| | - Liugui Lu
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, 530021Guangxi, China
| | - Zhaoxi Huang
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, 530021Guangxi, China
| | - Yong Gao
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Rd, Nanning, 530021Guangxi, China
| |
Collapse
|
9
|
Bai L, Luo T, Tang J, Zhang J, Tan X, Tang J, Huang L, Dong X, Li N, Li P, Liu Z. Ultrasound-Induced Tumor Perfusion Changes and Doxorubicin Delivery: A Study on Pulse Length and Pulse Repetition Frequency. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:253-263. [PMID: 37853950 DOI: 10.1002/jum.16355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/26/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023]
Abstract
OBJECTIVES To investigate the appropriate combination of pulse length (PL) and pulse repetition frequency (PRF) when performing ultrasound stimulated microbubble (USMB) to enhance doxorubicin (DOX) delivery to tumors. METHODS A total of 48 tumor-bearing mice were divided into four groups, namely groups A-D. The mice in groups B-D were treated with chemotherapy and USMB treatment with different combinations of PL and PRF, and group A was control. Contrast-enhanced ultrasound imaging was conducted to analyze tumor blood perfusion. Fluorescence microscopy and high-performance liquid chromatography were used to qualitatively and quantitatively analyse DOX release. The structural changes of tumors were observed under light microscope and transmission electron microscope. Furthermore, another 24 tumor-bearing mice were treated with sonochemotherapy and some related inflammatory factors were measured to explore the underlying mechanism. RESULTS With PL of three cycles and PRF of 2 kHz, the tumor perfusion area ratio increased by 26.67%, and the DOX concentration was 4.69 times higher than the control (P < .001). With PL of 34.5 cycles and PRF of 200 Hz, the tumor perfusion area ratio decreased by 12.7% and DOX did not exhibit increased extravasation compared with the control. Microvascular rupture and hemorrhage were observed after long PL and low PRF treatment. While vasodilation and higher levels of some vasodilator inflammatory factors were found after treatment with short PL and high PRF. CONCLUSIONS USMB treatment using short PL and high PRF could enhance tumor blood perfusion and increase DOX delivery, whereas long PL and low PRF could not serve the same purpose.
Collapse
Affiliation(s)
- Luhua Bai
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Tingting Luo
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Jiawei Tang
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Jing Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xi Tan
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Junhui Tang
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Leidan Huang
- Department of Ultrasound, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Xiaoxiao Dong
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Ningshan Li
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Peijing Li
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Zheng Liu
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
10
|
Zhang Y, Zhang J, Luo T, Cai Z, Yang G, Li H, Wei J, Zhu Q, Li P, Dong X, Liu Z. Sononeoperfusion effect by ultrasound and microbubble promotes nitric oxide release to alleviate hypoxia in a mouse MC38 tumor model. ULTRASONICS SONOCHEMISTRY 2023; 100:106619. [PMID: 37757603 PMCID: PMC10550768 DOI: 10.1016/j.ultsonch.2023.106619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/02/2023] [Accepted: 09/22/2023] [Indexed: 09/29/2023]
Abstract
Tumor hypoperfusion not only impedes therapeutic drug delivery and accumulation, but also leads to a hypoxic and acidic tumor microenvironment, resulting in tumor proliferation, invasion, and therapeutic resistance. Sononeoperfusion effect refers to tumor perfusion enhancement using ultrasound and microbubbles. This study aimed to further investigate hypoxia alleviation by sononeoperfusion effect and explore the characteristics and mechanism of sononeoperfusion effect. To stimulate the sononeoperfusion effect, mice bearing MC38 colon cancers were included in this study and diagnostic ultrasound for therapy was set at a mechanical index (MI) of 0.1, 0.3, and 0.5, frequency of 3 MHz, pulse length of 5 cycles, and pulse repetition frequency of 2000 Hz. The results demonstrated that a single ultrasound and microbubble (USMB) treatment resulted in tumor perfusion enhancement at MI = 0.3, and nitric oxide (NO) concentration increased at MI = 0.3/0.5 (P < 0.05). However, there were no significant difference in the hypoxia-inducible factor-1α (HIF-1α) or D-lactate (D-LA) (P > 0.05) levels. Multiple sononeoperfusion effects were observed at MI = 0.3/0.5 (P < 0.05). For each treatment, USMB slightly but steadily improved the tumor tissue oxygen partial pressure (pO2) during and post treatment. It alleviated tumor hypoxia by decreasing HIF-1α, D-LA level and the hypoxic immunofluorescence intensity at MI = 0.3/0.5 (P < 0.05). The sononeoperfusion effect was not stimulated after eNOS inhibition. In conclusion, USMB with appropriate MI could lead to a sononeoperfusion effect via NO release, resulting in hypoxia amelioration. The tumors were not resistant to multiple sononeoperfusion effects. Repeated sononeoperfusion is a promising approach for relieving tumor hypoxia and resistance to therapy.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jing Zhang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Tingting Luo
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zhiping Cai
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China; Department of Ultrasound, General Hospital of Guangzhou Military Command of PLA, Guangzhou, China
| | - Guoliang Yang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Hui Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Junshuai Wei
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China; Department of Ultrasound, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China
| | - Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China; Department of Ultrasound, 953th Hospital, Shigatse Branch, Xinqiao Hospital, Army Medical University (Third Military Medical University), Shigatse, China
| | - Peijing Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiaoxiao Dong
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China.
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
11
|
Li H, He H, Tang J, Luo T, Yang G, Huang L, Dong X, Liu Z. A new sonoablation using acoustic droplet vaporization and focused ultrasound: A feasibility study. Med Phys 2023; 50:6663-6672. [PMID: 37731063 DOI: 10.1002/mp.16742] [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: 03/08/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Histotripsy and boiling histotripsy are two methods of mechanical ablation that use high-pressure focused ultrasound (FUS). PURPOSE Here, a new bubble sonoablation technique was investigated using low-pressure FUS in combination with local injection of perfluoropentane (PFP) in rabbit liver. METHODS Fifteen healthy New Zealand white rabbits were treated with FUS alone, FUS + PFP or PFP alone. FUS was performed using a single-element focused transducer (frequency 596 kHz, 0.27 ms pulses, 0.54% duty cycle, and peak negative pressure 2.0 MPa). Ten minutes before FUS treatment, the PFP droplet was locally injected into the rabbit liver, where the ultrasound was focused. Contrast-enhanced ultrasound (CEUS) of the liver was performed, and the temperature at the liver surface in the targeted liver region was recorded during treatment. The livers were collected for pathological examination. Statistical significance was set at p < 0.05. Paired t-tests were used to compare the pre- and post-treatment values. One-way analysis of variance was performed to compare multiple groups, and the least significant difference method was used for further comparisons between the two groups. RESULTS Analysis of CEUS data showed that the values of area under the curve (AUC) were significantly different in the PFP + FUS group pre- (10453.644 ± 1182.93) and post-treatment (4058.098 ± 2720.41), and the AUC values of PFP + FUS post-treatment (4058.098 ± 2720.41) were also significantly lower than those of the FUS (9946.694 ± 1071.54) and the PFP (10364.794 ± 2181.53) groups. The peak intensity values also showed the same results, the value of peak intensity of PFP+FUS post-treatment was 82.958 ± 13.99, whereas there was no difference between FUS (106.61 ± 7.61) and PFP (104.136 ± 10.55). Hematoxylin and eosin (H&E) staining revealed that the pathological damage ratings of the PFP + FUS, PFP, and FUS groups were grade 3, grade 1, and grade 0, respectively. Specifically, the area of liver necrosis in the PFP + FUS group (0.99 ± 0.29 cm2 ) was 198 times higher than that in the PFP group (0.005 ± 0.008 cm2 ), whereas no necrosis was observed in the livers treated with FUS alone. Simultaneously, the number of vacuoles in the liver of the PFP + FUS group (35.50 ± 23.31) was approximately five times that of the PFP group (7.00 ± 12.88), whereas no vacuoles were found in the liver treated with FUS alone. CONCLUSION PFP droplets combined with FUS can destroy liver tissue and cause tissue necrosis in the droplet injection area, without affecting the structure of surrounding tissue.
Collapse
Affiliation(s)
- Hui Li
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Huan He
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Jiawei Tang
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Tingting Luo
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Guoliang Yang
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Leidan Huang
- Department of Ultrasound, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiaoxiao Dong
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Zheng Liu
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
12
|
Drzał A, Delalande A, Dziurman G, Pichon C, Swakoń J, Elas M. Ultrasound sensitive O 2 microbubbles radiosensitize murine breast cancer but lead to higher metastatic spread. Free Radic Biol Med 2023; 199:166-176. [PMID: 36858326 DOI: 10.1016/j.freeradbiomed.2023.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/06/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023]
Abstract
The inadequate level of oxygenation in tumors has been shown to correlate not only with greater invasiveness of cancer cells, but also with a reduction in their sensitivity to anticancer therapies. Over the years, many attempts have been made to increase the oxygenation level of cancer, but most of them have been ineffective. We investigated the heterogeneous response of tumor tissue to phospholipid-coated oxygen microbubbles (OMB) in murine tumors in vivo using oxygen and hemoglobin saturation mapping and the influence of OMB treatment on microvasculature, perfusion, and radiotherapy effectiveness. Intravenous administration of OMB followed by ultrasound pulse leads to increased oxygenation of a tumor, found mainly in the vicinity of tumor vessels, while intratumoral delivery resulted in areas of increased pO2 more evenly distributed within the tumor. Furthermore, hemoglobin contributes little to the increase in tumor oxygenation caused by oxygen microbubbles. Extensive vasculature disruption was observed in the groups treated with both oxygen/nitrogen microbubbles and ultrasound pulse. This therapy also led to a reduction in the coverage of the vessels by pericytes, while the density of the microvessels was unchanged. Radiotherapy with a single dose of 12Gy reduced tumor growth by 50% in all treated groups. Unfortunately, at the same time, the number of macroscopic metastases in the lungs increased significantly after intravenous administration of oxygen/nitrogen microbubbles and the application of an ultrasound pulse. In conclusion, ultrasound-sensitive oxygen microbubbles are effective in delivering oxygen to tumor tissue, thus increasing the effectiveness of radiotherapy. However, cavitation effects and destruction of the integrity of tumor vessels result in greater spread of cancer cells in the host organism.
Collapse
Affiliation(s)
- Agnieszka Drzał
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Kraków, Poland
| | - Anthony Delalande
- University of Orleans, 45067, Orleans, France; Center for Molecular Biophysics, CNRS Orleans, 45071, Orleans, France
| | - Gabriela Dziurman
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Kraków, Poland
| | - Chantal Pichon
- University of Orleans, 45067, Orleans, France; Institut Universitaire de France, 75231, Paris, France; Center for Molecular Biophysics, CNRS Orleans, 45071, Orleans, France
| | - Jan Swakoń
- Institute of Nuclear Physics, Polish Academy of Sciences (IFJ PAN), Krakow, Poland
| | - Martyna Elas
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Kraków, Poland.
| |
Collapse
|
13
|
Tang N, Tang J, Tang J, Zhu Q, Dong X, Zhang Y, Li N, Liu Z. Sononeoperfusion: a new therapeutic effect to enhance tumour blood perfusion using diagnostic ultrasound and microbubbles. Cancer Imaging 2023; 23:29. [PMID: 36959681 PMCID: PMC10035258 DOI: 10.1186/s40644-023-00545-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 03/12/2023] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND Hypoperfusion or resultant hypoxia in solid tumours is a main reason for therapeutic resistance. Augmenting the blood perfusion of hypovascular tumours might improve both hypoxia and drug delivery. Cavitation is known to result in microstreaming and sonoporation and to enhance drug diffusion into tumours. Here, we report the ability to enhance both tumour blood perfusion and doxorubicin (Dox) delivery using a new sononeoperfusion effect causing a cavitation effect on tumour perfusion in subcutaneous Walker-256 tumours of rats using ultrasound stimulated microbubble (USMB). METHODS To induce the sononeoperfusion effect, USMB treatment was performed with a modified diagnostic ultrasound (DUS) system and SonoVue® microbubbles. The therapeutic pulse was operated with a peak negative pressure of 0.26 to 0.32 MPa and a pulse repetition frequency (PRF) of 50 Hz to 2 kHz. Contrast-enhanced ultrasound (CEUS) was used for tumour perfusion assessment. RESULTS The USMB treatment of 0.26 MPa and 1 kHz could significantly enhance tumour perfusion with a 20.29% increase in the CEUS peak intensity and a 21.42% increment in the perfusion area for more than 4 hours (P < 0.05). The treatment also increased Dox delivery to tumours by approximately 3.12-fold more than that of the control (P < 0.05). Furthermore, ELISAs showed that vasodilators and inflammatory factors increased 4 hours after treatment (P < 0.05), suggesting that the inflammatory response plays an important role in the sononeoperfusion effect. CONCLUSION The USMB-induced sononeoperfusion effect could significantly enhance the blood perfusion of Walker-256 tumours and promote drug delivery. It might be a novel physical method for overcoming the therapeutic resistance of hypoperfused or hypoxic tumours.
Collapse
Affiliation(s)
- Najiao Tang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, 83 Xinqiao Street, Chongqing, 400037, PR China
| | - Jiawei Tang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, 83 Xinqiao Street, Chongqing, 400037, PR China
| | - Junhui Tang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, 83 Xinqiao Street, Chongqing, 400037, PR China
| | - Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, 83 Xinqiao Street, Chongqing, 400037, PR China
| | - Xiaoxiao Dong
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, 83 Xinqiao Street, Chongqing, 400037, PR China
| | - Yi Zhang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, 83 Xinqiao Street, Chongqing, 400037, PR China.
| | - Ningshan Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, 83 Xinqiao Street, Chongqing, 400037, PR China
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, 83 Xinqiao Street, Chongqing, 400037, PR China.
| |
Collapse
|
14
|
Ho YJ, Hsu HC, Wu BH, Lin YC, Liao LD, Yeh CK. Preventing ischemia-reperfusion injury by acousto-mechanical local oxygen delivery. J Control Release 2023; 356:481-492. [PMID: 36921723 DOI: 10.1016/j.jconrel.2023.03.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 02/28/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023]
Abstract
Ischemia-reperfusion (I/R) injury is a pathological process that causes vascular damage and dysfunction which increases recurrence and/or mortality in myocardial infarction, ischemic stroke, and organ transplantation. We hypothesized that ultrasound-stimulated oxygen-loaded microbubble (O2-MB) cavitation would enhance mechanical force on endothelium and simultaneously release oxygen locally at the targeted vessels. This cooperation between biomechanical and biochemical stimuli might modulate endothelial metabolism, providing a potential clinical approach to the prevention of I/R injury. Murine hindlimb and cardiac I/R models were used to demonstrate the feasibility of injury prevention by O2-MB cavitation. Increased mechanical force on endothelium induced eNOS-activated vasodilation and angiogenesis to prevent re-occlusion at the I/R vessels. Local oxygen therapy increased endothelial oxygenation that inhibited HIF-1α expression, increased ATP generation, and activated cyclin D1 for cell repair. Moreover, a decrease in interstitial H2O2 level reduced the expression of caspase3, NFκB, TNFα, and IL6, thus ameliorating inflammatory responses. O2-MB cavitation showed efficacy in maintaining cardiac function and preventing myocardial fibrosis after I/R. Finally, we present a potential pathway for the modulation of endothelial metabolism by O2-MB cavitation in relation to I/R injury, wound healing, and vascular bioeffects.
Collapse
Affiliation(s)
- Yi-Ju Ho
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.
| | - Hui-Ching Hsu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Bing-Huan Wu
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Chun Lin
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan; Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Lun-De Liao
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
15
|
Luo T, Bai L, Zhang Y, Huang L, Li H, Gao S, Dong X, Li N, Liu Z. Optimal treatment occasion for ultrasound stimulated microbubbles in promoting gemcitabine delivery to VX2 tumors. Drug Deliv 2022; 29:2796-2804. [PMID: 36047064 PMCID: PMC9448370 DOI: 10.1080/10717544.2022.2115163] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Ultrasound stimulated microbubbles (USMB) is a widely used technology that can promote chemotherapeutic delivery to tumors yet the best treatment occasion for USMB is unknown or ignored. We aimed to determine the optimal treatment occasion for USMB treatment to enhance tumor chemotherapy to achieve the highest drug concentration in tumors. Experiments were conducted on VX2 tumors implanted in 60 rabbits. Gemcitabine (GEM) was intravenously infused as a chemotherapeutic agent and USMB was administered before, during or after chemotherapy. USMB was conducted with a modified diagnostic ultrasound at 3 MHz employing short bursts (5 cycles and 0.125% duty cycle) at 0.26 MPa in combination with a lipid microbubble. Subsequently, tumor blood perfusion quantitation, drug concentration detection, and fluorescence microscopy were performed. The results showed that the group that received USMB treatment immediately after GEM infusion had the highest drug concentration in tumors, which was 2.83 times that of the control group. Fifteen tumors were then treated repeatedly with the optimal USMB-plus-GEM combination, and along with the GEM and the control groups, were studied for tumor growth, tumor cell proliferation, apoptosis, and related cytokine contents. The combined treatment significantly inhibited tumor growth and promoted apoptosis. The levels of related cytokines, including HIF-1α, decreased after six combination therapies. These results suggest that the optimal treatment occasion for USMB occurs immediately after chemotherapy and tumor hypoxia improves after multiple combination therapies.
Collapse
Affiliation(s)
- Tingting Luo
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Luhua Bai
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yi Zhang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Leidan Huang
- Department of Ultrasound, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Hui Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Shunji Gao
- Department of Ultrasound, General Hospital of Central Theatre Command, Wuhan, China
| | - Xiaoxiao Dong
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ningshan Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
16
|
Cheng Z, Yang Q, He H, Li R, Li X, Jiang H, Zhao X, Li J, Wang L, Zhou S, Zhang S. Intravoxel incoherent motion diffusion-weighted imaging and shear wave elastography for evaluating peritumoral liver fibrosis after transarterial chemoembolization in a VX2 rabbit liver tumor model. Front Physiol 2022; 13:893925. [PMID: 36311244 PMCID: PMC9597251 DOI: 10.3389/fphys.2022.893925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
In this study, we sought to evaluate changes in peritumoral fibrosis after transarterial chemoembolization (TACE) in a rabbit VX2 liver tumor model using intravoxel incoherent motion diffusion-weighted imaging (IVIM DWI) and ultrasound shear wave elastography (SWE). A total of 20 rabbits underwent implantation of VX2 tumor tissues in the left lobe of the liver. The rabbits were randomly divided into an experimental group (n = 10) or a control group (n = 10). Those in the experimental group were treated with an emulsion of lipiodol and pirarubicin through a microcatheter 2–3 weeks after implantation; those in the control group were treated with sterile water. Compared with the control group, the true diffusion coefficient (D) and pseudodiffusion coefficient (D*) values in liver tissues were significantly lower (p < 0.05 for all) and liver stiffness values (LSV) (10.58 ± 0.89 kPa) were higher in the experimental group (7.65 ± 0.86 kPa; p < 0.001). The median stage of liver fibrosis based on METAVIR scores was 1 (1,1) in the control group and 2 (2,3) in the experimental group (Z = 4.15, p < 0.001). D, D*, and LSV were significantly correlated with pathologic staining in the assessment of liver fibrosis (r = −0.54 p = 0.015; r = −0.50, p = 0.025; r = 0.91, p < 0.001; respectively). These data suggest that TACE aggravates liver injury and liver fibrosis, especially surrounding the tumor, in a rabbit VX2 liver tumor model. IVIM DWI and SWE can be used to evaluate the change in liver fibrosis.
Collapse
Affiliation(s)
- Zhimei Cheng
- Institute of Image, Guizhou Medical University, Guiyang, China
| | - Qin Yang
- Institute of Image, Guizhou Medical University, Guiyang, China
| | - Huizhou He
- Institute of Image, Guizhou Medical University, Guiyang, China
| | - Ran Li
- Institute of Image, Guizhou Medical University, Guiyang, China
| | - Xueying Li
- Institute of Image, Guizhou Medical University, Guiyang, China
| | - Hongyu Jiang
- GCP Institution Office, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Xuya Zhao
- Department of Interventional Radiology, the Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Junxiang Li
- Institute of Image, Guizhou Medical University, Guiyang, China
- Department of Interventional Radiology, the Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Lizhou Wang
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, China Branch of National Clinical Research Center for Interventional Medicine, Guiyang, China
| | - Shi Zhou
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, China Branch of National Clinical Research Center for Interventional Medicine, Guiyang, China
- *Correspondence: Shi Zhou, ; Shuai Zhang,
| | - Shuai Zhang
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, China Branch of National Clinical Research Center for Interventional Medicine, Guiyang, China
- *Correspondence: Shi Zhou, ; Shuai Zhang,
| |
Collapse
|
17
|
Sharma D, Czarnota GJ. Involvement of Ceramide Signalling in Radiation-Induced Tumour Vascular Effects and Vascular-Targeted Therapy. Int J Mol Sci 2022; 23:ijms23126671. [PMID: 35743121 PMCID: PMC9223569 DOI: 10.3390/ijms23126671] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 02/04/2023] Open
Abstract
Sphingolipids are well-recognized critical components in several biological processes. Ceramides constitute a class of sphingolipid metabolites that are involved in important signal transduction pathways that play key roles in determining the fate of cells to survive or die. Ceramide accumulated in cells causes apoptosis; however, ceramide metabolized to sphingosine promotes cell survival and angiogenesis. Studies suggest that vascular-targeted therapies increase endothelial cell ceramide resulting in apoptosis that leads to tumour cure. Specifically, ultrasound-stimulated microbubbles (USMB) used as vascular disrupting agents can perturb endothelial cells, eliciting acid sphingomyelinase (ASMase) activation accompanied by ceramide release. This phenomenon results in endothelial cell death and vascular collapse and is synergistic with other antitumour treatments such as radiation. In contrast, blocking the generation of ceramide using multiple approaches, including the conversion of ceramide to sphingosine-1-phosphate (S1P), abrogates this process. The ceramide-based cell survival "rheostat" between these opposing signalling metabolites is essential in the mechanotransductive vascular targeting following USMB treatment. In this review, we aim to summarize the past and latest findings on ceramide-based vascular-targeted strategies, including novel mechanotransductive methodologies.
Collapse
Affiliation(s)
- Deepa Sharma
- Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
- Correspondence: ; Tel.: +1-416-480-6100 (ext. 89533)
| | - Gregory J. Czarnota
- Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
| |
Collapse
|
18
|
Thies M, Oelze ML. Combined Therapy Planning, Real-Time Monitoring, and Low Intensity Focused Ultrasound Treatment Using a Diagnostic Imaging Array. IEEE TRANSACTIONS ON MEDICAL IMAGING 2022; 41:1410-1419. [PMID: 34986094 PMCID: PMC9199060 DOI: 10.1109/tmi.2021.3140176] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Low intensity focused ultrasound (FUS) therapies use low intensity focused ultrasound waves, typically in combination with microbubbles, to non-invasively induce a variety of therapeutic effects. FUS therapies require pre-therapy planning and real-time monitoring during treatment to ensure the FUS beam is correctly targeted to the desired tissue region. To facilitate more streamlined FUS treatments, we present a system for pre-therapy planning, real-time FUS beam visualization, and low intensity FUS treatment using a single diagnostic imaging array. Therapy planning was accomplished by manually segmenting a B-mode image captured by the imaging array and calculating a sonication pattern for the treatment based on the user-input region of interest. For real-time monitoring, the imaging array transmitted a visualization pulse which was focused to the same location as the FUS therapy beam and ultrasonic backscatter from this pulse was used to reconstruct the intensity field of the FUS beam. The therapy planning and beam monitoring techniques were demonstrated in a tissue-mimicking phantom and in a rat tumor in vivo while a mock FUS treatment was carried out. The FUS pulse from the imaging array was excited with an MI of 0.78, which suggests that the array could be used to administer select low intensity FUS treatments involving microbubble activation.
Collapse
|