1
|
Chulanova Y, Breier D, Peer D. Delivery of genetic medicines for muscular dystrophies. Cell Rep Med 2025; 6:101885. [PMID: 39765231 DOI: 10.1016/j.xcrm.2024.101885] [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: 04/02/2024] [Revised: 08/29/2024] [Accepted: 12/05/2024] [Indexed: 01/24/2025]
Abstract
Muscular dystrophies are a group of heterogenic disorders characterized by progressive muscle weakness, the most common of them being Duchenne muscular dystrophy (DMD). Muscular dystrophies are caused by mutations in over 50 distinct genes, and many of them are caused by different genetic mechanisms. Currently, none of these diseases have a cure. However, in recent years, significant progress has been made to correct the underlying genetic cause. The clinical development of adeno-associated viral vector-based therapies has simultaneously produced excitement and disappointment in the research community due to the moderate effect, making it clear that new methods of muscle delivery have to be created. Herein, we review the main characteristics of major muscular dystrophies and outline various muscle-targeted delivery methods being explored for genetic medicines.
Collapse
Affiliation(s)
- Yulia Chulanova
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel-Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dor Breier
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel-Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dan Peer
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel-Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
2
|
Jayasankar G, Koilpillai J, Narayanasamy D. A Systematic Study on Long-acting Nanobubbles: Current Advancement and Prospects on Theranostic Properties. Adv Pharm Bull 2024; 14:278-301. [PMID: 39206408 PMCID: PMC11347731 DOI: 10.34172/apb.2024.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/16/2024] [Accepted: 03/17/2024] [Indexed: 09/04/2024] Open
Abstract
Delivery of diagnostic drugs via nanobubbles (NBs) has shown to be an emerging field of study. Due to their small size, NBs may more easily travel through constricted blood vessels and precisely target certain bodily parts. NB is considered the major treatment for cancer treatment and other diseases which are difficult to diagnose. The field of NBs is dynamic and continues to grow as researchers discover new properties and seek practical applications in various fields. The predominant usage of NBs in novel drug delivery is to enhance the bioavailability, and controlled drug release along with imaging properties NBs are important because they may change interfacial characteristics including surface force, lubrication, and absorption. The quick diffusion of gas into the water was caused by a hypothetical film that was stimulated and punctured by a strong acting force at the gas/water contact of the bubble. In this article, various prominent aspects of NBs have been discussed, along with the long-acting nature, and the theranostical aspect which elucidates the potential marketed drugs along with clinical trial products. The article also covers quality by design aspects, different production techniques that enable method-specific therapeutic applications, increasing the floating time of the bubble, and refining its properties to enhance the prepared NB's quality. NB containing both analysis and curing properties makes it special from other nano-carriers. This work includes all the possible methods of preparing NB, its application, all marketed drugs, and products in clinical trials.
Collapse
Affiliation(s)
| | | | - Damodharan Narayanasamy
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institution of Science and Technology, Kattankulathur, Chengalpattu, India
| |
Collapse
|
3
|
Sotoudeh Bagha P, Kolanthai E, Wei F, Neal CJ, Kumar U, Braun G, Coathup M, Seal S, Razavi M. Ultrasound-Responsive Nanobubbles for Combined siRNA-Cerium Oxide Nanoparticle Delivery to Bone Cells. Pharmaceutics 2023; 15:2393. [PMID: 37896153 PMCID: PMC10609961 DOI: 10.3390/pharmaceutics15102393] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
This study aims to present an ultrasound-mediated nanobubble (NB)-based gene delivery system that could potentially be applied in the future to treat bone disorders such as osteoporosis. NBs are sensitive to ultrasound (US) and serve as a controlled-released carrier to deliver a mixture of Cathepsin K (CTSK) siRNA and cerium oxide nanoparticles (CeNPs). This platform aimed to reduce bone resorption via downregulating CTSK expression in osteoclasts and enhance bone formation via the antioxidant and osteogenic properties of CeNPs. CeNPs were synthesized and characterized using transmission electron microscopy and X-ray photoelectron spectroscopy. The mixture of CTSK siRNA and CeNPs was adsorbed to the surface of NBs using a sonication method. The release profiles of CTSK siRNA and CeNPs labeled with a fluorescent tag molecule were measured after low-intensity pulsed ultrasound (LIPUS) stimulation using fluorescent spectroscopy. The maximum release of CTSK siRNA and the CeNPs for 1 mg/mL of NB-(CTSK siRNA + CeNPs) was obtained at 2.5 nM and 1 µg/mL, respectively, 3 days after LIPUS stimulation. Then, Alizarin Red Staining (ARS) was applied to human bone marrow-derived mesenchymal stem cells (hMSC) and tartrate-resistant acid phosphatase (TRAP) staining was applied to human osteoclast precursors (OCP) to evaluate osteogenic promotion and osteoclastogenic inhibition effects. A higher mineralization and a lower number of osteoclasts were quantified for NB-(CTSK siRNA + CeNPs) versus control +RANKL with ARS (p < 0.001) and TRAP-positive staining (p < 0.01). This study provides a method for the delivery of gene silencing siRNA and CeNPs using a US-sensitive NB system that could potentially be used in vivo and in the treatment of bone fractures and disorders such as osteoporosis.
Collapse
Affiliation(s)
- Pedram Sotoudeh Bagha
- BiionixTM (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA; (P.S.B.); (F.W.); (M.C.)
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (E.K.); (C.J.N.); (U.K.); (S.S.)
| | - Fei Wei
- BiionixTM (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA; (P.S.B.); (F.W.); (M.C.)
| | - Craig J. Neal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (E.K.); (C.J.N.); (U.K.); (S.S.)
| | - Udit Kumar
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (E.K.); (C.J.N.); (U.K.); (S.S.)
| | - Gillian Braun
- Department of Biological Sciences, Mount Holyoke College, South Hadley, MA 01075, USA;
| | - Melanie Coathup
- BiionixTM (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA; (P.S.B.); (F.W.); (M.C.)
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (E.K.); (C.J.N.); (U.K.); (S.S.)
| | - Mehdi Razavi
- BiionixTM (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA; (P.S.B.); (F.W.); (M.C.)
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
4
|
Abstract
The conventional microbubble-based ultrasound biomedicine clinically plays a vital role in providing the dynamic detection of macro and microvasculature and disease theranostics. However, the intrinsic limitation of particle size severely decreases the treatment effectiveness due to their vascular transport characteristics, which promotes the development and application of multifunctional ultrasound-responsive nanomaterials. Herein, we put forward a research field of "ultrasound nanomedicine and materdicine", referring to the interdiscipline of ultrasound, nanobiotechnology and materials, which seeks to produce specific biological effects for addressing the challenges faced and dilemma of conventional ultrasound medicine. We comprehensively summarize the state-of-the-art scientific advances in the latest progress in constructing ultrasound-based platforms and ultrasound-activated sonosensitizers, ranging from the synthesis strategies, biological functions to ultrasound-triggered therapeutic applications. Ultimately, the unresolved challenges and clinical-translation potentials of ultrasound nanomedicine and materdicine are discussed and prospected in this evolving field.
Collapse
Affiliation(s)
- Zeyu Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Xue Wang
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Meiqi Chang
- Central Laboratory of Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China.
| | - Jia Guo
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| |
Collapse
|
5
|
Pasupathy R, Pandian P, Selvamuthukumar S. Nanobubbles: A Novel Targeted Drug Delivery System. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e19604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
6
|
Zeng F, Du M, Chen Z. Nanosized Contrast Agents in Ultrasound Molecular Imaging. Front Bioeng Biotechnol 2021; 9:758084. [PMID: 34912789 PMCID: PMC8666542 DOI: 10.3389/fbioe.2021.758084] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Applying nanosized ultrasound contrast agents (nUCAs) in molecular imaging has received considerable attention. nUCAs have been instrumental in ultrasound molecular imaging to enhance sensitivity, identification, and quantification. nUCAs can achieve high performance in molecular imaging, which was influenced by synthetic formulations and size. This review presents an overview of nUCAs from different synthetic formulations with a discussion on imaging and detection technology. Then we also review the progress of nUCAs in preclinical application and highlight the recent challenges of nUCAs.
Collapse
Affiliation(s)
- Fengyi Zeng
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, China.,Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China.,Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Meng Du
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, China.,Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhiyi Chen
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, China.,Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
7
|
Walsh AP, Gordon HN, Peter K, Wang X. Ultrasonic particles: An approach for targeted gene delivery. Adv Drug Deliv Rev 2021; 179:113998. [PMID: 34662671 PMCID: PMC8518240 DOI: 10.1016/j.addr.2021.113998] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/24/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023]
Abstract
Gene therapy has been widely investigated for the treatment of genetic, acquired, and infectious diseases. Pioneering work utilized viral vectors; however, these are suspected of causing serious adverse events, resulting in the termination of several clinical trials. Non-viral vectors, such as lipid nanoparticles, have attracted significant interest, mainly due to their successful use in vaccines in the current COVID-19 pandemic. Although they allow safe delivery, they come with the disadvantage of off-target delivery. The application of ultrasound to ultrasound-sensitive particles allows for a direct, site-specific transfer of genetic materials into the organ/site of interest. This process, termed ultrasound-targeted gene delivery (UTGD), also increases cell membrane permeability and enhances gene uptake. This review focuses on the advances in ultrasound and the development of ultrasonic particles for UTGD across a range of diseases. Furthermore, we discuss the limitations and future perspectives of UTGD.
Collapse
Affiliation(s)
- Aidan P.G. Walsh
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Henry N. Gordon
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Biochemistry and Pharmacology, University of Melbourne, VIC, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Medicine, Monash University, Melbourne, VIC, Australia,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Xiaowei Wang
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Medicine, Monash University, Melbourne, VIC, Australia,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia,Corresponding author at: Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| |
Collapse
|
8
|
Xie L, Wang J, Zhao S, Lai ML, Jiang T, Yan F. An acoustic field-based conformal transfection system for improving the gene delivery efficiency. Biomater Sci 2021; 9:4127-4138. [PMID: 33954320 DOI: 10.1039/d1bm00251a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Ultrasound-activated microbubble destruction is a promising platform for gene delivery due to the low toxicity, non-invasiveness, and high specificity. However, the gene transfection efficiency is still low, especially for suspension cells. It is desirable to develop a universal gene delivery tool that overcomes the drawbacks existing in ultrasound-mediated methods. Here, we present a three-dimensional acoustic field-based conformal transfection (AFCT) system by designing a Sono-hole that can fit the three-dimensional acoustic field to maximally utilize the acoustic energy from bubble cavitation, thus greatly promoting the gene delivery efficiency. Surprisingly, compared with the traditional two-dimensional transfection system, the gene transfection efficiency of the AFCT system increased by more than 3 times, achieving nearly 30%. The parameters including acoustic pressure, duration, duty cycle, DNA concentrations, and bubble kinds were optimized to obtain higher gene transfection. In conclusion, our study provides an effective ultrasound-based gene delivery approach for gene transfection, especially for suspension-cultured cells.
Collapse
Affiliation(s)
- Liting Xie
- Department of Ultrasound, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China. and CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Jieqiong Wang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Shuai Zhao
- Department of Ultrasound, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Man Lin Lai
- Department of Ultrasound, The First Affiliated Hospital, Shenzhen University school of medicine, Shenzhen, 518061, China
| | - Tianan Jiang
- Department of Ultrasound, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Fei Yan
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
9
|
Suppression of Peritoneal Fibrosis by Sonoporation of Hepatocyte Growth Factor Gene-Encoding Plasmid DNA in Mice. Pharmaceutics 2021; 13:pharmaceutics13010115. [PMID: 33477422 PMCID: PMC7829751 DOI: 10.3390/pharmaceutics13010115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/15/2021] [Accepted: 01/15/2021] [Indexed: 12/14/2022] Open
Abstract
Gene therapy is expected to be used for the treatment of peritoneal fibrosis, which is a serious problem associated with long-term peritoneal dialysis. Hepatocyte growth factor (HGF) is a well-known anti-fibrotic gene. We developed an ultrasound and nanobubble-mediated (sonoporation) gene transfection system, which selectively targets peritoneal tissues. Thus, we attempted to treat peritoneal fibrosis by sonoporation-based human HGF (hHGF) gene transfection in mice. To prepare a model of peritoneal fibrosis, mice were intraperitoneally injected with chlorhexidine digluconate. We evaluated the preventive and curative effects of sonoporation-based hHGF transfection by analyzing the following factors: hydroxyproline level, peritoneum thickness, and the peritoneal equilibration test. The transgene expression characteristics of sonoporation were also evaluated using multicolor deep imaging. In early-stage fibrosis in mice, transgene expression by sonoporation was observed in the submesothelial layer. Sonoporation-based hHGF transfection showed not only a preventive effect but also a curative effect for early-stage peritoneal fibrosis. Sonoporation-based hHGF transfection may be suitable for the treatment of peritoneal fibrosis regarding the transfection characteristics of transgene expression in the peritoneum under fibrosis.
Collapse
|
10
|
Su C, Ren X, Nie F, Li T, Lv W, Li H, Zhang Y. Current advances in ultrasound-combined nanobubbles for cancer-targeted therapy: a review of the current status and future perspectives. RSC Adv 2021; 11:12915-12928. [PMID: 35423829 PMCID: PMC8697319 DOI: 10.1039/d0ra08727k] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/16/2021] [Indexed: 12/14/2022] Open
Abstract
The non-specific distribution, non-selectivity towards cancerous cells, and adverse off-target side effects of anticancer drugs and other therapeutic molecules lead to their inferior clinical efficacy. Accordingly, ultrasound-based targeted delivery of therapeutic molecules loaded in smart nanocarriers is currently gaining wider acceptance for the treatment and management of cancer. Nanobubbles (NBs) are nanosize carriers, which are currently used as effective drug/gene delivery systems because they can deliver drugs/genes selectively to target sites. Thus, combining the applications of ultrasound with NBs has recently demonstrated increased localization of anticancer molecules in tumor tissues with triggered release behavior. Consequently, an effective therapeutic concentration of drugs/genes is achieved in target tumor tissues with ultimately increased therapeutic efficacy and minimal side-effects on other non-cancerous tissues. This review illustrates present developments in the field of ultrasound-nanobubble combined strategies for targeted cancer treatment. The first part of this review discusses the composition and the formulation parameters of NBs. Next, we illustrate the interactions and biological effects of combining NBs and ultrasound. Subsequently, we explain the potential of NBs combined with US for targeted cancer therapeutics. Finally, the present and future directions for the improvement of current methods are proposed. NBs combined with ultrasound demonstrated the ability to enhance the targeting of anticancer agents and improve the efficacy.![]()
Collapse
Affiliation(s)
- Chunhong Su
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
- Department of Pain, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
| | - XiaoJun Ren
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
| | - Fang Nie
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
| | - Tiangang Li
- Department of Ultrasound Diagnosis, Gansu Provincial Maternity and Child-Care Hospital, Lanzhou, 730030, Gansu Province, China
| | - Wenhao Lv
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
| | - Hui Li
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
- Department of Pneumology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
| | - Yao Zhang
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
- Department of Emergency, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
| |
Collapse
|
11
|
Decker RE, Lamantia ZE, Emrick TS, Figueiredo ML. Sonodelivery in Skeletal Muscle: Current Approaches and Future Potential. Bioengineering (Basel) 2020; 7:E107. [PMID: 32916815 PMCID: PMC7552685 DOI: 10.3390/bioengineering7030107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/27/2020] [Accepted: 09/04/2020] [Indexed: 11/16/2022] Open
Abstract
There are currently multiple approaches to facilitate gene therapy via intramuscular gene delivery, such as electroporation, viral delivery, or direct DNA injection with or without polymeric carriers. Each of these methods has benefits, but each method also has shortcomings preventing it from being established as the ideal technique. A promising method, ultrasound-mediated gene delivery (or sonodelivery) is inexpensive, widely available, reusable, minimally invasive, and safe. Hurdles to utilizing sonodelivery include choosing from a large variety of conditions, which are often dependent on the equipment and/or research group, and moderate transfection efficiencies when compared to some other gene delivery methods. In this review, we provide a comprehensive look at the breadth of sonodelivery techniques for intramuscular gene delivery and suggest future directions for this continuously evolving field.
Collapse
Affiliation(s)
- Richard E. Decker
- Department of Basic Medical Sciences, Purdue University, 625 Harrison St., West Lafayette, IN 47907, USA; (R.E.D.); (Z.E.L.)
| | - Zachary E. Lamantia
- Department of Basic Medical Sciences, Purdue University, 625 Harrison St., West Lafayette, IN 47907, USA; (R.E.D.); (Z.E.L.)
| | - Todd S. Emrick
- Department of Polymer Science & Engineering, University of Massachusetts, 120 Governors Drive, Amherst, MA 01003, USA;
| | - Marxa L. Figueiredo
- Department of Basic Medical Sciences, Purdue University, 625 Harrison St., West Lafayette, IN 47907, USA; (R.E.D.); (Z.E.L.)
| |
Collapse
|
12
|
Brzoska E, Kalkowski L, Kowalski K, Michalski P, Kowalczyk P, Mierzejewski B, Walczak P, Ciemerych MA, Janowski M. Muscular Contribution to Adolescent Idiopathic Scoliosis from the Perspective of Stem Cell-Based Regenerative Medicine. Stem Cells Dev 2020; 28:1059-1077. [PMID: 31170887 DOI: 10.1089/scd.2019.0073] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is a relatively frequent disease within a range 0.5%-5.0% of population, with higher frequency in females. While a resultant spinal deformity is usually medically benign condition, it produces far going psychosocial consequences, which warrant attention. The etiology of AIS is unknown and current therapeutic approaches are symptomatic only, and frequently inconvenient or invasive. Muscular contribution to AIS is widely recognized, although it did not translate to clinical routine as yet. Muscle asymmetry has been documented by pathological examinations as well as systemic muscle disorders frequently leading to scoliosis. It has been also reported numerous genetic, metabolic and radiological alterations in patients with AIS, which are linked to muscular and neuromuscular aspects. Therefore, muscles might be considered an attractive and still insufficiently exploited therapeutic target for AIS. Stem cell-based regenerative medicine is rapidly gaining momentum based on the tremendous progress in understanding of developmental biology. It comes also with a toolbox of various stem cells such as satellite cells or mesenchymal stem cells, which could be transplanted; also, the knowledge acquired in research on regenerative medicine can be applied to manipulation of endogenous stem cells to obtain desired therapeutic goals. Importantly, paravertebral muscles are located relatively superficially; therefore, they can be an easy target for minimally invasive approaches to treatment of AIS. It comes in pair with a fast progress in image guidance, which allows for precise delivery of therapeutic agents, including stem cells to various organs such as brain, muscles, and others. Summing up, it seems that there is a link between AIS, muscles, and stem cells, which might be worth of further investigations with a long-term goal of setting foundations for eventual bench-to-bedside translation.
Collapse
Affiliation(s)
- Edyta Brzoska
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Lukasz Kalkowski
- 2Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Kamil Kowalski
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Pawel Michalski
- 3Spine Surgery Department, Institute of Mother and Child, Warsaw, Poland
| | - Pawel Kowalczyk
- 4Department of Neurosurgery, Children's Memorial Health Institute, Warsaw, Poland
| | - Bartosz Mierzejewski
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Piotr Walczak
- 5Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,6Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Maria A Ciemerych
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Miroslaw Janowski
- 5Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,6Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
13
|
Cai X, Jiang Y, Lin M, Zhang J, Guo H, Yang F, Leung W, Xu C. Ultrasound-Responsive Materials for Drug/Gene Delivery. Front Pharmacol 2020; 10:1650. [PMID: 32082157 PMCID: PMC7005489 DOI: 10.3389/fphar.2019.01650] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/16/2019] [Indexed: 12/15/2022] Open
Abstract
Ultrasound is one of the most commonly used methods in the diagnosis and therapy of diseases due to its safety, deep penetration into tissue, and non-invasive nature. In the drug/gene delivery systems, ultrasound shows many advantages in terms of site-specific delivery and spatial release control of drugs/genes and attracts increasing attention. Microbubbles are the most well-known ultrasound-responsive delivery materials. Recently, nanobubbles, droplets, micelles, and nanoliposomes have been developed as novel carriers in this field. Herein, we review advances of novel ultrasound-responsive materials (nanobubbles, droplets, micelles and nanoliposomes) and discuss the challenges of ultrasound-responsive materials in delivery systems to boost the development of ultrasound-responsive materials as delivery carriers.
Collapse
Affiliation(s)
- Xiaowen Cai
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuan Jiang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mei Lin
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiyong Zhang
- Department of Pediatrics, Shenzhen Maternity and Child Health Care Hospital, Shenzhen, China
| | - Huanhuan Guo
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Fanwen Yang
- Department of Biomedical Engineering, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Wingnang Leung
- Asia-Pacific Institute of Aging Studies, Lingnan University, Tuen Mun, Hong Kong, Hong Kong
| | - Chuanshan Xu
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
14
|
Nonviral ultrasound-mediated gene delivery in small and large animal models. Nat Protoc 2019; 14:1015-1026. [PMID: 30804568 DOI: 10.1038/s41596-019-0125-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 12/18/2018] [Indexed: 12/15/2022]
Abstract
Ultrasound-mediated gene delivery (sonoporation) is a minimally invasive, nonviral and clinically translatable method of gene therapy. This method offers a favorable safety profile over that of viral vectors and is less invasive as compared with other physical gene delivery approaches (e.g., electroporation). We have previously used sonoporation to overexpress transgenes in different skeletal tissues in order to induce tissue regeneration. Here, we provide a protocol that could easily be adapted to address various other targets of tissue regeneration or additional applications, such as cancer and neurodegenerative diseases. This protocol describes how to prepare, conduct and optimize ultrasound-mediated gene delivery in both a murine and a porcine animal model. The protocol includes the preparation of a microbubble-DNA mix and in vivo sonoporation under ultrasound imaging. Ultrasound-mediated gene delivery can be accomplished within 10 min. After DNA delivery, animals can be followed to monitor gene expression, protein secretion and other transgene-specific outcomes, including tissue regeneration. This procedure can be accomplished by a competent graduate student or technician with prior experience in ultrasound imaging or in performing in vivo procedures.
Collapse
|
15
|
Bez M, Sheyn D, Tawackoli W, Avalos P, Shapiro G, Giaconi JC, Da X, David SB, Gavrity J, Awad HA, Bae HW, Ley EJ, Kremen TJ, Gazit Z, Ferrara KW, Pelled G, Gazit D. In situ bone tissue engineering via ultrasound-mediated gene delivery to endogenous progenitor cells in mini-pigs. Sci Transl Med 2018; 9:9/390/eaal3128. [PMID: 28515335 DOI: 10.1126/scitranslmed.aal3128] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/08/2017] [Accepted: 04/14/2017] [Indexed: 12/20/2022]
Abstract
More than 2 million bone-grafting procedures are performed each year using autografts or allografts. However, both options carry disadvantages, and there remains a clear medical need for the development of new therapies for massive bone loss and fracture nonunions. We hypothesized that localized ultrasound-mediated, microbubble-enhanced therapeutic gene delivery to endogenous stem cells would induce efficient bone regeneration and fracture repair. To test this hypothesis, we surgically created a critical-sized bone fracture in the tibiae of Yucatán mini-pigs, a clinically relevant large animal model. A collagen scaffold was implanted in the fracture to facilitate recruitment of endogenous mesenchymal stem/progenitor cells (MSCs) into the fracture site. Two weeks later, transcutaneous ultrasound-mediated reporter gene delivery successfully transfected 40% of cells at the fracture site, and flow cytometry showed that 80% of the transfected cells expressed MSC markers. Human bone morphogenetic protein-6 (BMP-6) plasmid DNA was delivered using ultrasound in the same animal model, leading to transient expression and secretion of BMP-6 localized to the fracture area. Micro-computed tomography and biomechanical analyses showed that ultrasound-mediated BMP-6 gene delivery led to complete radiographic and functional fracture healing in all animals 6 weeks after treatment, whereas nonunion was evident in control animals. Collectively, these findings demonstrate that ultrasound-mediated gene delivery to endogenous mesenchymal progenitor cells can effectively treat nonhealing bone fractures in large animals, thereby addressing a major orthopedic unmet need and offering new possibilities for clinical translation.
Collapse
Affiliation(s)
- Maxim Bez
- Skeletal Biotech Laboratory, Hadassah Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem 91120, Israel.,Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Dmitriy Sheyn
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Wafa Tawackoli
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Pablo Avalos
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Galina Shapiro
- Skeletal Biotech Laboratory, Hadassah Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem 91120, Israel
| | - Joseph C Giaconi
- Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Xiaoyu Da
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Shiran Ben David
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jayne Gavrity
- Department of Biomedical Engineering and the Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hani A Awad
- Department of Biomedical Engineering and the Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hyun W Bae
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Eric J Ley
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Thomas J Kremen
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zulma Gazit
- Skeletal Biotech Laboratory, Hadassah Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem 91120, Israel.,Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Katherine W Ferrara
- Department of Biomedical Engineering, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Gadi Pelled
- Skeletal Biotech Laboratory, Hadassah Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem 91120, Israel.,Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dan Gazit
- Skeletal Biotech Laboratory, Hadassah Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem 91120, Israel. .,Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
16
|
Injected Human Muscle Precursor Cells Overexpressing PGC-1 α Enhance Functional Muscle Regeneration after Trauma. Stem Cells Int 2018. [PMID: 29531537 PMCID: PMC5827889 DOI: 10.1155/2018/4658503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
While many groups demonstrated new muscle tissue formation after muscle precursor cell (MPC) injection, the capacity of these cells to heal muscle damage, for example, sphincter in stress urinary incontinence, in long-term is still limited. Therefore, the first goal of our project was to optimize the functional regenerative potential of hMPC by genetic modification to overexpress human peroxisome proliferator-activated receptor gamma coactivator 1-alpha (hPGC-1α), key regulator of exercise-mediated adaptation. Moreover, we aimed at establishing a feasible methodology for noninvasive PET visualization of implanted cells and their microenvironment in muscle crush injury model. PGC-1α-bioengineered muscles showed enhanced marker expression for myogenesis (α-actinin, MyHC, and Desmin), vascularization (VEGF), neuronal (ACHE), and mitochondrial (COXIV) activity. Consistently, use of hPGC-1α_hMPCs produced significantly increased contractile force one to three weeks postinjury. PET imaging showed distinct differences in radiotracer signals ([18F]Fallypride and [11C]Raclopride (both targeting dopamine 2 receptors (D2R)) and [64Cu]NODAGA-RGD (targeting neovascularization)) between GFP_hMPCs and hD2R_hPGC-1α_hMPCs. After muscle harvesting, inflammation levels were in parallel to radiotracer uptake amount, with significantly lower uptake in hPGC-1α overexpressing samples. In summary, we facilitated early functional muscle tissue regeneration, introducing a novel approach to improve skeletal muscle regeneration. Besides successful tracking of hMPCs in muscle crush injuries, we showed that in high-inflammation areas, the specificity of radioligands might be significantly reduced, addressing a possible bottleneck of neovascularization PET imaging.
Collapse
|
17
|
Preparation and characterization of a novel silicon-modified nanobubble. PLoS One 2017; 12:e0178031. [PMID: 28557995 PMCID: PMC5448765 DOI: 10.1371/journal.pone.0178031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 05/08/2017] [Indexed: 01/05/2023] Open
Abstract
Nanobubbles (NBs) opened a new field of ultrasound imaging. There is still no practical method to control the diameter of bubbles. In this study, we developed a new method to control the size by incorporating of silicon hybrid lipids into the bubble membrane. The range of particle size of resulting NBs is between 523.02 ± 46.45 to 857.18 ± 82.90, smaller than the conventional microbubbles. The size of resulting NBs increased with the decrease in amount of silicon hybrid lipids, indicating the diameter of NBs can be regulated through modulating the ratio of silicon hybrid lipids in the bubble shell. Typical harmonic signals could be detected. The in vitro and in vivo ultrasound imaging experiments demonstrated these silicon-modified NBs had significantly improved ultrasound contrast enhancement abilities. Cytotoxicity assays revealed that these NBs had no obvious cytotoxicity to the 293 cell line at the tested bubble concentration. Our results showed that the novel NBs could use as nanoscale ultrasound contrast agents, providing the foundation for NBs in future applications including contrast-enhanced imaging and drug/gene delivery.
Collapse
|
18
|
Güvener N, Appold L, de Lorenzi F, Golombek SK, Rizzo LY, Lammers T, Kiessling F. Recent advances in ultrasound-based diagnosis and therapy with micro- and nanometer-sized formulations. Methods 2017; 130:4-13. [PMID: 28552267 DOI: 10.1016/j.ymeth.2017.05.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/11/2017] [Accepted: 05/21/2017] [Indexed: 01/15/2023] Open
Abstract
Ultrasound (US) is one of the most frequently used imaging methods in the clinic. The broad spectrum of its applications can be increased by the use of gas-filled microbubbles (MB) as ultrasound contrast agents (UCA). In recent years, also nanoscale UCA like nanobubbles (NB), echogenic liposomes (ELIP) and nanodroplets have been developed, which in contrast to MB, are able to extravasate from the vessels into the tissue. New disease-specific UCA have been designed for the assessment of tissue biomarkers and advanced US to a molecular imaging modality. For this purpose, specific binding moieties were coupled to the UCA surface. The vascular endothelial growth factor receptor-2 (VEGFR-2) and P-/E-selectin are prominent examples of molecular US targets to visualize tumor blood vessels and inflammatory diseases, respectively. Besides their application in contrast-enhanced imaging, MB can also be employed for drug delivery to tumors and across the blood-brain barrier (BBB). This review summarizes the development of micro- and nanoscaled UCA and highlights recent advances in diagnostic and therapeutic applications, which are ready for translation into the clinic.
Collapse
Affiliation(s)
- Nihan Güvener
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 20, 52074 Aachen, Germany
| | - Lia Appold
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 20, 52074 Aachen, Germany
| | - Federica de Lorenzi
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 20, 52074 Aachen, Germany
| | - Susanne K Golombek
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 20, 52074 Aachen, Germany
| | - Larissa Y Rizzo
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 20, 52074 Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 20, 52074 Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 20, 52074 Aachen, Germany.
| |
Collapse
|
19
|
Abstract
In recent decades ultrasound-guided delivery of drugs loaded on nanocarriers has been the focus of increasing attention to improve therapeutic treatments. Ultrasound has often been used in combination with microbubbles, micron-sized spherical gas-filled structures stabilized by a shell, to amplify the biophysical effects of the ultrasonic field. Nanometer size bubbles are defined nanobubbles. They were designed to obtain more efficient drug delivery systems. Indeed, their small sizes allow extravasation from blood vessels into surrounding tissues and ultrasound-targeted site-specific release with minimal invasiveness. Additionally, nanobubbles might be endowed with improved stability and longer residence time in systemic circulation. This review will describe the physico-chemical properties of nanobubbles, the formulation parameters and the drug loading approaches, besides potential applications as a therapeutic tool.
Collapse
|
20
|
Shapiro G, Wong AW, Bez M, Yang F, Tam S, Even L, Sheyn D, Ben-David S, Tawackoli W, Pelled G, Ferrara KW, Gazit D. Multiparameter evaluation of in vivo gene delivery using ultrasound-guided, microbubble-enhanced sonoporation. J Control Release 2015; 223:157-164. [PMID: 26682505 DOI: 10.1016/j.jconrel.2015.12.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 10/27/2015] [Accepted: 12/01/2015] [Indexed: 10/22/2022]
Abstract
More than 1800 gene therapy clinical trials worldwide have targeted a wide range of conditions including cancer, cardiovascular diseases, and monogenic diseases. Biological (i.e. viral), chemical, and physical approaches have been developed to deliver nucleic acids into cells. Although viral vectors offer the greatest efficiency, they also raise major safety concerns including carcinogenesis and immunogenicity. The goal of microbubble-mediated sonoporation is to enhance the uptake of drugs and nucleic acids. Insonation of microbubbles is thought to facilitate two mechanisms for enhanced uptake: first, deflection of the cell membrane inducing endocytotic uptake, and second, microbubble jetting inducing the formation of pores in the cell membrane. We hypothesized that ultrasound could be used to guide local microbubble-enhanced sonoporation of plasmid DNA. With the aim of optimizing delivery efficiency, we used nonlinear ultrasound and bioluminescence imaging to optimize the acoustic pressure, microbubble concentration, treatment duration, DNA dosage, and number of treatments required for in vivo Luciferase gene expression in a mouse thigh muscle model. We found that mice injected with 50μg luciferase plasmid DNA and 5×10(5) microbubbles followed by ultrasound treatment at 1.4MHz, 200kPa, 100-cycle pulse length, and 540 Hz pulse repetition frequency (PRF) for 2min exhibited superior transgene expression compared to all other treatment groups. The bioluminescent signal measured for these mice on Day 4 post-treatment was 100-fold higher (p<0.0001, n=5 or 6) than the signals for controls treated with DNA injection alone, DNA and microbubble injection, or DNA injection and ultrasound treatment. Our results indicate that these conditions result in efficient gene delivery and prolonged gene expression (up to 21days) with no evidence of tissue damage or off-target delivery. We believe that these promising results bear great promise for the development of microbubble-enhanced sonoporation-induced gene therapies.
Collapse
Affiliation(s)
- Galina Shapiro
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Ein Kerem, Jerusalem 91120, Israel
| | - Andrew W Wong
- University of California, Davis, Department of Biomedical Engineering, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Maxim Bez
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Ein Kerem, Jerusalem 91120, Israel
| | - Fang Yang
- University of California, Davis, Department of Biomedical Engineering, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Sarah Tam
- University of California, Davis, Department of Biomedical Engineering, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Lisa Even
- University of California, Davis, Department of Biomedical Engineering, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Dmitriy Sheyn
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Shiran Ben-David
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Wafa Tawackoli
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gadi Pelled
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Ein Kerem, Jerusalem 91120, Israel; Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Katherine W Ferrara
- University of California, Davis, Department of Biomedical Engineering, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Dan Gazit
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Ein Kerem, Jerusalem 91120, Israel; Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
21
|
Sanches PG, Mühlmeister M, Seip R, Kaijzel E, Löwik C, Böhmer M, Tiemann K, Grüll H. Ultrasound-mediated gene delivery of naked plasmid DNA in skeletal muscles: A case for bolus injections. J Control Release 2014; 195:130-7. [DOI: 10.1016/j.jconrel.2014.06.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 06/06/2014] [Accepted: 06/20/2014] [Indexed: 12/17/2022]
|
22
|
Fabiilli ML, Piert MR, Koeppe RA, Sherman PS, Quesada CA, Kripfgans OD. Assessment of the biodistribution of an [(18) F]FDG-loaded perfluorocarbon double emulsion using dynamic micro-PET in rats. CONTRAST MEDIA & MOLECULAR IMAGING 2013; 8:366-74. [PMID: 23613440 DOI: 10.1002/cmmi.1532] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 12/17/2012] [Accepted: 01/04/2013] [Indexed: 12/11/2022]
Abstract
Perfluorocarbon (PFC) double emulsions loaded with a water-soluble, therapeutic agent can be triggered by ultrasound in a process known as acoustic droplet vaporization. Elucidating the stability and biodistribution of these sonosensitive vehicles and encapsulated agents is critical in developing targeted drug delivery strategies using ultrasound. [(18) F]fluorodeoxyglucose (FDG) was encapsulated in a PFC double emulsion and the in vitro diffusion of FDG was assessed using a Franz diffusion cell. Using dynamic micro-positron emission tomography and direct tissue sampling, the biodistribution of FDG administered as a solution (i.e. non-emulsified) or as an emulsion was studied in Fisher 344 rats (n = 6) bearing subcutaneous 9L gliosarcoma. Standardized uptake values (SUVs) and area under the curve of the SUV (AUCSUV ) of FDG were calculated for various tissues. The FDG flux from the emulsion decreased by up to a factor of 6.9 compared with the FDG solution. FDG uptake, calculated from the AUCSUV , decreased by 36% and 44% for brain and tumor, respectively, when comparing FDG solution vs FDG emulsion (p < 0.01). Decreases in AUCSUV in highly metabolic tissues such as brain and tumor demonstrated retention of FDG within the double emulsion. No statistically significant differences in lung AUCSUV were observed, suggesting minimal accumulation of the emulsion in the pulmonary capillary bed. The liver AUCSUV increased by 356% for the FDG emulsion, thus indicating significant hepatic retention of the emulsion.
Collapse
Affiliation(s)
- Mario L Fabiilli
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Horie S, Chen R, Li L, Mori S, Kodama T. Contrast-enhanced high-frequency ultrasound imaging of early stage liver metastasis in a preclinical mouse model. Cancer Lett 2013; 339:208-13. [PMID: 23791880 DOI: 10.1016/j.canlet.2013.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 05/28/2013] [Accepted: 06/02/2013] [Indexed: 12/21/2022]
Abstract
Monitoring angiogenesis is potentially an effective strategy for the early detection of cancer. In this study, early detection was achieved by evaluating blood vessel density in the liver using a three-dimensional contrast-enhanced high-frequency ultrasound (CE-HFUS) system and Sonazoid microbubbles. Three-dimensional CE-HFUS detected an increase in blood vessel density in the liver after intrasplenic injection of breast tumor cells into mice. The results were in agreement with immunohistochemical analysis of blood vessel density. Three-dimensional CE-HFUS using microbubbles is an attractive, novel approach for the early detection of liver metastases through quantification of new, pathological vascular growth (i.e. tumor angiogenesis).
Collapse
Affiliation(s)
- Sachiko Horie
- Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo-machi, Aoba, Sendai, Miyagi 980-8575, Japan; Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba, Sendai, Miyagi 980-8575, Japan.
| | | | | | | | | |
Collapse
|
24
|
Optimization of Acoustic Liposomes for Improved In Vitro and In Vivo Stability. Pharm Res 2012; 30:218-24. [DOI: 10.1007/s11095-012-0864-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 08/13/2012] [Indexed: 10/27/2022]
|
25
|
Richard-Fiardo P, Franken PR, Lamit A, Marsault R, Guglielmi J, Cambien B, Graslin F, Lindenthal S, Darcourt J, Pourcher T, Vassaux G. Normalisation to blood activity is required for the accurate quantification of Na/I symporter ectopic expression by SPECT/CT in individual subjects. PLoS One 2012; 7:e34086. [PMID: 22470517 PMCID: PMC3309932 DOI: 10.1371/journal.pone.0034086] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 02/21/2012] [Indexed: 12/21/2022] Open
Abstract
The utilisation of the Na/I symporter (NIS) and associated radiotracers as a reporter system for imaging gene expression is now reaching the clinical setting in cancer gene therapy applications. However, a formal assessment of the methodology in terms of normalisation of the data still remains to be performed, particularly in the context of the assessment of activities in individual subjects in longitudinal studies. In this context, we administered to mice a recombinant, replication-incompetent adenovirus encoding rat NIS, or a human colorectal carcinoma cell line (HT29) encoding mouse NIS. We used (99m)Tc pertechnetate as a radiotracer for SPECT/CT imaging to determine the pattern of ectopic NIS expression in longitudinal kinetic studies. Some animals of the cohort were culled and NIS expression was measured by quantitative RT-PCR and immunohistochemistry. The radioactive content of some liver biopsies was also measured ex vivo. Our results show that in longitudinal studies involving datasets taken from individual mice, the presentation of non-normalised data (activity expressed as %ID/g or %ID/cc) leads to 'noisy', and sometimes incoherent, results. This variability is due to the fact that the blood pertechnetate concentration can vary up to three-fold from day to day. Normalisation of these data with blood activities corrects for these inconsistencies. We advocate that, blood pertechnetate activity should be determined and used to normalise the activity measured in the organ/region of interest that expresses NIS ectopically. Considering that NIS imaging has already reached the clinical setting in the context of cancer gene therapy, this normalisation may be essential in order to obtain accurate and predictive information in future longitudinal clinical studies in biotherapy.
Collapse
Affiliation(s)
- Peggy Richard-Fiardo
- INSERM U948, Biothérapies Hépatiques, CHU Hôtel Dieu, Nantes, France
- CHU de Nantes, Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - Philippe R. Franken
- Centre Antoine Lacassagne, Nice, France
- Faculté de Médecine, Université de Nice Sophia-Antipolis, Nice, France
- Laboratoire TIRO, UMRE 4320, iBEB, DSV, Commissariat à l'Energie Atomique, Nice, France
| | - Audrey Lamit
- Centre Antoine Lacassagne, Nice, France
- Faculté de Médecine, Université de Nice Sophia-Antipolis, Nice, France
- Laboratoire TIRO, UMRE 4320, iBEB, DSV, Commissariat à l'Energie Atomique, Nice, France
| | - Robert Marsault
- Centre Antoine Lacassagne, Nice, France
- Faculté de Médecine, Université de Nice Sophia-Antipolis, Nice, France
- Laboratoire TIRO, UMRE 4320, iBEB, DSV, Commissariat à l'Energie Atomique, Nice, France
| | - Julien Guglielmi
- Centre Antoine Lacassagne, Nice, France
- Faculté de Médecine, Université de Nice Sophia-Antipolis, Nice, France
- Laboratoire TIRO, UMRE 4320, iBEB, DSV, Commissariat à l'Energie Atomique, Nice, France
| | - Béatrice Cambien
- Centre Antoine Lacassagne, Nice, France
- Faculté de Médecine, Université de Nice Sophia-Antipolis, Nice, France
- Laboratoire TIRO, UMRE 4320, iBEB, DSV, Commissariat à l'Energie Atomique, Nice, France
| | - Fanny Graslin
- Centre Antoine Lacassagne, Nice, France
- Faculté de Médecine, Université de Nice Sophia-Antipolis, Nice, France
- Laboratoire TIRO, UMRE 4320, iBEB, DSV, Commissariat à l'Energie Atomique, Nice, France
| | - Sabine Lindenthal
- Centre Antoine Lacassagne, Nice, France
- Faculté de Médecine, Université de Nice Sophia-Antipolis, Nice, France
- Laboratoire TIRO, UMRE 4320, iBEB, DSV, Commissariat à l'Energie Atomique, Nice, France
| | - Jacques Darcourt
- Centre Antoine Lacassagne, Nice, France
- Faculté de Médecine, Université de Nice Sophia-Antipolis, Nice, France
- Laboratoire TIRO, UMRE 4320, iBEB, DSV, Commissariat à l'Energie Atomique, Nice, France
| | - Thierry Pourcher
- Centre Antoine Lacassagne, Nice, France
- Faculté de Médecine, Université de Nice Sophia-Antipolis, Nice, France
- Laboratoire TIRO, UMRE 4320, iBEB, DSV, Commissariat à l'Energie Atomique, Nice, France
| | - Georges Vassaux
- INSERM U948, Biothérapies Hépatiques, CHU Hôtel Dieu, Nantes, France
- CHU de Nantes, Institut des Maladies de l'Appareil Digestif, Nantes, France
- * E-mail:
| |
Collapse
|
26
|
Yin T, Wang P, Zheng R, Zheng B, Cheng D, Zhang X, Shuai X. Nanobubbles for enhanced ultrasound imaging of tumors. Int J Nanomedicine 2012; 7:895-904. [PMID: 22393289 PMCID: PMC3289446 DOI: 10.2147/ijn.s28830] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The fabrication and initial applications of nanobubbles (NBs) have shown promising results in recent years. A small particle size is a basic requirement for ultrasound contrast-enhanced agents that penetrate tumor blood vessel pores to allow for targeted imaging and therapy. However, the nanoscale size of the particles used has the disadvantage of weakening the imaging ability of clinical diagnostic ultrasound. In this work, we fabricated a lipid NBs contrast-enhanced ultrasound agent and evaluated its passive targeting ability in vivo. The results showed that the NBs were small (436.8 ± 5.7 nm), and in vitro ultrasound imaging suggested that the ultrasonic imaging ability is comparable to that of microbubbles (MBs). In vivo experiments confirmed the ability of NBs to passively target tumor tissues. The NBs remained in the tumor area for a longer period because they exhibited enhanced permeability and retention. Direct evidence was obtained by direct observation of red fluorescence-dyed NBs in tumor tissue using confocal laser scanning microscopy. We have demonstrated the ability to fabricate NBs that can be used for the in vivo contrast-enhanced imaging of tumor tissue and that have potential for drug/gene delivery.
Collapse
Affiliation(s)
- Tinghui Yin
- Department of Medical Ultrasonic, Third Affiliated Hospital, Guangzhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
27
|
Fernandez-Fernandez A, Manchanda R, McGoron AJ. Theranostic applications of nanomaterials in cancer: drug delivery, image-guided therapy, and multifunctional platforms. Appl Biochem Biotechnol 2011; 165:1628-51. [PMID: 21947761 PMCID: PMC3239222 DOI: 10.1007/s12010-011-9383-z] [Citation(s) in RCA: 235] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Accepted: 09/07/2011] [Indexed: 12/18/2022]
Abstract
Successful cancer management depends on accurate diagnostics along with specific treatment protocols. Current diagnostic techniques need to be improved to provide earlier detection capabilities, and traditional chemotherapy approaches to cancer treatment are limited by lack of specificity and systemic toxicity. This review highlights advances in nanotechnology that have allowed the development of multifunctional platforms for cancer detection, therapy, and monitoring. Nanomaterials can be used as MRI, optical imaging, and photoacoustic imaging contrast agents. When used as drug carriers, nanoformulations can increase tumor exposure to therapeutic agents and result in improved treatment effects by prolonging circulation times, protecting entrapped drugs from degradation, and enhancing tumor uptake through the enhanced permeability and retention effect as well as receptor-mediated endocytosis. Multiple therapeutic agents such as chemotherapy, antiangiogenic, or gene therapy agents can be simultaneously delivered by nanocarriers to tumor sites to enhance the effectiveness of therapy. Additionally, imaging and therapy agents can be co-delivered to provide seamless integration of diagnostics, therapy, and follow-up, and different therapeutic modalities such as chemotherapy and hyperthermia can be co-administered to take advantage of synergistic effects. Liposomes, metallic nanoparticles, polymeric nanoparticles, dendrimers, carbon nanotubes, and quantum dots are examples of nanoformulations that can be used as multifunctional platforms for cancer theranostics. Nanomedicine approaches in cancer have great potential for clinically translatable advances that can positively impact the overall diagnostic and therapeutic process and result in enhanced quality of life for cancer patients. However, a concerted scientific effort is still necessary to fully explore long-term risks, effects, and precautions for safe human use.
Collapse
Affiliation(s)
- Alicia Fernandez-Fernandez
- Department of Biomedical Engineering, Florida International University, 10555 West Flagler Street, Miami, FL 33174, USA
| | | | | |
Collapse
|
28
|
Horie S, Watanabe Y, Ono M, Mori S, Kodama T. Evaluation of antitumor effects following tumor necrosis factor-α gene delivery using nanobubbles and ultrasound. Cancer Sci 2011; 102:2082-9. [PMID: 21824220 PMCID: PMC11158812 DOI: 10.1111/j.1349-7006.2011.02056.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The antitumor effects of tumor necrosis factor (TNF-α) were evaluated following transfection of TNF-α plasmid DNA into solid mouse tumors using the nanobubbles (NBs) and ultrasound (US) gene delivery system. Murine breast carcinoma (EMT6) cells expressing luciferase (1 × 10(6) cells) were injected intradermally into the flanks of 6-7-week-old male SCID mice on day 0. Ten microliters of TNF-α (5 μg/μL) or TNF-α mock plasmid DNA (5 μg/μL) with/without NBs (15 μL) and saline was injected intratumorally in a total volume of 30 μL, and tumors were exposed to US (frequency, 1 MHz; intensity, 3.0 W/cm(2); duty cycle, 20%; number of pulses, 200; and exposure time, 60 s) on days 2, 4, 7, and 9. Changes in tumor size were measured with an in vivo bioluminescent imaging system and a mechanical caliper. Changes in tumor vessel area were quantified using contrast-enhanced US imaging with Sonazoid and a high frequency US imaging system (40 MHz) and immunohistochemistry (CD31). At the mRNA level, expression of TNF-α, caspase-3, and p53 were quantified using real-time quantitative RT-PCR. At the protein level, expression of caspase-3 and p53 were confirmed by immunohistochemistry. We show that repeated TNF-α gene delivery using NBs and US can lead to the local production of TNF-α. This results in antitumor effects, including activation of p53-dependent apoptosis, decrease in tumor vessel density, and suppression of tumor size. In this study, we showed the effectiveness of using NBs and US for TNF-α gene delivery into tumor cells.
Collapse
Affiliation(s)
- Sachiko Horie
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | | | | | | | | |
Collapse
|
29
|
Klutz K, Willhauck MJ, Dohmen C, Wunderlich N, Knoop K, Zach C, Senekowitsch-Schmidtke R, Gildehaus FJ, Ziegler S, Fürst S, Göke B, Wagner E, Ogris M, Spitzweg C. Image-guided tumor-selective radioiodine therapy of liver cancer after systemic nonviral delivery of the sodium iodide symporter gene. Hum Gene Ther 2011; 22:1563-74. [PMID: 21851208 DOI: 10.1089/hum.2011.041] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We reported the induction of tumor-selective iodide uptake and therapeutic efficacy of (131)I in a hepatocellular carcinoma (HCC) xenograft mouse model, using novel polyplexes based on linear polyethylenimine (LPEI), shielded by polyethylene glycol (PEG), and coupled with the epidermal growth factor receptor-specific peptide GE11 (LPEI-PEG-GE11). The aim of the current study in the same HCC model was to evaluate the potential of biodegradable nanoparticle vectors based on pseudodendritic oligoamines (G2-HD-OEI) for systemic sodium iodide symporter (NIS) gene delivery and to compare efficiency and tumor specificity with LPEI-PEG-GE11. Transfection of HCC cells with NIS cDNA, using G2-HD-OEI, resulted in a 44-fold increase in iodide uptake in vitro as compared with a 22-fold increase using LPEI-PEG-GE11. After intravenous application of G2-HD-OEI/NIS HCC tumors accumulated 6-11% ID/g (123)I (percentage of the injected dose per gram tumor tissue) with an effective half-life of 10 hr (tumor-absorbed dose, 281 mGy/MBq) as measured by (123)I scintigraphic gamma camera or single-photon emission computed tomography computed tomography (SPECT CT) imaging, as compared with 6.5-9% ID/g with an effective half-life of only 6 hr (tumor-absorbed dose, 47 mGy/MBq) for LPEI-PEG-GE11. After only two cycles of G2-HD-OEI/NIS/(131)I application, a significant delay in tumor growth was observed with markedly improved survival. A similar degree of therapeutic efficacy had been observed after four cycles of LPEI-PEG-GE11/(131)I. These results clearly demonstrate that biodegradable nanoparticles based on OEI-grafted oligoamines show increased efficiency for systemic NIS gene transfer in an HCC model with similar tumor selectivity as compared with LPEI-PEG-GE11, and therefore represent a promising strategy for NIS-mediated radioiodine therapy of HCC.
Collapse
Affiliation(s)
- Kathrin Klutz
- Department of Internal Medicine II, Ludwig-Maximilians-University, 81377 Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
|
31
|
Caskey CF, Hu X, Ferrara KW. Leveraging the power of ultrasound for therapeutic design and optimization. J Control Release 2011; 156:297-306. [PMID: 21835212 DOI: 10.1016/j.jconrel.2011.07.032] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 07/21/2011] [Indexed: 12/19/2022]
Abstract
Contrast agent-enhanced ultrasound can facilitate personalized therapeutic strategies by providing the technology to measure local blood flow rate, to selectively image receptors on the vascular endothelium, and to enhance localized drug delivery. Ultrasound contrast agents are micron-diameter encapsulated bubbles that circulate within the vascular compartment and can be selectively imaged with ultrasound. Microbubble transport-based estimates of local blood flow can quantify changes resulting from anti-angiogenic therapies and facilitate differentiation of angiogenic mechanisms. Microbubbles that are conjugated with targeting ligands attach to endothelial surface receptors that are upregulated in disease, providing high signal-to-noise ratio images of pathological vasculature. In addition to imaging applications, microbubbles can be used to enhance localized gene and drug delivery, either by changing membrane and vascular permeability or by carrying and locally releasing cargo. Our goal in this review is to provide an overview of the use of contrast-enhanced ultrasound methodologies in the design and evaluation of therapeutic strategies with emphases on quantitative blood flow mapping, molecular imaging, and enhanced drug delivery.
Collapse
Affiliation(s)
- Charles F Caskey
- Department of Biomedical Engineering, University of California, Davis, One Shields Ave, Davis, CA 95616, USA
| | | | | |
Collapse
|
32
|
Richard-Fiardo P, Franken PR, Harrington KJ, Vassaux G, Cambien B. The use of molecular imaging of gene expression by radiotracers in gene therapy. Expert Opin Biol Ther 2011; 11:1273-85. [DOI: 10.1517/14712598.2011.588596] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
33
|
Epidermal growth factor receptor-targeted (131)I-therapy of liver cancer following systemic delivery of the sodium iodide symporter gene. Mol Ther 2011; 19:676-85. [PMID: 21245850 DOI: 10.1038/mt.2010.296] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
We recently demonstrated tumor-selective iodide uptake and therapeutic efficacy of radioiodine in neuroblastoma tumors after systemic nonviral polyplex-mediated sodium iodide symporter (NIS) gene delivery. In the present study, we used novel polyplexes based on linear polyethylenimine (LPEI), polyethylene glycol (PEG), and the synthetic peptide GE11 as an epidermal growth factor receptor (EGFR)-specific ligand to target a NIS-expressing plasmid to hepatocellular carcinoma (HCC) (HuH7). Incubation of HuH7 cells with LPEI-PEG-GE11/NIS polyplexes resulted in a 22-fold increase in iodide uptake, which was confirmed in other cancer cell lines correlating well with EGFR expression levels. Using (123)I-scintigraphy and ex vivo γ-counting, HuH7 xenografts accumulated 6.5-9% injected dose per gram (ID/g) (123)I, resulting in a tumor-absorbed dose of 47 mGray/Megabecquerel (mGy/MBq) (131)Iodide ((131)I) after intravenous (i.v.) application of LPEI-PEG-GE11/NIS. No iodide uptake was observed in other tissues. After pretreatment with the EGFR-specific antibody cetuximab, tumoral iodide uptake was markedly reduced confirming the specificity of EGFR-targeted polyplexes. After three or four cycles of polyplex/(131)I application, a significant delay in tumor growth was observed associated with prolonged survival. These results demonstrate that systemic NIS gene transfer using polyplexes coupled with an EGFR-targeting ligand is capable of inducing tumor-specific iodide uptake, which represents a promising innovative strategy for systemic NIS gene therapy in metastatic cancers.
Collapse
|