1
|
McCorkell G, Piva T, Highgate D, Nakayama M, Geso M. Ultrasound-stimulated microbubbles to enhance radiotherapy: A scoping review. J Med Imaging Radiat Oncol 2024; 68:740-769. [PMID: 39250692 DOI: 10.1111/1754-9485.13740] [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: 02/24/2024] [Accepted: 07/22/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Primarily used as ultrasound contrast agents, microbubbles have recently emerged as a versatile therapeutic vector that can be 'burst' to deliver payloads in the presence of suitably optimised ultrasound fields. Ultrasound-stimulated microbubbles (USMB) have recently demonstrated improvements in treatment outcomes across a variety of clinical applications. This scoping review investigates whether this potential translates into the context of radiation therapy by evaluating the application of this technology across all three phases of radiation action. METHODS Primary research articles, excluding poster presentations and conference proceedings, were identified through systematic searches of the PubMed NCBI/Medline, Embase/OVID, Web of Science and CINAHL/EBSCOhost databases, with additional articles identified via manual Google Scholar searching. Articles were dual screened for inclusion using the Covidence systematic review platform and classified against all three phases of radiation action. RESULTS Overall, 57 eligible publications from a total of 1389 identified articles were included in the review, with studies dating back to 2012. Study heterogeneity prevented formal statistical analysis; however, most articles reported improved outcomes using USMB in the presence of radiation compared to that of radiation alone. These improvements appear to result from the use of USMB as either a biovascular disruptor causing tumour cell damage via indirect mechanisms, or as a localised treatment vector that directly increases tumour cell uptake of other therapeutic and physical agents designed to enhance radiation action. CONCLUSIONS USMB demonstrate exciting potential to enhance the effects of radiation treatments due to their versatility and capacity to target all three phases of radiation action.
Collapse
Affiliation(s)
- Giulia McCorkell
- RMIT University, Melbourne, Victoria, Australia
- The University of Melbourne, Melbourne, Victoria, Australia
| | | | | | - Masao Nakayama
- RMIT University, Melbourne, Victoria, Australia
- Kobe University, Kobe, Hyogo, Japan
- Kita-Harima Medical Center, Ono, Hyogo, Japan
| | - Moshi Geso
- RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Wu SY, Wang CH, Kang ST, Yu CF, Chen FH, Chiang CS. Power-Doppler-based NH002 microbubble sonoporation with chemotherapy relieves hypoxia and enhances the efficacy of chemotherapy and immunotherapy for pancreatic tumors. Sci Rep 2024; 14:8532. [PMID: 38830912 PMCID: PMC11148017 DOI: 10.1038/s41598-024-54432-y] [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: 11/07/2023] [Accepted: 02/13/2024] [Indexed: 06/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses challenges due to late-stage diagnosis and limited treatment response, often attributed to the hypoxic tumor microenvironment (TME). Sonoporation, combining ultrasound and microbubbles, holds promise for enhancing therapy. However, additional preclinical research utilizing commercially available ultrasound equipment for PDAC treatment while delving into the TME's intricacies is necessary. This study investigated the potential of using a clinically available ultrasound system and phase 2-proven microbubbles to relieve tumor hypoxia and enhance the efficacy of chemotherapy and immunotherapy in a murine PDAC model. This approach enables early PDAC detection and blood-flow-sensitive Power-Doppler sonoporation in combination with chemotherapy. It significantly extended treated mice's median survival compared to chemotherapy alone. Mechanistically, this combination therapy enhanced tumor perfusion and substantially reduced tumor hypoxia (77% and 67%, 1- and 3-days post-treatment). Additionally, cluster of differentiation 8 (CD8) T-cell infiltration increased four-fold afterward. The combined treatment demonstrated a strengthening of the anti-programmed death-ligand 1(αPDL1) therapy against PDAC. Our study illustrates the feasibility of using a clinically available ultrasound system with NH-002 microbubbles for early tumor detection, alleviating hypoxic TME, and improving chemotherapy and immunotherapy. It suggests the development of an adjuvant theragnostic protocol incorporating Power-Doppler sonoporation for pancreatic tumor treatment.
Collapse
Affiliation(s)
- Sheng-Yan Wu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | | | | | - Ching-Fang Yu
- Research Center for Radiation Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan, 33382, Taiwan
| | - Fang-Hsin Chen
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, 30013, Taiwan.
- The BNCT Research Center, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan.
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, 30013, Taiwan.
- The BNCT Research Center, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
3
|
Qi N, Zhou X, Ma N, Zhang J, Wang Z, Zhang X, Li A. Integrin αvβ3 and LHRH Receptor Double Directed Nano-Analogue Effective Against Ovarian Cancer in Mice Model. Int J Nanomedicine 2024; 19:3071-3086. [PMID: 38562611 PMCID: PMC10984207 DOI: 10.2147/ijn.s442921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction The high mortality rate of malignant ovarian cancer is attributed to the absence of effective early diagnosis methods. The LHRH receptor is specifically overexpressed in most ovarian cancers, and the integrin αvβ3 receptor is also overexpressed on the surface of ovarian cancer cells. In this study, we designed LHRH analogues (LHRHa)/RGD co-modified paclitaxel liposomes (LHRHa-RGD-LP-PTX) to target LHRH receptor-positive ovarian cancers more effectively and enhance the anti-ovarian cancer effects. Methods LHRHa-RGD-LP-PTX liposomes were prepared using the thin film hydration method. The morphology, physicochemical properties, cellular uptake, and cell viability were assessed. Additionally, the cellular uptake mechanism of the modified liposomes was investigated using various endocytic inhibitors. The inhibitory effect of the formulations on tumor spheroids was observed under a microscope. The co-localization with lysosomes was visualized using confocal laser scanning microscopy (CLSM), and the in vivo tumor-targeting ability of the formulations was assessed using the IVIS fluorescent imaging system. Finally, the in vivo anti-tumor efficacy of the formulations was evaluated in the armpits of BALB/c nude mice. Results The results indicated that LHRHa-RGD-LP-PTX significantly enhanced cellular uptake in A2780 cells, increased cytotoxicity, and hand a more potent inhibitory effect on tumor spheroids of A2780 cells. It also showed enhanced co-localization with endosomes or lysosome in A2780 cells, improved tumor-targeting capability, and demonstrated an enhanced anti-tumor effect in LHRHR-positive ovarian cancers. Conclusion The designed LHRHa-RGD-LP-PTX liposomes significantly enhanced the tumor-targeting ability and therapeutic efficacy for LHRH receptor-positive ovarian cancers.
Collapse
Affiliation(s)
- Na Qi
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, People's Republic of China
| | - Xiantai Zhou
- Department of Pharmacy, Guilin Medical University, Guilin, 541004, People's Republic of China
| | - Ningzhu Ma
- Department of Pharmacy, Guilin Medical University, Guilin, 541004, People's Republic of China
| | - Jianguo Zhang
- Department of Pharmacy, Guilin Medical University, Guilin, 541004, People's Republic of China
| | - Zhenlin Wang
- Department of Pharmacy, Guilin Medical University, Guilin, 541004, People's Republic of China
| | - Xin Zhang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, People's Republic of China
| | - Aimin Li
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, People's Republic of China
| |
Collapse
|
4
|
Roy M, Alix C, Burlaud-Gaillard J, Fouan D, Raoul W, Bouakaz A, Blanchard E, Lecomte T, Viaud-Massuard MC, Sasaki N, Serrière S, Escoffre JM. Delivery of Anticancer Drugs Using Microbubble-Assisted Ultrasound in a 3D Spheroid Model. Mol Pharm 2024; 21:831-844. [PMID: 38174896 DOI: 10.1021/acs.molpharmaceut.3c00921] [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] [Indexed: 01/05/2024]
Abstract
Tumor spheroids are promising three-dimensional (3D) in vitro tumor models for the evaluation of drug delivery methods. The design of noninvasive and targeted drug methods is required to improve the intratumoral bioavailability of chemotherapeutic drugs and reduce their adverse off-target effects. Among such methods, microbubble-assisted ultrasound (MB-assisted US) is an innovative modality for noninvasive targeted drug delivery. The aim of the present study is to evaluate the efficacy of this US modality for the delivery of bleomycin, doxorubicin, and irinotecan in colorectal cancer (CRC) spheroids. MB-assisted US permeabilized the CRC spheroids to propidium iodide, which was used as a drug model without affecting their growth and viability. Histological analysis and electron microscopy revealed that MB-assisted US affected only the peripheral layer of the CRC spheroids. The acoustically mediated bleomycin delivery induced a significant decrease in CRC spheroid growth in comparison to spheroids treated with bleomycin alone. However, this US modality did not improve the therapeutic efficacy of doxorubicin and irinotecan on CRC spheroids. In conclusion, this study demonstrates that tumor spheroids are a relevant approach to evaluate the efficacy of MB-assisted US for the delivery of chemotherapeutics.
Collapse
Affiliation(s)
- Marie Roy
- UMR 1253, iBrain, Université de Tours, Inserm, 37032 Tours, France
| | - Corentin Alix
- UMR 1253, iBrain, Université de Tours, Inserm, 37032 Tours, France
| | - Julien Burlaud-Gaillard
- Inserm U1259, Université de Tours et CHRU de Tours & Plateforme IBiSA des Microscopies, PPF ASB, CHRU de Tours, 37032 Tours, France
| | - Damien Fouan
- UMR 1253, iBrain, Université de Tours, Inserm, 37032 Tours, France
| | - William Raoul
- Inserm UMR 1069, Nutrition Croissance et Cancer (N2C), Université de Tours, 37032 Tours, France
| | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, 37032 Tours, France
| | - Emmanuelle Blanchard
- Inserm U1259, Université de Tours et CHRU de Tours & Plateforme IBiSA des Microscopies, PPF ASB, CHRU de Tours, 37032 Tours, France
| | - Thierry Lecomte
- Inserm UMR 1069, Nutrition Croissance et Cancer (N2C), Université de Tours, 37032 Tours, France
- Department of Hepato-Gastroenterology & Digestive Oncology, CHRU de Tours, 37000 Tours, France
| | | | - Noboru Sasaki
- Laboratory of Veterinary Internal Medicine, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, 060-0818 Sapporo, Japan
| | - Sophie Serrière
- UMR 1253, iBrain, Université de Tours, Inserm, 37032 Tours, France
- Département d'Imagerie Préclinique, Plateforme Scientifique et Technique Analyse des Systèmes Biologiques, Université de Tours, 37032 Tours, France
| | | |
Collapse
|
5
|
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: 5] [Impact Index Per Article: 5.0] [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
|
6
|
Tan X, Li S, Sheng R, Zhang Q, Li C, Liu L, Zhang Y, Ge L. Biointerfacial giant capsules with high paclitaxel loading and magnetic targeting for breast tumor therapy. J Colloid Interface Sci 2023; 633:1055-1068. [PMID: 36516681 DOI: 10.1016/j.jcis.2022.11.151] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/23/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
High drug loading, targeted delivery, prolonged drug release, and low systemic toxicity are effective weapons for hydrophobic drug delivery systems to solve serious concerns in poor water-solubility and toxicity of paclitaxel (PTX). Herein, we reported that biointerfacial giant multilayer microcapsules (BGMs) with the feature of high-density drug loading and high-efficiency magnetic delivery were fabricated templated by PTX-liposome-microbubble complex using the layer-by-layer self-assembly (LbL) technique. The drug loading capacity of BGMs was improved by optimizing the structure of microbubbles and capsules to increase the PTX-contained layers, and the resultant BGMs exhibited high drug loading content (50.56 ± 0.09 %) and sustained drug release properties. The BGMs with an average diameter of 74.1 ± 12.1 µm and an average thickness of 275.5 ± 48.4 nm contained abundant magnetic nanoparticles (MNPs) in their cavity, which endowed these capsules with outstanding magnetic properties and fast magnetophoretic velocity in the blood (∼0.3 mm/s, ▽B = 1 T/mm). Moreover, both in vitro and in vivo studies demonstrated that the biocompatible PTX-loaded magnetic BGMs (Capsule@PLMPPL) caused notable death (71.3 ± 2.9 %) of 4 T1 breast cancer cells through PTX diffusion, capsules degradation, and subsequent endocytosis by cancer cells, and ultimately effectively inhibited tumor growth. In general, the developed BGM with good deformability and degradation was the first reported giant polyelectrolyte capsule to be used in tumor therapy, which could notably improve the therapeutic efficacy of PTX while reducing its side effects.
Collapse
Affiliation(s)
- Xin Tan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China
| | - Shiming Li
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009 PR China
| | - Renwang Sheng
- School of Medicine, Southeast University, Nanjing 210009, PR China
| | - Qianli Zhang
- School of Chemistry and Life Science, Suzhou University of Science and Technology, Suzhou 215009, PR China
| | - Chunyang Li
- Institute of Agroproducts Processing Jiangsu Academy of Agricultural Sciences, Nanjing 210014, PR China
| | - Ling Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009 PR China.
| | - Yu Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China.
| | - Liqin Ge
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China.
| |
Collapse
|
7
|
Chen KW, Hsu PH, Huang HL, Liu HL, Lin YT, Hsu CY, Lin JH, Lin YH. Targeting nanoparticle-conjugated microbubbles combined with ultrasound-mediated microbubble destruction for enhanced tumor therapy. Pharmacol Res 2022; 186:106532. [DOI: 10.1016/j.phrs.2022.106532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/20/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
|
8
|
Wang YC, Tian JY, Han YY, Liu YF, Chen SY, Guo FJ. Evaluation of the potential of ultrasound-mediated drug delivery for the treatment of ovarian cancer through preclinical studies. Front Oncol 2022; 12:978603. [PMID: 36132133 PMCID: PMC9483181 DOI: 10.3389/fonc.2022.978603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Ovarian cancer (OC) has the greatest mortality rate among gynecological cancers, with a five-year survival rate of <50%. Contemporary adjuvant chemotherapy mostly fails in the case of OCs that are refractory, metastatic, recurrent, and drug-resistant. Emerging ultrasound (US)-mediated technologies show remarkable promise in overcoming these challenges. Absorption of US waves by the tissue results in the generation of heat due to its thermal effect causing increased diffusion of drugs from the carriers and triggering sonoporation by increasing the permeability of the cancer cells. Certain frequencies of US waves could also produce a cavitation effect on drug-filled microbubbles (MBs, phospholipid bilayers) thereby generating shear force and acoustic streaming that could assist drug release from the MBs, and promote the permeability of the cell membrane. A new class of nanoparticles that carry therapeutic agents and are guided by US contrast agents for precision delivery to the site of the ovarian tumor has been developed. Phase-shifting of nanoparticles by US sonication has also been engineered to enhance the drug delivery to the ovarian tumor site. These technologies have been used for targeting the ovarian cancer stem cells and protein moieties that are particularly elevated in OCs including luteinizing hormone-releasing hormone, folic acid receptor, and vascular endothelial growth factor. When compared to healthy ovarian tissue, the homeostatic parameters at the tissue microenvironment including pH, oxygen levels, and glucose metabolism differ significantly in ovarian tumors. US-based technologies have been developed to take advantage of these tumor-specific alterations for precision drug delivery. Preclinical efficacy of US-based targeting of currently used clinical chemotherapies presented in this review has the potential for rapid human translation, especially for formulations that use all substances that are deemed to be generally safe by the U.S. Food and Drug Administration.
Collapse
Affiliation(s)
- Yi-Chao Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Jing-Yan Tian
- Department of Urology, The Second Division of the First Hospital of Jilin University, Changchun, China
| | - Ying-Ying Han
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Yun-Fei Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Si-Yao Chen
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Feng-Jun Guo
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Feng-Jun Guo,
| |
Collapse
|
9
|
He J, Liu Z, Zhu X, Xia H, Gao H, Lu J. Ultrasonic Microbubble Cavitation Enhanced Tissue Permeability and Drug Diffusion in Solid Tumor Therapy. Pharmaceutics 2022; 14:1642. [PMID: 36015267 PMCID: PMC9414228 DOI: 10.3390/pharmaceutics14081642] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/31/2022] [Accepted: 08/04/2022] [Indexed: 01/21/2023] Open
Abstract
Chemotherapy has an essential role not only in advanced solid tumor therapy intervention but also in society's health at large. Chemoresistance, however, seriously restricts the efficiency and sensitivity of chemotherapeutic agents, representing a significant threat to patients' quality of life and life expectancy. How to reverse chemoresistance, improve efficacy sensitization response, and reduce adverse side effects need to be tackled urgently. Recently, studies on the effect of ultrasonic microbubble cavitation on enhanced tissue permeability and retention (EPR) have attracted the attention of researchers. Compared with the traditional targeted drug delivery regimen, the microbubble cavitation effect, which can be used to enhance the EPR effect, has the advantages of less trauma, low cost, and good sensitization effect, and has significant application prospects. This article reviews the research progress of ultrasound-mediated microbubble cavitation in the treatment of solid tumors and discusses its mechanism of action to provide new ideas for better treatment strategies.
Collapse
Affiliation(s)
- Jide He
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Zenan Liu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Xuehua Zhu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Haizhui Xia
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Jian Lu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
10
|
Wang X, Dong H. A convergent synthetic platform for anticancer drugs formulation with nanoparticle delivery for the treatment and nursing care of glioma cancer. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
11
|
Jangjou A, Meisami AH, Jamali K, Niakan MH, Abbasi M, Shafiee M, Salehi M, Hosseinzadeh A, Amani AM, Vaez A. The promising shadow of microbubble over medical sciences: from fighting wide scope of prevalence disease to cancer eradication. J Biomed Sci 2021; 28:49. [PMID: 34154581 PMCID: PMC8215828 DOI: 10.1186/s12929-021-00744-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/10/2021] [Indexed: 12/29/2022] Open
Abstract
Microbubbles are typically 0.5-10 μm in size. Their size tends to make it easier for medication delivery mechanisms to navigate the body by allowing them to be swallowed more easily. The gas included in the microbubble is surrounded by a membrane that may consist of biocompatible biopolymers, polymers, surfactants, proteins, lipids, or a combination thereof. One of the most effective implementation techniques for tiny bubbles is to apply them as a drug carrier that has the potential to activate ultrasound (US); this allows the drug to be released by US. Microbubbles are often designed to preserve and secure medicines or substances before they have reached a certain area of concern and, finally, US is used to disintegrate microbubbles, triggering site-specific leakage/release of biologically active drugs. They have excellent therapeutic potential in a wide range of common diseases. In this article, we discussed microbubbles and their advantageous medicinal uses in the treatment of certain prevalent disorders, including Parkinson's disease, Alzheimer's disease, cardiovascular disease, diabetic condition, renal defects, and finally, their use in the treatment of various forms of cancer as well as their incorporation with nanoparticles. Using microbubble technology as a novel carrier, the ability to prevent and eradicate prevalent diseases has strengthened the promise of effective care to improve patient well-being and life expectancy.
Collapse
Affiliation(s)
- Ali Jangjou
- Department of Emergency Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Hossein Meisami
- Department of Emergency Medicine, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kazem Jamali
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hadi Niakan
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Abbasi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mostafa Shafiee
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Salehi
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Ahmad Hosseinzadeh
- Thoracic and Vascular Surgery Research Center, Nemazee Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Zhu X, Yan S, Xiao F, Xue M. PLGA nanoparticles delivering CPT-11 combined with focused ultrasound inhibit platinum resistant ovarian cancer. Transl Cancer Res 2021; 10:1732-1743. [PMID: 35116498 PMCID: PMC8797841 DOI: 10.21037/tcr-20-3171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/22/2021] [Indexed: 11/06/2022]
Abstract
Background Ovarian cancer cells show resistance to platinum drugs treatment, which brings a big challenge to clinical therapeutics. This study aimed to construct effective drug delivering nanoparticles specifically targeting ovarian cancer cell. Methods Poly lactic-co-glycolic acid (PLGA) were used to form Nano-spheres by double emulsion method, and to deliver CPT-11. Connected with targeted LHRH-a molecules, their effects were tested by ovarian cancer cell A2780/DDP in vitro and in vivo. Results We successfully constructed PLGA nanoparticles carrying LHRH-a (Luteinizing hormone releasing hormone analogue) and CPT-11 (irinotecan HCl trihydrate), which can specifically target LHRH receptor high expression ovarian cancer cell A2780/DDP (cisplatin). Combined with focused ultrasound in vitro, LHRH-a/CPT-11/PLGA nanoparticles significantly inhibited the proliferation of A2780/DDP cells (a cisplatin-resistant A2780 cell line), and the cells were obviously arrested at S phase. Both the mRNA expression and protein level of Caspase3 increased, while Bcl-2 and MMP2 declined, which promoted apoptosis. In vivo, LHRH-a/CPT-11/PLGA nanoparticles bind specifically with LHRH receptor on xenograft tumors of A2780/DDP. With focused ultrasound, LHRH-a/CPT-11/PLGA nanoparticles inhibited the growth of A2780/DDP xenograft tumors significantly. The expression level of VEGF, Bcl-2 and MMP2 reduced, while Caspase3 increased in tumors. Conclusions CPT-11 delivering PLGA nanoparticles with LHRH-a specifically target ovarian cancer cell A2780/DDP, and work locally when combined with focused ultrasound. They increase local drug concentration and reduce side effects. This research may provide a new effective therapeutic strategy for recurrent platinum resistant ovarian cancer.
Collapse
Affiliation(s)
- Xiaogang Zhu
- Department of Gynecology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Siqi Yan
- Department of Oncology, Second Xiangya Hospital, Central South University, Changsha, China.,Department of Radiation Oncology, Hunan Academy of Traditional Chinese Medicine Affiliated Hospital, Changsha, China
| | - Fang Xiao
- Department of Gynecology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Min Xue
- Department of Gynecology, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
13
|
Han W, Liu X, Wang L, Zhou X. Engineering of lipid microbubbles-coated copper and selenium nanoparticles: Ultrasound-stimulated radiation of anticancer activity ian human ovarian cancer cells. Process Biochem 2020. [DOI: 10.1016/j.procbio.2020.07.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
14
|
Recent advances in ultrasound-triggered drug delivery through lipid-based nanomaterials. Drug Discov Today 2020; 25:2182-2200. [PMID: 33010479 DOI: 10.1016/j.drudis.2020.09.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/24/2020] [Accepted: 09/22/2020] [Indexed: 12/28/2022]
Abstract
The high prescribed dose of anticancer drugs and their resulting adverse effects on healthy tissue are significant drawbacks to conventional chemotherapy (CTP). Ideally, drugs should have the lowest possible degree of interaction with healthy cells, which would diminish any adverse effects. Therefore, an ideal scenario to bring about improvements in CTP is the use of technological strategies to ensure the efficient, specific, and selective transport and/or release of drugs to the target site. One practical and feasible solution to promote the efficiency of conventional CTP is the use of ultrasound (US). In this review, we highlight the potential role of US in combination with lipid-based carriers to achieve a targeted CTP strategy in engineered smart drug delivery systems.
Collapse
|
15
|
Chandan R, Mehta S, Banerjee R. Ultrasound-Responsive Carriers for Therapeutic Applications. ACS Biomater Sci Eng 2020; 6:4731-4747. [PMID: 33455210 DOI: 10.1021/acsbiomaterials.9b01979] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ultrasound (US)-responsive carriers have emerged as promising theranostic candidates because of their ability to enhance US-contrast, promote image-guided drug delivery, cause on-demand pulsatile release of drugs in response to ultrasound stimuli, as well as to enhance the permeability of physiological barriers such as the stratum corneum, the vascular endothelium, and the blood-brain barrier (BBB). US-responsive carriers include microbubbles MBs, liposomes, droplets, hydrogels, and nanobubble-nanoparticle complexes and have been explored for cavitation-mediated US-responsive drug delivery. Recently, a transient increase in the permeability of the BBB by microbubble (MB)-assisted low-frequency US has shown promise in enhancing the delivery of therapeutic agents in the case of neurological disorders. Further, the periodic mechanical stimulus generated by US-responsive MBs have also been explored in tissue engineering and has directly influenced the differentiation of mesenchymal stem cells into cartilage. This Review discusses the various types of US-responsive carriers and explores their emerging roles in therapeutics ranging from drug delivery to tissue engineering.
Collapse
Affiliation(s)
- Rajeet Chandan
- Nanomedicine Lab, Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Sourabh Mehta
- Nanomedicine Lab, Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.,IIT Bombay-Monash Research Academy, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Rinti Banerjee
- Nanomedicine Lab, Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
16
|
Tavsan Z, Kayali HA. Protein Kinase C regulates the complex between cell membrane molecules in ovarian cancer. Process Biochem 2020. [DOI: 10.1016/j.procbio.2020.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
17
|
Ding Y, Cao Q, Qian S, Chen X, Xu Y, Chen J, Shen H. Optimized Anti-Prostate-Specific Membrane Antigen Single-Chain Variable Fragment-Loaded Nanobubbles as a Novel Targeted Ultrasound Contrast Agent for the Diagnosis of Prostate Cancer. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2020; 39:761-773. [PMID: 31702068 PMCID: PMC7155035 DOI: 10.1002/jum.15155] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 07/12/2019] [Accepted: 09/22/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVES To prepare optimized prostate-specific membrane antigen (PSMA) single-chain variable fragment (scFv)-loaded nanobubbles (NBs) as a novel targeted ultrasound (US) contrast agent for diagnosis and treatment of prostate cancer (PCa). METHODS Prostate-specific membrane antigen scFv-loaded NBs were prepared by membrane hydration and biotin-streptavidin conjugation. Flow cytometry was used to observe the binding rate of the targeted NBs to PSMA-expressing cells. Contrast-enhanced US was used to monitor targeted and nontargeted NBs administered to nude mice with 22RV1, LNCaP, and PC-3 xenograft tumors. The specific binding ability of the targeted NBs was further examined by fluorescence imaging of tumor cryosections. RESULTS Uniformly sized targeted NBs were successfully prepared (mean ± SD, 485.3 ± 28.4 nm). The NBs showed good stability and bound specifically to LNCaP and 22RV1 cells with high PSMA expression in vitro but did not bind to PC-3 cells without PSMA expression. The targeted NBs presented good US enhancement, and the results of the in vivo xenograft tumor nude mouse model showed that the peak contrast intensity in LNCaP and 22RV1 cells was significantly higher for the targeted NBs than the nontargeted NBs (P < .05), whereas there was no significant difference in PC-3 cells. Immunofluorescence results obtained from tumor sections confirmed that the targeted NBs were capable of targeting PSMA-expressing tumor cells. CONCLUSIONS These novel PSMA scFv-loaded NBs have proven to be an excellent US contrast agent for imaging PSMA-expressing PCa and have the potential to not only enable efficient and safe molecular imaging but also to serve as a delivery system for targeted PCa therapies.
Collapse
Affiliation(s)
- Yu Ding
- Department of Urology, Xinhua Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Qifeng Cao
- Department of Urology, Xinhua Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Subo Qian
- Department of Urology, Xinhua Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xiaolong Chen
- Zhejiang California International NanoSystems InstituteZhejiang UniversityHangzhouChina
| | - Yuhong Xu
- School of PharmacyShanghai Jiao Tong UniversityShanghaiChina
| | - Jian Chen
- School of PharmacyShanghai Jiao Tong UniversityShanghaiChina
| | - Haibo Shen
- Department of Urology, Xinhua Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
18
|
Mohamed TMD, Ji-Bin LMD, John REP. Recent Advances in Microbubble-Augmented Cancer Therapy. ADVANCED ULTRASOUND IN DIAGNOSIS AND THERAPY 2020. [DOI: 10.37015/audt.2020.200055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
19
|
Drug-Loaded Microbubbles Combined with Ultrasound for Thrombolysis and Malignant Tumor Therapy. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6792465. [PMID: 31662987 PMCID: PMC6791276 DOI: 10.1155/2019/6792465] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/22/2019] [Accepted: 09/14/2019] [Indexed: 12/14/2022]
Abstract
Cardiac-cerebral thrombosis and malignant tumor endanger the safety of human life seriously. Traditional chemotherapy drugs have side effects which restrict their applications. Drug-loaded microbubbles can be destroyed by ultrasound irradiation at the focus position and be used for thrombolysis and tumor therapy. Compared with traditional drug treatment, the drug-loaded microbubbles can be excited by ultrasound and release drugs to lesion sites, increasing the local drug concentration and the exposure dose to nonfocal regions, thus reducing the cytotoxicity and side effects of drugs. This article reviews the applications of drug-loaded microbubbles combined with ultrasound for thrombolysis and tumor therapy. We focus on highlighting the advantages of using this new technique for disease treatment and concluding with recommendations for future efforts on the applications of this technology.
Collapse
|
20
|
Fu JW, Lin YS, Gan SL, Li YR, Wang Y, Feng ST, Li H, Zhou GF. Multifunctionalized Microscale Ultrasound Contrast Agents for Precise Theranostics of Malignant Tumors. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:3145647. [PMID: 31360144 PMCID: PMC6642784 DOI: 10.1155/2019/3145647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/26/2019] [Accepted: 06/10/2019] [Indexed: 11/17/2022]
Abstract
In ultrasonography, ultrasound contrast agents (UCAs) that possess high acoustic impedance mismatch with the bulk medium are frequently employed to highlight the borders between tissues by enhanced ultrasound scattering in a clinic. Typically, the most common UCA, microbubble, is generally close in size to a red blood cell (<∼10 μm). These microscale UCAs cannot be directly entrapped into the target cells but generate several orders of magnitude stronger echo signals than the nanoscale ones. And their large containment and high ultrasound responsiveness also greatly facilitate to perform combined treatments, e.g., drug delivery and other imaging techniques. So multifunctionalized microscale UCAs appear on this scene and keep growing toward a promising direction for precise theranostics. In this review, we systematically summarize the new advances in the principles and preparations of multifunctionalized microscale UCAs and their medical applications for malignant tumors.
Collapse
Affiliation(s)
- Jia-Wei Fu
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology & Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou 510006, China
| | - Yi-Sheng Lin
- Department of Radiology, The First Affiliated Hospital, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Sheng-Long Gan
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology & Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou 510006, China
| | - Yong-Rui Li
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology & Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou 510006, China
| | - Yao Wang
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology & Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou 510006, China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Hao Li
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology & Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou 510006, China
| | - Guo-Fu Zhou
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology & Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou 510006, China
| |
Collapse
|
21
|
Qu N, Shi D, Shang M, Duan S, Guo L, Ning S, Li J. Breast Cancer Cell Line Phenotype Affects Sonoporation Efficiency Under Optimal Ultrasound Microbubble Conditions. Med Sci Monit 2018; 24:9054-9062. [PMID: 30546004 PMCID: PMC6302661 DOI: 10.12659/msm.910790] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Ultrasound/microbubble (USMB)-mediated sonoporation is a new strategy with minimal procedural invasiveness for targeted and site-specific drug delivery to tumors. The purpose of this study was to explore the effect of different breast cancer cell lines on sonoporation efficiency, and then to identify an optimal combination of USMB parameters to maximize the sonoporation efficiency for each tumor cell line. Material/Methods Three drug-sensitive breast cell lines – MCF-7, MDA-MB-231, and MDA-MB-468 – and 1 multidrug resistance (MDR) cell line – MCF-7/ADR – were chosen. An orthogonal array experimental design approach based on 3 levels of 3 parameters (A: microbubble concentration, 10%, 20%, and 30%, B: sound intensity, 0.5, 1.0, and 1.5 W/cm2, C: irradiation time, 30, 60, and 90 s) was employed to optimize the sonoporation efficiency. Results The optimal USMB parameter combinations for different cell lines were diverse. Under optimal parameter combinations, the maximum sonoporation efficiency differences between different breast tumor cell lines were statistically significant (MDA-MB-231: 46.70±5.79%, MDA-MB-468: 53.44±5.69%, MCF-7: 59.88±5.53%, MCF-7/ADR: 65.39±4.01%, P<0.05), so were between drug-sensitive cell line and MDR cell line (MCF-7: 59.88±5.53%, MCF-7/ADR: 65.39±4.01%, p=0.026). Conclusions Different breast tumor cell lines have their own optimal sonoporation. Drug-resistant MCF-7/ADR cells had higher sonoporation efficiency than drug-sensitive MCF-7 cells. The molecular subtype of tumors should be considered when sonoporation is applied, and optimal parameter combination may have the potential to improve drug-delivery efficiency by increasing the sonoporation efficiency.
Collapse
Affiliation(s)
- Nina Qu
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland).,Department of Ultrasound, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China (mainland)
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Sujuan Duan
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Song Ning
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
22
|
Skachkov I, Luan Y, van Tiel ST, van der Steen AFW, de Jong N, Bernsen MR, Kooiman K. SPIO labeling of endothelial cells using ultrasound and targeted microbubbles at diagnostic pressures. PLoS One 2018; 13:e0204354. [PMID: 30235336 PMCID: PMC6147550 DOI: 10.1371/journal.pone.0204354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 09/06/2018] [Indexed: 02/07/2023] Open
Abstract
In vivo cell tracking of therapeutic, tumor, and endothelial cells is an emerging field and a promising technique for imaging cardiovascular disease and cancer development. Site-specific labeling of endothelial cells with the MRI contrast agent superparamagnetic iron oxide (SPIO) in the absence of toxic agents is challenging. Therefore, the aim of this in vitro study was to find optimal parameters for efficient and safe SPIO-labeling of endothelial cells using ultrasound-activated CD31-targeted microbubbles for future MRI tracking. Ultrasound at a frequency of 1 MHz (10,000 cycles, repetition rate of 20 Hz) was used for varying applied peak negative pressures (10–160 kPa, i.e. low mechanical index (MI) of 0.01–0.16), treatment durations (0–30 s), time of SPIO addition (-5 min– 15 min with respect to the start of the ultrasound), and incubation time after SPIO addition (5 min– 3 h). Iron specific Prussian Blue staining in combination with calcein-AM based cell viability assays were applied to define the most efficient and safe conditions for SPIO-labeling. Optimal SPIO labeling was observed when the ultrasound parameters were 40 kPa peak negative pressure (MI 0.04), applied for 30 s just before SPIO addition (0 min). Compared to the control, this resulted in an approximate 12 times increase of SPIO uptake in endothelial cells in vitro with 85% cell viability. Therefore, ultrasound-activated targeted ultrasound contrast agents show great potential for effective and safe labeling of endothelial cells with SPIO.
Collapse
Affiliation(s)
- Ilya Skachkov
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
| | - Ying Luan
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
| | - Sandra T. van Tiel
- Department of Radiology & Nucleair Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Antonius F. W. van der Steen
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
- Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Nico de Jong
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
- Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Monique R. Bernsen
- Department of Radiology & Nucleair Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Klazina Kooiman
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
- * E-mail:
| |
Collapse
|
23
|
An experimental study of ovarian cancer imaging and therapy by paclitaxel-loaded phase-transformation lipid nanoparticles combined with low-intensity focused ultrasound. Biochem Biophys Res Commun 2018; 504:203-210. [PMID: 30180951 DOI: 10.1016/j.bbrc.2018.08.156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 08/26/2018] [Indexed: 01/03/2023]
Abstract
Drug-loaded phase-transformation lipid nanoparticles (NPs) combined with low-intensity focused ultrasound (LIFU) for ultrasound molecular imaging and therapy, which is a very promising drug carrier and can provide both physical and chemical therapeutics, simultaneously. We successfully prepared the paclitaxel (PTX) loaded anti-LHRHR targeted phase-transformation lipid nanoparticles (PTX-anti-LHRHR-PTNPs) for ovarian cancer in this study combined with LIFU has the following characteristics: On the one hand, it showed smaller size and greater stability than blood cells, which significantly prolonged its half-life in the body, and can actively target ovarian cancer OVCAR-3 cells, and smoothly penetrate the endothelial gap into the tumor site for specifically killing the ovarian cancer cells. Thereby, the special drug carrier improved the therapeutic effect and reduced toxic and side effects, maximized the protection of normal tissues and minimized adverse reactions. On the other hand, PTX-anti-LHRHR-PTNPs can be targeted to focus after being injected intravenously and remain in the tumor target tissue for a long time. At the same time, liquid-gas phase-transformation can occur under LIFU triggering, resulting in more ideal and sustained ultrasound imaging effects. Then acoustic contrast agent is used to develop the molecular level of ultrasound scattering, so as to evaluate the diseased tissue from the molecular level.
Collapse
|
24
|
van den Brand D, Mertens V, Massuger LF, Brock R. siRNA in ovarian cancer – Delivery strategies and targets for therapy. J Control Release 2018; 283:45-58. [DOI: 10.1016/j.jconrel.2018.05.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 12/21/2022]
|
25
|
Roy J, Kaake M, Srinivasarao M, Low PS. Targeted Tubulysin B Hydrazide Conjugate for the Treatment of Luteinizing Hormone-Releasing Hormone Receptor-Positive Cancers. Bioconjug Chem 2018; 29:2208-2214. [DOI: 10.1021/acs.bioconjchem.8b00164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
26
|
Shi F, Li M, Wang J, Wu D, Pan M, Guo M, Dou J. Induction of multiple myeloma cancer stem cell apoptosis using conjugated anti-ABCG2 antibody with epirubicin-loaded microbubbles. Stem Cell Res Ther 2018; 9:144. [PMID: 29784015 PMCID: PMC5963075 DOI: 10.1186/s13287-018-0885-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/05/2018] [Accepted: 04/23/2018] [Indexed: 02/08/2023] Open
Abstract
Background Multiple myeloma (MM) currently remains largely incurable. Cancer stem cells (CSCs) are believed to be responsible for drug resistance and eventual relapse. In this study, we exploited a novel agent to evaluate its inhibitory effect on MM CSCs. Methods Epirubicin (EPI)-loaded lipid microbubbles (MBs) conjugated with anti-ABCG2 monoclonal antibody (EPI-MBs + mAb) were developed and their effect on MM 138−CD34− CSCs isolated from human MM RPMI 8226 cell line plus ultrasound exposure in vitro and in vivo in a nonobese diabetic/severe combined immunodeficient mouse model were assessed. Results EPI-MBs + mAb combined with ultrasound led to a significant decrease in the clone formation ability and the mitochondrial membrane potential along with an increase in MM CSC apoptosis. Moreover, treatment with EPI-MBs + mAb with ultrasound exposure remarkably inhibited the growth of MM CSC-derived tumors in xenograft nonobese diabetic/severe combined immunodeficient mice compared with a single agent or EPI-MBs + mAb without ultrasound exposure. The inhibitive efficacy was also correlated with an increased expression of caspase-3, Bax, and TUNEL and decreased expressions of PCNA, Bcl-2, and CD31. Conclusions Our findings reveal that the EPI-MBs + mAb combined with therapeutic ultrasound may confer an effective approach for treatment of MM by induction of an apoptotic pathway in MM CSCs.
Collapse
Affiliation(s)
- Fangfang Shi
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.,Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, 87# Ding Jiaqiao Rd., Nanjing, 210009, China
| | - Miao Li
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, 87# Ding Jiaqiao Rd., Nanjing, 210009, China
| | - Jing Wang
- Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Di Wu
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, 87# Ding Jiaqiao Rd., Nanjing, 210009, China.,Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Meng Pan
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, 87# Ding Jiaqiao Rd., Nanjing, 210009, China
| | - Mei Guo
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jun Dou
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, 87# Ding Jiaqiao Rd., Nanjing, 210009, China.
| |
Collapse
|
27
|
Jang KW, Seol D, Ding L, Lim TH, Frank JA, Martin JA. Ultrasound-Mediated Microbubble Destruction Suppresses Melanoma Tumor Growth. ULTRASOUND IN MEDICINE & BIOLOGY 2018; 44:831-839. [PMID: 29361373 PMCID: PMC5826859 DOI: 10.1016/j.ultrasmedbio.2017.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/29/2017] [Accepted: 12/13/2017] [Indexed: 05/23/2023]
Abstract
Melanoma is one of the most aggressive types of cancer, and its incidence has increased rapidly in the past few decades. In this study, we investigated a novel treatment approach, the use of low-intensity ultrasound (2.3 W/cm2 at 1 MHz)-mediated Optison microbubble (MB) destruction (UMMD) to treat melanoma in a flank tumor model. The effect of UMMD was first evaluated in the melanoma cell line B16 F10 (B16) in vitro and then in mice inoculated with B16 cells. MB+B16 cells were exposed to US in vitro, resulting in significant cell death proportional to duty cycle (R2 = 0.74): approximately 30%, 50%, 80% and 80% cell death at 10%, 30%, 50% and 100% DC respectively. Direct implantation of tumors with MBs, followed by sonication, resulted in retarded tumor growth and improved survival (p = 0.0106). Immunohistochemical analyses confirmed the significant changes in expression of the cell proliferation marker Ki67 (p = 0.037) and a microtubule-associated protein 2 (p = 0.048) after US + MB treatment. These results suggest that UMMD could be used as a possible treatment approach in isolated melanoma and has the potential to translate to clinical trials.
Collapse
Affiliation(s)
- Kee W Jang
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA.
| | - Dongrim Seol
- Department of Orthopaedics and Rehabilitation, The University of Iowa, Iowa City, Iowa, USA
| | - Lei Ding
- Jiangnan University Wuxi Medical School, Wuxi, Jiangsu, China
| | - Tae-Hong Lim
- Department of Biomedical Engineering, The University of Iowa, Iowa City, Iowa, USA
| | - Joseph A Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA; National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | - James A Martin
- Department of Orthopaedics and Rehabilitation, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
28
|
Shi D, Guo L, Duan S, Shang M, Meng D, Cheng L, Li J. Influence of tumor cell lines derived from different tissue on sonoporation efficiency under ultrasound microbubble treatment. ULTRASONICS SONOCHEMISTRY 2017; 38:598-603. [PMID: 27562907 DOI: 10.1016/j.ultsonch.2016.08.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/14/2016] [Accepted: 08/15/2016] [Indexed: 06/06/2023]
Abstract
To reveal the effect of tumor cell lines derived from different tissue on sonoporation efficiency under ultrasound microbubble (USMB) treatment, and meanwhile to determine the optimum parameter combination for each tumor cell line. Human breast tumor (MCF-7), ovarian tumor (A2780), liver tumor (Bel7402) and thyroid tumor (ARO) were exposed to ultrasound in the presence of SonoVue MBs. The major parameters for the designed experiments including MB concentration (A1:10%, A2:20%, A3:30%), sound intensity (B1:0.5, B2:1.0, B3:1.5W/cm2), irradiation time (C1:30, C2:60, C3:90s). An orthogonal array experimental design based on three levels L9 (33) of the above three parameters was employed to optimize the sonoporation efficiency. MTT experiment was used to calculate cell survival rate. FD500 uptake assay and cytometry were performed to evaluate transference percentage. The optimum parameter combination for each tumor cell line was different (MCF-7: A3B1C1, A2780: A1B3C3, Bel7402: A2B3C2, ARO: A2B3C2). Under their own optimum parameter combination, four kinds of tumor cell line exhibited different optimum sonoporation efficiency (MCF-7: 55.05±5.29%; A2780: 45.62±7.35%; Bel7402: 39.37±4.11%; ARO: 53.37±3.94%). Multiple comparison with LSD-t test showed that the sonoporation efficiency between four kinds of cell line was statistically significant (P<0.05), with the exception of MCF-7 VS. ARO (P=0.487). Tumor cell lines derived from different tissue can impact the sonoporation efficiency, and optimizing the exposure parameters can safely and efficiently increase the cell membrane permeability.
Collapse
Affiliation(s)
- Dandan Shi
- Department of Ultrasound, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Sujuan Duan
- Department of Ultrasound, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Dong Meng
- Department of Ultrasound, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Lin Cheng
- Department of Ultrasound, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital, Shandong University, Jinan 250012, China.
| |
Collapse
|
29
|
Ultrasound-triggered PLGA microparticle destruction and degradation for controlled delivery of local cytotoxicity and drug release. Int J Biol Macromol 2017; 106:1211-1217. [PMID: 28851638 DOI: 10.1016/j.ijbiomac.2017.08.125] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 08/15/2017] [Accepted: 08/22/2017] [Indexed: 11/21/2022]
Abstract
In this study, we investigated the low intensity ultrasound (US)-controlled delivery of local cytotoxicity and drug release via induced destruction and degradation of microparticles (MPs) made of poly(lactic-co-glycolic acid) (PLGA). This study was conducted in vitro with potential application towards tumor treatment in conjunction with direct injection. MPs, either loaded with or without doxorubicin (DOX), were prepared using a double-emulsion solvent-evaporation technique. First, the MPs were exposed to US with duty cycle (DC)-modulation. The destruction and degradation of MPs were evaluated using light and scanning electron microscopy. Second, the effects of US-mediated destruction/degradation of MPs on the local cytotoxicity as well as DOX release were evaluated. US-triggered MP destruction/degradation significantly enhanced nearby cell death and DOX release. These affects occurred in proportion to the DC. Our findings indicate that controlled cytotoxicity and DOX release by US could be useful in developing the minimally invasive therapeutic applications for tumor treatment.
Collapse
|
30
|
Shi F, Li M, Wu S, Yang F, Di W, Pan M, Zhao F, Luo S, Gu N, Dou J. Enhancing the anti-multiple myeloma efficiency in a cancer stem cell xenograft model by conjugating the ABCG2 antibody with microbubbles for a targeted delivery of ultrasound mediated epirubicin. Biochem Pharmacol 2017; 132:18-28. [PMID: 28232025 DOI: 10.1016/j.bcp.2017.02.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/16/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Although multiple myeloma (MM) treatment has improved in the last decade, it remains largely incurable. One of main reasons is that there are cancer stem cells (CSCs) in MM, which are responsible for MM's drug resistance and relapse. In this study, we used the targeting microbubbles (MBs) conjugated with anti-ABCG2 monoclonal antibody (mAb) for ultrasound mediated epirubicin (EPI) delivery to evaluate the therapeutic effectiveness of the novel agent in MM CSC xenograft model. METHODS MM CSCs, marked by CD138-CD34- cell phenotypes were isolated from human MM RPMI8226 cell line using immune magnetic activated cell sorting system, and inoculated into nonobese diabetic/severe combined immunodeficient mice by subcutaneous or intravenous injection. After the mice developed MM, they were intravenous injection treated with EPI, EPI-MBs+mAb, and EPI-MBs+mAb with ultrasound exposure, respectively. RESULTS All treated mice showed inhibited tumor sizes or bone lesions, decreased renal damages and anemia, and increased MM bearing mice' survival. In particular, the EPI-MBs+mAb plus ultrasound exhibited significantly enhanced therapeutic MM effectiveness by inducing apoptosis compared with other biologic agents. CONCLUSION The data provide evidence that EPI-MBs+mAb with ultrasound exposure might be available for treatment MM patients in clinic.
Collapse
Affiliation(s)
- Fangfang Shi
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Miao Li
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Songyan Wu
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Fang Yang
- School of Biological Science and Technology, Southeast University, Nanjing 210096, China
| | - Wu Di
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Meng Pan
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Fengshu Zhao
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Shouhua Luo
- School of Biological Science and Technology, Southeast University, Nanjing 210096, China
| | - Ning Gu
- School of Biological Science and Technology, Southeast University, Nanjing 210096, China
| | - Jun Dou
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
31
|
Zhou S, Zhang X, Wang C. Rituximab-conjugated, doxorubicin-loaded microbubbles as a theranostic modality in B-cell lymphoma. Oncotarget 2017; 8:4760-4772. [PMID: 27902473 PMCID: PMC5354869 DOI: 10.18632/oncotarget.13587] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 11/07/2016] [Indexed: 11/25/2022] Open
Abstract
This study evaluated rituximab-conjugated, doxorubicin-loaded microbubbles (RDMs) in combination with ultrasound as molecular imaging agents for early diagnosis of B cell lymphomas, and as a targeted drug delivery system. Rituximab, a monoclonal CD20 antibody, was attached to the surfaces of doxorubicin-loaded microbubbles. RDM binding to B cell lymphoma cells was assessed using immunofluorescence. The cytotoxic effects of RDMs in combination with ultrasound (RDMs+US) were evaluated in vitro in CD20+ and CD20– cell lines, and its antitumor activities were assessed in Raji (CD20+) and Jurkat (CD20–) lymphoma cell-grafted mice. RDMs specifically bound to CD20+ cells in vitro and in vivo. Contrast enhancement was monitored in vivo via ultrasound. RDM peak intensities and contrast enhancement durations were higher in Raji than in Jurkat cell-grafted mice (P<0.05). RDMs+US treatment resulted in improved antitumor effects and reduced systemic toxicity in Raji cell-grafted mice compared with other treatments (P<0.05). Our results showed that RDMs+US enhanced tumor targeting, reduced systemic toxicity, and inhibited CD20+ B cell lymphoma growth in vivo. Targeted RDMs could be employed as ultrasound molecular imaging agents for early diagnosis, and are an effective targeted drug delivery system in combination with ultrasound for CD20+ B cell malignancy treatment.
Collapse
Affiliation(s)
- Shoubing Zhou
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Xiu Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Cailian Wang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
32
|
Mulvana H, Browning RJ, Luan Y, de Jong N, Tang MX, Eckersley RJ, Stride E. Characterization of Contrast Agent Microbubbles for Ultrasound Imaging and Therapy Research. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2017; 64:232-251. [PMID: 27810805 DOI: 10.1109/tuffc.2016.2613991] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The high efficiency with which gas microbubbles can scatter ultrasound compared with the surrounding blood pool or tissues has led to their widespread employment as contrast agents in ultrasound imaging. In recent years, their applications have been extended to include super-resolution imaging and the stimulation of localized bio-effects for therapy. The growing exploitation of contrast agents in ultrasound and in particular these recent developments have amplified the need to characterize and fully understand microbubble behavior. The aim in doing so is to more fully exploit their utility for both diagnostic imaging and potential future therapeutic applications. This paper presents the key characteristics of microbubbles that determine their efficacy in diagnostic and therapeutic applications and the corresponding techniques for their measurement. In each case, we have presented information regarding the methods available and their respective strengths and limitations, with the aim of presenting information relevant to the selection of appropriate characterization methods. First, we examine methods for determining the physical properties of microbubble suspensions and then techniques for acoustic characterization of both suspensions and single microbubbles. The next section covers characterization of microbubbles as therapeutic agents, including as drug carriers for which detailed understanding of their surface characteristics and drug loading capacity is required. Finally, we discuss the attempts that have been made to allow comparison across the methods employed by various groups to characterize and describe their microbubble suspensions and promote wider discussion and comparison of microbubble behavior.
Collapse
|
33
|
Luo T, Sun J, Zhu S, He J, Hao L, Xiao L, Zhu Y, Wang Q, Pan X, Wang Z, Chang S. Ultrasound-mediated destruction of oxygen and paclitaxel loaded dual-targeting microbubbles for intraperitoneal treatment of ovarian cancer xenografts. Cancer Lett 2016; 391:1-11. [PMID: 28043912 DOI: 10.1016/j.canlet.2016.12.032] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 12/20/2022]
Abstract
Folate receptor (FR) is overexpressed in many epithelial cancers and tumor-associated macrophages (TAMs), which enable it to function as an appropriate target for cancer treatment. We have successfully synthesized multifunctional folate-targeted and oxygen/paclitaxel loaded microbubbles (TOPLMBs) for ultrasound (US) mediated delivery for combination therapy in an intraperitoneal ovarian cancer xenograft model. The TOPLMBs target both ovarian cancer cells and TAMs and provide a promising drug delivery strategy for the combination treatment of ovarian cancer and tumor microenvironment. Microscopic imaging and flow cytometric analysis showed that TOPLMBs significantly penetrated into ovarian cancer cells and tumor-associated macrophages (TAMs) within tumor ascites fluid and the tumor nodules. Immunohistochemical analyses of dissected tumor tissue confirmed the increased tumor apoptosis, the reduced expression of vascular endothelial growth factor (VEGF) and microvascular density (MVD), and the reduced expression of CD68 after treatment (P < 0.05). Our experiment results suggest that intraperitoneal injection of dual-targeting TOPLMBs followed by US mediation provide a promising drug delivery strategy for combination treatment of ovarian cancer and tumor microenvironment with the therapeutic outcome superior to that of conventional therapeutic options.
Collapse
Affiliation(s)
- Tingting Luo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China; Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Ultrasound Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jiangchuan Sun
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Shenyin Zhu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Juan He
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Lan Hao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Linlin Xiao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yi Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qianqian Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xin Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhigang Wang
- Institute of Ultrasound Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Shufang Chang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
34
|
Sennoga CA, Kanbar E, Auboire L, Dujardin PA, Fouan D, Escoffre JM, Bouakaz A. Microbubble-mediated ultrasound drug-delivery and therapeutic monitoring. Expert Opin Drug Deliv 2016; 14:1031-1043. [DOI: 10.1080/17425247.2017.1266328] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Charles A. Sennoga
- UMR Imagerie et Cerveau, Inserm U930, Université François Rabelais, Tours, France
| | - Emma Kanbar
- UMR Imagerie et Cerveau, Inserm U930, Université François Rabelais, Tours, France
| | - Laurent Auboire
- UMR Imagerie et Cerveau, Inserm U930, Université François Rabelais, Tours, France
| | | | - Damien Fouan
- UMR Imagerie et Cerveau, Inserm U930, Université François Rabelais, Tours, France
| | - Jean-Michel Escoffre
- UMR Imagerie et Cerveau, Inserm U930, Université François Rabelais, Tours, France
| | - Ayache Bouakaz
- UMR Imagerie et Cerveau, Inserm U930, Université François Rabelais, Tours, France
| |
Collapse
|
35
|
Cao LB, Zeng S, Zhao W. Highly Stable PEGylated Poly(lactic-co-glycolic acid) (PLGA) Nanoparticles for the Effective Delivery of Docetaxel in Prostate Cancers. NANOSCALE RESEARCH LETTERS 2016; 11:305. [PMID: 27325521 PMCID: PMC4916070 DOI: 10.1186/s11671-016-1509-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 05/30/2016] [Indexed: 05/07/2023]
Abstract
In the present study, a highly stable luteinizing-hormone-releasing hormone (LHRH)-conjugated PEGylated poly(lactic-co-glycolic acid) (PLGA) nanoparticles were developed for the successful treatment of prostate cancers. We have demonstrated that a unique combination of targeted drug delivery and controlled drug release is effective against prostate cancer therapy. The docetaxel (DTX)/PLGA-LHRH micelles possessed a uniform spherical shape with an average diameter of ~170 nm. The micelles exhibited a controlled drug release for up to 96 h which can minimize the non-specific systemic spread of toxic drugs during circulation while maximizing the efficiency of tumor-targeted drug delivery. The LHRH-conjugated micelles showed enhanced cellular uptake and exhibited significantly higher cytotoxicity against LNCaP cancer cells. We have showed that PLGA-LHRH induced greater caspase-3 activity indicating its superior apoptosis potential. Consistently, LHRH-conjugated micelles induced threefold and twofold higher G2/M phase arrest than compared to free DTX or PLGA NP-treated groups. Overall, results indicate that use of LHRH-conjugated nanocarriers may potentially be an effective nanocarrier to effectively treat prostate cancer.
Collapse
Affiliation(s)
- Long-Bin Cao
- Department of Urology, Liaocheng Hospital, 67, Dongchang Xi Road, Liaocheng, Shandong, 252000, People's Republic of China.
| | - Sha Zeng
- Central Laboratory, Liaocheng Hospital, Liaocheng, Shandong, 252000, People's Republic of China
| | - Wei Zhao
- Department of Urology, Liaocheng Hospital, 67, Dongchang Xi Road, Liaocheng, Shandong, 252000, People's Republic of China
| |
Collapse
|
36
|
Ye H, Liu X, Sun J, Zhu S, Zhu Y, Chang S. Enhanced therapeutic efficacy of LHRHa-targeted brucea javanica oil liposomes for ovarian cancer. BMC Cancer 2016; 16:831. [PMID: 27793127 PMCID: PMC5086058 DOI: 10.1186/s12885-016-2870-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 10/21/2016] [Indexed: 02/06/2023] Open
Abstract
Background Although brucea javanica oil liposomes (BJOLs) have been used clinically to treat ovarian cancer, its clinical efficacy is often limited by systemic side effects due to non-specific distribution. Luteinizing hormone releasing hormone receptor (LHRHR) is overexpressed in most ovarian cancers but negligibly expressed in most of the other visceral organs. In this study, we aimed to develop a novel LHRHa targeted and BJO-loaded liposomes (LHRHa-BJOLs), and investigate its characteristics, targeting ability and anti-ovarian cancer efficiency both in vitro and in vivo. Methods The LHRHa-BJOLs were prepared by film-dispersion and biotin-streptavidin linkage methods, and characterized in terms of its morphology, particle size, zeta potential, ligand conjugation, encapsulation efficiency and stability. The targeting nature and antitumor effects of the liposomes were evaluated in vitro using cultured human ovarian cancer A2780/DDP cells, and in vivo using ovarian cancer-bearing nude mice. Results The LHRHa-BJOLs were successfully synthesized, with a uniformly spherical shape, appropriate particle size and zeta potential, as well as a high encapsulation efficiency. Compared to non-targeted liposomes and BJO emulsion, the LHRHa-BJOLs could significantly increase specific intracellular uptaking rate, enhance cell inhibitory effect and induce cell apoptosis in A2780/DDP cells in vitro. Meanwhile, LHRHa-BJOLs also had a significantly stronger activity of targeting tumor tissue, inhibiting tumor growth, inducing tumor apoptosis and prolonging survival time in ovarian cancer-bearing mice in vivo. Conclusions Our experiment suggests that LHRHa-BJOLs may be a useful targeted drug for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Hongxia Ye
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong district, Chongqing, 400010, China
| | - Xiaojuan Liu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong district, Chongqing, 400010, China
| | - Jiangchuan Sun
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong district, Chongqing, 400010, China
| | - Shenyin Zhu
- Department of Pharmacy, First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong district, Chongqing, 400010, China
| | - Yi Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong district, Chongqing, 400010, China
| | - Shufang Chang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong district, Chongqing, 400010, China.
| |
Collapse
|
37
|
van Rooij T, Skachkov I, Beekers I, Lattwein KR, Voorneveld JD, Kokhuis TJ, Bera D, Luan Y, van der Steen AF, de Jong N, Kooiman K. Viability of endothelial cells after ultrasound-mediated sonoporation: Influence of targeting, oscillation, and displacement of microbubbles. J Control Release 2016; 238:197-211. [DOI: 10.1016/j.jconrel.2016.07.037] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/20/2016] [Accepted: 07/24/2016] [Indexed: 10/21/2022]
|
38
|
Fan X, Guo Y, Wang L, Xiong X, Zhu L, Fang K. Diagnosis of prostate cancer using anti-PSMA aptamer A10-3.2-oriented lipid nanobubbles. Int J Nanomedicine 2016; 11:3939-50. [PMID: 27574424 PMCID: PMC4990382 DOI: 10.2147/ijn.s112951] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
In this study, the lipid targeted nanobubble carrying the A10-3.2 aptamer against prostate specific membrane antigen was fabricated, and its effect in the ultrasound imaging of prostate cancer was investigated. Materials including 2-dipalmitoyl-sn-glycero-3-phosphocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphatidic acid, 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn-glycero-3-phosphoglycerol, carboxyl-modified 1,2-distearoyl-sn-glycero-3-phosphoethanolamine, and polyethyleneglycol-2000 were for mechanical oscillation, and nanobubbles were obtained through the centrifugal flotation method. After mice were injected with nanobubbles, abdominal color Doppler blood flow imaging significantly improved. Through left ventricular perfusion with normal saline to empty the circulating nanobubbles, nanobubbles still existed in tumor tissue sections, which demonstrated that nanobubbles could enter tissue spaces via the permeability and retention effect. Fluorinated A10-3.2 aptamers obtained by chemical synthesis had good specificity for PSMA-positive cells, and were linked with carboxyl-modified 1,2-distearoyl-sn-glycero-3-phosphoethanolamine lipid molecules from the outer shell of nanobubbles via amide reaction to construct targeted nanobubbles. Gel electrophoresis and immunofluorescence confirmed that targeted nanobubbles were fabricated successfully. Next, targeted nanobubbles could bind with PSMA-positive cells (C4-2 cells), while not with PSMA-negative cells (PC-3 cells), using in vitro binding experiments and flow cytometry at the cellular level. Finally, C4-2 and PC-3 xenografts in mice were used to observe changes in parameters of targeted and non-targeted nanobubbles in the contrast-enhanced ultrasound mode, and the distribution of Cy5.5-labeled targeted nanobubbles in fluorescent imaging of live small animals. Comparison of ultrasound indicators between targeted and non-targeted nanobubbles in C4-2 xenografts showed that they had similar peak times (P>0.05), while the peak intensity, half time of peak intensity, and area under the curve of ½ peak intensity were significantly different (P<0.05). In PC-3 xenografts, there were no differences in these four indicators. Fluorescent imaging indicated that targeted nanobubbles had an aggregation ability in C4-2 xenograft tumors. In conclusion, targeted nanobubbles carrying the anti-PSMA A10-3.2 aptamer have a targeted imaging effect in prostate cancer.
Collapse
Affiliation(s)
- Xiaozhou Fan
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Yanli Guo
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Luofu Wang
- Department of Urology, Daping Hospital, Institute of Surgery Research, Third Military Medical University, Chongqing, People's Republic of China
| | - Xingyu Xiong
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Lianhua Zhu
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Kejing Fang
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| |
Collapse
|
39
|
Yang L, Moghaddas S, Dezvareh H, Belkacemi L, Bark SJ, Bose RN, Do LH. Insights into the anti-angiogenic properties of phosphaplatins. J Inorg Biochem 2016; 164:5-16. [PMID: 27591123 DOI: 10.1016/j.jinorgbio.2016.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 07/19/2016] [Accepted: 07/26/2016] [Indexed: 01/06/2023]
Abstract
Phosphaplatins are platinum-based antitumor compounds that, unlike other clinically utilized platinum drugs (i.e. cisplatin, carboplatin, and oxaliplatin), appear to target proteins rather than DNA. Because of their unique mode of action, phosphaplatins are promising drug candidates for cisplatin-resistant cancers. In this study, we discovered that Pt(II) and Pt(IV) phosphaplatins possess diverse antitumor properties. In addition to targeting apoptosis antigen (FAS) and proapoptotic gene products as described previously, phosphaplatins also target angiogenesis. We demonstrate that phosphaplatins inhibit human umbilical vein endothelial cell (HUVEC) migration and tube formation in vitro and suppress tumor angiogenesis and growth in immunodeficient mice that were inoculated with A2780 ovarian cancer cells in vivo. To provide insight into this novel antitumor mechanism, phosphaplatin-treated HUVECs were found to exhibit lower gene expression levels of vascular endothelial growth factors (VEGFs) and the VEGFR-2 receptor compared to untreated cells. Kinase inhibition studies suggest that phosphaplatins are inhibitors of VEGFR-2. In ligand exchange experiments using both Pt atomic absorption and 31P NMR spectroscopies, we show that phosphaplatins most likely bind to VEGFR-2 through metal-ligand coordination rather than electrostatic interactions. These studies enhance our understanding of the diverse and novel mechanisms of action of the phosphaplatin antitumor agents, which could potentially be used as chemotherapeutic agents against cisplatin-resistant cancers.
Collapse
Affiliation(s)
- Lu Yang
- Departments of Chemistry, University of Houston, Houston, TX 77004, United States
| | - Shadi Moghaddas
- Departments of Biology and Biochemistry, University of Houston, Houston, TX 77004, United States
| | - Homa Dezvareh
- Departments of Biology and Biochemistry, University of Houston, Houston, TX 77004, United States
| | - Louiza Belkacemi
- Departments of Biology and Biochemistry, University of Houston, Houston, TX 77004, United States
| | - Steven J Bark
- Departments of Biology and Biochemistry, University of Houston, Houston, TX 77004, United States
| | - Rathindra N Bose
- Departments of Chemistry, University of Houston, Houston, TX 77004, United States; Departments of Biology and Biochemistry, University of Houston, Houston, TX 77004, United States
| | - Loi H Do
- Departments of Chemistry, University of Houston, Houston, TX 77004, United States.
| |
Collapse
|
40
|
Wang P, Yin T, Li J, Zheng B, Wang X, Wang Y, Zheng J, Zheng R, Shuai X. Ultrasound-responsive microbubbles for sonography-guided siRNA delivery. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1139-1149. [DOI: 10.1016/j.nano.2015.12.361] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 11/07/2015] [Accepted: 12/05/2015] [Indexed: 11/30/2022]
|
41
|
Qin J, Wang TY, Willmann JK. Sonoporation: Applications for Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 880:263-91. [PMID: 26486343 DOI: 10.1007/978-3-319-22536-4_15] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Therapeutic efficacy of both traditional chemotherapy and gene therapy in cancer is highly dependent on the ability to deliver drugs across natural barriers, such as the vessel wall or tumor cell membranes. In this regard, sonoporation induced by ultrasound-guided microbubble (USMB) destruction has been widely investigated in the enhancement of therapeutic drug delivery given it can help overcome these natural barriers, thereby increasing drug delivery into cancer. In this chapter we discuss challenges in current cancer therapy and how some of these challenges could be overcome using USMB-mediated drug delivery. We particularly focus on recent advances in delivery approaches that have been developed to further improve therapeutic efficiency and specificity of various cancer treatments. An example of clinical translation of USMB-mediated drug delivery is also shown.
Collapse
Affiliation(s)
- Jiale Qin
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, School of Medicine, Stanford, CA, USA
| | - Tzu-Yin Wang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, School of Medicine, Stanford, CA, USA
| | - Jürgen K Willmann
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, School of Medicine, Stanford, CA, USA.
| |
Collapse
|
42
|
Sun J, Yin M, Zhu S, Liu L, Zhu Y, Wang Z, Xu RX, Chang S. Ultrasound-mediated destruction of oxygen and paclitaxel loaded lipid microbubbles for combination therapy in hypoxic ovarian cancer cells. ULTRASONICS SONOCHEMISTRY 2016; 28:319-326. [PMID: 26384914 DOI: 10.1016/j.ultsonch.2015.08.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 08/14/2015] [Accepted: 08/15/2015] [Indexed: 06/05/2023]
Abstract
We synthesized oxygen and paclitaxel (PTX) loaded lipid microbubbles (OPLMBs) for ultrasound mediated combination therapy in hypoxic ovarian cancer cells. Our experiments successfully demonstrated that ultrasound induced OPLMBs destruction significantly enhanced the local oxygen release. We also demonstrated that OPLMBs in combination with ultrasound (300 kHz, 0.5 W/cm(2), 15s) yielded anti-proliferative activities of 52.8 ± 2.75% and cell apoptosis ratio of 35.25 ± 0.17% in hypoxic cells at 24h after the treatment, superior to other treatment groups such as PTX only and PTX-loaded MBs (PLMBs) with or without ultrasound mediation. RT-PCR and Western blot tests further confirmed the reduced expression of HIF-1α and MDR-1/P-gp after ultrasound mediation of OPLMBs. Our experiment suggests that ultrasound mediation of oxygen and drug-loaded MBs may be a useful method to overcome chemoresistance in the hypoxic ovarian cancer cells.
Collapse
Affiliation(s)
- Jiangchuan Sun
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China; Institute of Ultrasound Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Mingyue Yin
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Shenyin Zhu
- Department of Pharmacy, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Li Liu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yi Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhigang Wang
- Institute of Ultrasound Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Ronald X Xu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China; Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Shufang Chang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
43
|
Chang S, Si T, Zhang S, Merrick MA, Cohn DE, Xu RX. Ultrasound mediated destruction of multifunctional microbubbles for image guided delivery of oxygen and drugs. ULTRASONICS SONOCHEMISTRY 2016; 28:31-38. [PMID: 26384880 DOI: 10.1016/j.ultsonch.2015.06.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 05/18/2015] [Accepted: 06/24/2015] [Indexed: 06/05/2023]
Abstract
We synthesized multifunctional activatible microbubbles (MAMs) for ultrasound mediated delivery of oxygen and drugs with both ultrasound and fluorescence imaging guidance. Oxygen enriched perfluorocarbon (PFC) compound was encapsulated in liposome microbubbles (MBs) by a modified emulsification process. DiI dye was loaded as a model drug. The ultrasound targeted microbubble destruction (UTMD) process was guided by both ultrasonography and fluorescence imaging modalities. The process was validated in both a dialysis membrane tube model and a porcine carotid artery model. Our experiment results show that the UTMD process effectively facilitates the controlled delivery of oxygen and drug at the disease site and that the MAM agent enables ultrasound and fluorescence imaging guidance of the UTMD process. The proposed MAM agent can be potentially used for UTMD-mediated combination therapy in hypoxic ovarian cancer.
Collapse
Affiliation(s)
- Shufang Chang
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Ting Si
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; College of Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Shiwu Zhang
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; College of Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Mark A Merrick
- Division of Health & Rehabilitation Sciences, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - David E Cohn
- Department of Obstetrics and Gynecology, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Ronald X Xu
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; College of Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
44
|
Raavé R, de Vries RBM, Massuger LF, van Kuppevelt TH, Daamen WF. Drug delivery systems for ovarian cancer treatment: a systematic review and meta-analysis of animal studies. PeerJ 2015; 3:e1489. [PMID: 26713240 PMCID: PMC4690347 DOI: 10.7717/peerj.1489] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/19/2015] [Indexed: 01/04/2023] Open
Abstract
Current ovarian cancer treatment involves chemotherapy that has serious limitations, such as rapid clearance, unfavorable biodistribution and severe side effects. To overcome these limitations, drug delivery systems (DDS) have been developed to encapsulate chemotherapeutics for delivery to tumor cells. However, no systematic assessment of the efficacy of chemotherapy by DDS compared to free chemotherapy (not in a DDS) has been performed for animal studies. Here, we assess the efficacy of chemotherapy in DDS on survival and tumor growth inhibition in animal studies. We searched PubMed and EMBASE (via OvidSP) to systematically identify studies evaluating chemotherapeutics encapsulated in DDS for ovarian cancer treatment in animal studies. Studies were assessed for quality and risk of bias. Study characteristics were collected and outcome data (survival/hazard ratio or tumor growth inhibition) were extracted and used for meta-analyses. Meta-analysis was performed to identify and explore which characteristics of DDS influenced treatment efficacy. A total of 44 studies were included after thorough literature screening (2,735 studies found after initial search). The risk of bias was difficult to assess, mainly because of incomplete reporting. A total of 17 studies (377 animals) and 16 studies (259 animals) could be included in the meta-analysis for survival and tumor growth inhibition, respectively. In the majority of the included studies chemotherapeutics entrapped in a DDS significantly improved efficacy over free chemotherapeutics regarding both survival and tumor growth inhibition. Subgroup analyses, however, revealed that cisplatin entrapped in a DDS did not result in additional tumor growth inhibition compared to free cisplatin, although it did result in improved survival. Micelles did not show a significant tumor growth inhibition compared to free chemotherapeutics, which indicates that micelles may not be a suitable DDS for ovarian cancer treatment. Other subgroup analyses, such as targeted versus non-targeted DDS or IV versus IP administration route, did not identify specific characteristics of DDS that affected treatment efficacy. This systematic review shows the potential, but also the limitations of chemotherapy by drug delivery systems for ovarian cancer treatment. For future animal research, we emphasize that data need to be reported with ample attention to detailed reporting.
Collapse
Affiliation(s)
- René Raavé
- Department of Biochemistry, Radboud university medical center , Nijmegen , The Netherlands
| | - Rob B M de Vries
- Systematic Review Centre for Laboratory Animal Experimentation, Central Animal Facility, Radboud university medical center , Nijmegen , The Netherlands
| | - Leon F Massuger
- Department of Obstetrics and Gynaecology, Radboud university medical center , Nijmegen , The Netherlands
| | - Toin H van Kuppevelt
- Department of Biochemistry, Radboud university medical center , Nijmegen , The Netherlands
| | - Willeke F Daamen
- Department of Biochemistry, Radboud university medical center , Nijmegen , The Netherlands
| |
Collapse
|
45
|
ZHOU SHOUBING, ZHENG SHIYA, SHAN YONGFENG, LI LULU, ZHANG XIU, WANG CAILIAN. Rituximab-conjugated and doxorubicin-loaded microbubbles combined with ultrasound irradiation inhibits proliferation and induces apoptosis in Raji cell lines. Oncol Rep 2015; 35:801-8. [DOI: 10.3892/or.2015.4468] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 10/11/2015] [Indexed: 11/06/2022] Open
|
46
|
Zhang Y, Chang S, Sun J, Zhu S, Pu C, Li Y, Zhu Y, Wang Z, Xu RX. Targeted Microbubbles for Ultrasound Mediated Short Hairpin RNA Plasmid Transfection to Inhibit Survivin Gene Expression and Induce Apoptosis of Ovarian Cancer A2780/DDP Cells. Mol Pharm 2015. [PMID: 26212628 DOI: 10.1021/mp500835z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Yong Zhang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
- National Engineering Research Center of Ultrasound Medicine, Chongqing 400010,China
| | - Shufang Chang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jiangchuan Sun
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Shenyin Zhu
- Department
of Pharmacy, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Caixiu Pu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yaowei Li
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yi Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhigang Wang
- Institute of Ultrasound Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Ronald X. Xu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
- Department
of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
47
|
Abstract
The combination of microbubbles and ultrasound has emerged as a promising method for local drug delivery. Microbubbles can be locally activated by a targeted ultrasound beam, which can result in several bio-effects. For drug delivery, microbubble-assisted ultrasound is used to increase vascular- and plasma membrane permeability for facilitating drug extravasation and the cellular uptake of drugs in the treated region, respectively. In the case of drug-loaded microbubbles, these two mechanisms can be combined with local release of the drug following destruction of the microbubble. The use of microbubble-assisted ultrasound to deliver chemotherapeutic agents is also referred to as sonochemotherapy. In this review, the basic principles of sonochemotherapy are discussed, including aspects such as the type of (drug-loaded) microbubbles used, the routes of administration used in vivo, ultrasound devices and parameters, treatment schedules and safety issues. Finally, the clinical translation of sonochemotherapy is discussed, including the first clinical study using sonochemotherapy.
Collapse
Affiliation(s)
- Bart H A Lammertink
- Image Guided Therapy, Imaging Division, University Medical Center Utrecht Utrecht, Netherlands
| | - Clemens Bos
- Image Guided Therapy, Imaging Division, University Medical Center Utrecht Utrecht, Netherlands
| | - Roel Deckers
- Image Guided Therapy, Imaging Division, University Medical Center Utrecht Utrecht, Netherlands
| | - Gert Storm
- Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University Utrecht, Netherlands ; Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente Enschede, Netherlands
| | - Chrit T W Moonen
- Image Guided Therapy, Imaging Division, University Medical Center Utrecht Utrecht, Netherlands
| | - Jean-Michel Escoffre
- Image Guided Therapy, Imaging Division, University Medical Center Utrecht Utrecht, Netherlands
| |
Collapse
|
48
|
Ma J, Xu CS, Gao F, Chen M, Li F, Du LF. Diagnostic and therapeutic research on ultrasound microbubble/nanobubble contrast agents (Review). Mol Med Rep 2015; 12:4022-4028. [PMID: 26081968 DOI: 10.3892/mmr.2015.3941] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 12/12/2014] [Indexed: 11/06/2022] Open
Abstract
The contrast enhanced imaging function of ultrasound contrast agents (UCAs) has been extensively investigated using physical acoustic signatures. It has a number of novel applications, including tissue‑specific molecular imaging and multi‑modal imaging. In addition there are numerous other therapeutic applications of UCAs, for example as vehicles for drug or gene delivery. These uses are discussed, as well as the acoustically‑induced biological effects, including ultrasound targeted microbubble destruction (UTMD). This review also explores the considerations for the safe use of UCA from an acoustic standpoint. The scope of the application of UCA has markedly expanded in recent years, and it is a rapidly growing field of medical research. The current article reviews recent advances in the diagnostic and therapeutic applications of ultrasound microbubble/nanobubble contrast agents.
Collapse
Affiliation(s)
- Jing Ma
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Chang Song Xu
- Department of Ultrasound, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Feng Gao
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Ming Chen
- Department of Cardiovascular Ultrasound, Shanghai East Hospital Affiliated to Tongji University, Shanghai 200120, P.R. China
| | - Fan Li
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Lian Fang Du
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| |
Collapse
|
49
|
Hooker BA, Blomme EAG. Targeted drug delivery in oncology: where drug discovery meets physics. Vet J 2015; 205:120-1. [PMID: 26021890 DOI: 10.1016/j.tvjl.2015.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 04/28/2015] [Indexed: 11/16/2022]
Affiliation(s)
- Bradley A Hooker
- Integrated Science and Technology, Translational Imaging, AbbVie, North Chicago, IL, USA.
| | - Eric A G Blomme
- Scientific Editor, The Veterinary Journal; Senior Research Fellow, Investigative Toxicology and Pathology, AbbVie, North Chicago, IL, USA.
| |
Collapse
|
50
|
Liu L, Chang S, Sun J, Zhu S, Yin M, Zhu Y, Wang Z, Xu RX. Ultrasound-mediated destruction of paclitaxel and oxygen loaded lipid microbubbles for combination therapy in ovarian cancer xenografts. Cancer Lett 2015; 361:147-54. [PMID: 25754815 DOI: 10.1016/j.canlet.2015.02.052] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/23/2015] [Accepted: 02/27/2015] [Indexed: 12/18/2022]
Abstract
We have synthesized multifunctional oxygen and paclitaxel loaded microbubbles (OPLMBs) for ultrasound mediated delivery of combination therapy in an ovarian cancer xenograft model. In comparison with other therapeutic options, intravenous injection of OPLMBs followed by ultrasound mediation yielded a superior therapeutic outcome. Immunohistochemical analyses of the dissected tumor tissue confirmed the increased tumor apoptosis and the reduced VEGF expression after treatment. Western Blot tests further confirmed the decreased expressions of HIF-1α and P-gp. Our experiment suggests that ultrasound mediation of OPLMBs may provide a promising drug delivery strategy for the combination treatment of ovarian cancer.
Collapse
Affiliation(s)
- Li Liu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China; Institute of Ultrasound Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China; State Key Laboratory of Ultrasound Engineering in Medicine Co-founded by Chongqing and the Ministry of Science and Technology, Chongqing 400016, China
| | - Shufang Chang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Jiangchuan Sun
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Shenyin Zhu
- Department of Pharmacy, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Minyue Yin
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yi Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhigang Wang
- Institute of Ultrasound Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Ronald X Xu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China; Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|