1
|
Izadiyan Z, Misran M, Kalantari K, Webster TJ, Kia P, Basrowi NA, Rasouli E, Shameli K. Advancements in Liposomal Nanomedicines: Innovative Formulations, Therapeutic Applications, and Future Directions in Precision Medicine. Int J Nanomedicine 2025; 20:1213-1262. [PMID: 39911259 PMCID: PMC11794392 DOI: 10.2147/ijn.s488961] [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: 07/27/2024] [Accepted: 01/01/2025] [Indexed: 02/07/2025] Open
Abstract
Liposomal nanomedicines have emerged as a pivotal approach for the treatment of various diseases, notably cancer and infectious diseases. This manuscript provides an in-depth review of recent advancements in liposomal formulations, highlighting their composition, targeted delivery strategies, and mechanisms of action. We explore the evolution of liposomal products currently in clinical trials, emphasizing their potential in addressing diverse medical challenges. The integration of immunotherapeutic agents within liposomes marks a paradigm shift, enabling the design of 'immuno-modulatory hubs' capable of orchestrating precise immune responses while facilitating theranostic applications. The recent COVID-19 pandemic has accelerated research in liposomal-based vaccines and antiviral therapies, underscoring the need for improved delivery mechanisms to overcome challenges like rapid clearance and organ toxicity. Furthermore, we discuss the potential of "smart" liposomes, which can respond to specific disease microenvironments, enhancing treatment efficacy and precision. The integration of artificial intelligence and machine learning in optimizing liposomal designs promises to revolutionize personalized medicine, paving the way for innovative strategies in disease detection and therapeutic interventions. This comprehensive review underscores the significance of ongoing research in liposomal technologies, with implications for future clinical applications and enhanced patient outcomes.
Collapse
Affiliation(s)
- Zahra Izadiyan
- Department of Chemistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Misni Misran
- Department of Chemistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Katayoon Kalantari
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Thomas J Webster
- Biomedical Engineering, Hebei University of Technology, Tianjin, People’s Republic of China
- School of Engineering, Saveetha University, Chennai, India
| | - Pooneh Kia
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | | | - Elisa Rasouli
- Department of Electrical and Electronics Engineering, Nanyang Technological University, Nanyang, Singapore
| | - Kamyar Shameli
- School of Medicine, Institute of Virology, Technical University of Munich, Munich, Germany
| |
Collapse
|
2
|
Chen CM, Huang CY, Lai CH, Chen YC, Hwang YT, Lin CY. Neuroprotection effects of kynurenic acid-loaded micelles for the Parkinson's disease models. J Liposome Res 2024; 34:593-604. [PMID: 38779944 DOI: 10.1080/08982104.2024.2346986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024]
Abstract
Anti-glutamatergic agents may have neuroprotective effects against excitotoxicity that is known to be involved in the pathogenesis of Parkinson's disease (PD). One of these agents is kynurenic acid (KYNA), a tryptophan metabolite, which is an endogenous N-methyl-D-aspartic acid (NMDA) receptor antagonist. However, its pharmacological properties of poor water solubility and limited blood-brain barrier (BBB) permeability rules out its systemic administration in disorders affecting the central nervous system. Our aim in the present study was to investigate the neuroprotective effects of KYNA-loaded micelles (KYNA-MICs) against PD in vitro and in vivo. Lipid-based micelles (MICs) in conjunction with KYNA drug delivery have the potential to enhance the penetration of therapeutic drugs into a diseased brain without BBB obstacles. KYNA-MICs were characterized by particle size (105.8 ± 12.1 nm), loading efficiency (78.3 ± 4.23%), and in vitro drug release (approximately 30% at 24 h). The in vitro experiments showed that KYNA-MICs effectively reduced 2-fold protein aggregation. The in vivo studies revealed that KYNA was successfully delivered by 5-fold increase in neurotoxin-induced PD brains. The results showed significant enhancement of KYNA delivery into brain. We also found that the KYNA-MICs exhibited several therapeutic effects. The KYNA-MICs reduced protein aggregation of an in vitro PD model, ameliorated motor functions, and prevented loss of the striatal neurons in a PD animal model. The beneficial effects of KYNA-MICs are probably explained by the anti-excitotoxic activity of the treatment's complex. As the KYNA-MICs did not induce any appreciable side-effects at the protective dose applied to a chronic PD mouse model, our results demonstrate that KYNA provides neuroprotection and attenuates PD pathology.
Collapse
Affiliation(s)
- Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ching-Yun Huang
- Research Center for Radiation Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chin-Hui Lai
- Department of Neurology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Chieh Chen
- Department of Neurology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Ting Hwang
- Department of Statistics, National Taipei University, Taipei, Taiwan
| | - Chung-Yin Lin
- Department of Neurology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Radiation Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Statistics, National Taipei University, Taipei, Taiwan
| |
Collapse
|
3
|
Al Refaai KA, AlSawaftah NA, Abuwatfa W, Husseini GA. Drug Release via Ultrasound-Activated Nanocarriers for Cancer Treatment: A Review. Pharmaceutics 2024; 16:1383. [PMID: 39598507 PMCID: PMC11597164 DOI: 10.3390/pharmaceutics16111383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Conventional cancer chemotherapy often struggles with safely and effectively delivering anticancer therapeutics to target tissues, frequently leading to dose-limiting toxicity and suboptimal therapeutic outcomes. This has created a need for novel therapies that offer greater efficacy, enhanced safety, and improved toxicological profiles. Nanocarriers are nanosized particles specifically designed to enhance the selectivity and effectiveness of chemotherapy drugs while reducing their toxicity. A subset of drug delivery systems utilizes stimuli-responsive nanocarriers, which enable on-demand drug release, prevent premature release, and offer spatial and temporal control over drug delivery. These stimuli can be internal (such as pH and enzymes) or external (such as ultrasound, magnetic fields, and light). This review focuses on the mechanics of ultrasound-induced drug delivery and the various nanocarriers used in conjunction with ultrasound. It will also provide a comprehensive overview of key aspects related to ultrasound-induced drug delivery, including ultrasound parameters and the biological effects of ultrasound waves.
Collapse
Affiliation(s)
- Khaled Armouch Al Refaai
- Biomedical Engineering Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
| | - Nour A. AlSawaftah
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (N.A.A.); (W.A.)
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Biosciences and Bioengineering Ph.D. Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Waad Abuwatfa
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (N.A.A.); (W.A.)
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Biosciences and Bioengineering Ph.D. Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Biomedical Engineering Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (N.A.A.); (W.A.)
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Biosciences and Bioengineering Ph.D. Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| |
Collapse
|
4
|
Zhang J, Sun L, Jiang L, Xie X, Wang Y, Wu R, Tang Q, Sun S, Zhu S, Liang X, Cui L. Regulation of CTLs/Tregs via Highly Stable and Ultrasound-Responsive Cerasomal Nano-Modulators for Enhanced Colorectal Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400485. [PMID: 38552151 PMCID: PMC11165532 DOI: 10.1002/advs.202400485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/27/2024] [Indexed: 06/12/2024]
Abstract
Immunotherapy is showing good potential for colorectal cancer therapy, however, low responsive rates and severe immune-related drug side effects still hamper its therapeutic effectiveness. Herein, a highly stable cerasomal nano-modulator (DMC@P-Cs) with ultrasound (US)-controlled drug delivery capability for selective sonodynamic-immunotherapy is fabricated. DMC@P-Cs' lipid bilayer is self-assembled from cerasome-forming lipid (CFL), pyrophaeophorbid conjugated lipid (PL), and phospholipids containing unsaturated chemical bonds (DOPC), resulting in US-responsive lipid shell. Demethylcantharidin (DMC) as an immunotherapy adjuvant is loaded in the hydrophilic core of DMC@P-Cs. With US irradiation, reactive oxygen species (ROS) can be effectively generated from DMC@P-Cs, which can not only kill tumor cells for inducing immunogenic cell death (ICD), but also oxidize unsaturated phospholipids-DOPC to change the permeability of the lipid bilayers and facilitate controlled release of DMC, thus resulting in down-regulation of regulatory T cells (Tregs) and amplification of anti-tumor immune responses. After intravenous injection, DMC@P-Cs can efficiently accumulate at the tumor site, and local US treatment resulted in 94.73% tumor inhibition rate. In addition, there is no detectable systemic toxicity. Therefore, this study provides a highly stable and US-controllable smart delivery system to achieve synergistical sonodynamic-immunotherapy for enhanced colorectal cancer therapy.
Collapse
Affiliation(s)
- Jinxia Zhang
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100010P. R. China
- Department of UltrasoundPeking University Third HospitalBeijing100191P. R. China
| | - Lihong Sun
- Department of UltrasoundPeking University Third HospitalBeijing100191P. R. China
| | - Ling Jiang
- Department of UltrasoundPeking University Third HospitalBeijing100191P. R. China
| | - Xinxin Xie
- Department of UltrasoundPeking University Third HospitalBeijing100191P. R. China
| | - Yuan Wang
- Department of UltrasoundPeking University Third HospitalBeijing100191P. R. China
| | - Ruiqi Wu
- Department of UltrasoundPeking University Third HospitalBeijing100191P. R. China
| | - Qingshuang Tang
- Department of UltrasoundPeking University Third HospitalBeijing100191P. R. China
| | - Suhui Sun
- Department of UltrasoundPeking University Third HospitalBeijing100191P. R. China
| | - Shiwei Zhu
- Department of UltrasoundPeking University Third HospitalBeijing100191P. R. China
| | - Xiaolong Liang
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100010P. R. China
- Department of UltrasoundPeking University Third HospitalBeijing100191P. R. China
| | - Ligang Cui
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100010P. R. China
- Department of UltrasoundPeking University Third HospitalBeijing100191P. R. China
| |
Collapse
|
5
|
Zafar MN, Pitt WG, Husseini GA. Encapsulation and release of calcein from herceptin-conjugated eLiposomes. Heliyon 2024; 10:e27882. [PMID: 38524567 PMCID: PMC10958368 DOI: 10.1016/j.heliyon.2024.e27882] [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: 12/10/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/26/2024] Open
Abstract
Achieving an optimal therapeutic level is crucial in effectively eradicating cancer cells during treatment. However, conventional chemotherapy-associated systemic administration of anticancer agents leads to many side effects. To achieve the desired control over the target site, active targeting of HER2-positive breast cancer cells can be achieved by conjugating liposomal vesicles with Human Epidermal growth factor Receptor 2 (HER2) and inducing release of the encapsulated drug using ultrasound. To further enhance the delivery efficiency, nanoemulsion droplets exhibiting responsiveness to low-frequency ultrasound are encapsulated within these lipid vesicles. In this study, we prepared four different liposomal formulations, namely pegylated liposomes, emulsion liposomes (eLiposomes), HER-conjugated liposomes, and HER-conjugated eLiposomes, each loaded with calcein and subjected to a thorough characterization process. Their sizes, phospholipid concentration, and amount of antibody conjugation were compared and analyzed. Cryogenic transmission electron microscopy was used to confirm the encapsulation of nanoemulsion droplets within the liposomes. The drug-releasing performance of Herceptin-conjugated eLiposomes was found to surpass that of other liposomal formulations with a notably higher calcein release and established it as a highly effective nanocarrier. The study showcases the efficacy of calcein-loaded and Herceptin-conjugated eLiposomes, which demonstrate rapid and efficient drug release among other liposomal formulations when subjected to ultrasound. This discovery paves the way for a more targeted, efficient, and humane approach to cancer therapy.
Collapse
Affiliation(s)
- Mah Noor Zafar
- Biomedical Engineering Program, College of Engineering, American University of Sharjah, Sharjah, P.O. Box. 26666, United Arab Emirates
| | - William G. Pitt
- Department of Chemical Engineering, Brigham Young University, Provo, UT, 84602, USA
| | - Ghaleb A. Husseini
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah, P.O. Box. 26666, United Arab Emirates
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah, P.O. Box 26666, United Arab Emirates
| |
Collapse
|
6
|
El-Tanani M, Nsairat H, Aljabali AA, Matalka II, Alkilany AM, Tambuwala MM. Dual-loaded liposomal carriers to combat chemotherapeutic resistance in breast cancer. Expert Opin Drug Deliv 2024; 21:309-324. [PMID: 38284386 DOI: 10.1080/17425247.2024.2311812] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/25/2024] [Indexed: 01/30/2024]
Abstract
INTRODUCTION The resistance to chemotherapy is a significant hurdle in breast cancer treatment, prompting the exploration of innovative strategies. This review discusses the potential of dual-loaded liposomal carriers to combat chemoresistance and improve outcomes for breast cancer patients. AREAS COVERED This review discusses breast cancer chemotherapy resistance and dual-loaded liposomal carriers. Drug efflux pumps, DNA repair pathways, and signaling alterations are discussed as chemoresistance mechanisms. Liposomes can encapsulate several medicines and cargo kinds, according to the review. It examines how these carriers improve medication delivery, cancer cell targeting, and tumor microenvironment regulation. Also examined are dual-loaded liposomal carrier improvement challenges and techniques. EXPERT OPINION The use of dual-loaded liposomal carriers represents a promising and innovative strategy in the battle against chemotherapy resistance in breast cancer. This article has explored the various mechanisms of chemoresistance in breast cancer, emphasizing the potential of dual-loaded liposomal carriers to overcome these challenges. These carriers offer versatility, enabling the encapsulation and precise targeting of multiple drugs with different modes of action, a crucial advantage when dealing with the complexity of breast cancer treatment.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Pharmacological and Diagnostic Research Center, Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Pharmacy, Yarmouk University, Irbid, Jordan
| | - Ismail I Matalka
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Department of Pathology and Microbiology, Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | | | | |
Collapse
|
7
|
Koroleva M. Multicompartment colloid systems with lipid and polymer membranes for biomedical applications. Phys Chem Chem Phys 2023; 25:21836-21859. [PMID: 37565484 DOI: 10.1039/d3cp01984e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Multicompartment structures have the potential for biomedical applications because they can act as multifunctional systems and provide simultaneous delivery of drugs and diagnostics agents of different types. Moreover, some of them mimic biological cells to some extent with organelles as separate sub-compartments. This article analyses multicompartment colloidal structures with smaller sub-units covered with lipid or polymer membranes that provide additional protection for the encapsulated substances. Vesosomes with small vesicles encapsulated in the inner pools of larger liposomes are the most studied systems to date. Dendrimer molecules are enclosed by a lipid bilayer shell in dendrosomes. Capsosomes, polymersomes-in-polymer capsules, and cubosomes-in-polymer capsules are composed of sub-compartments encapsulated within closed multilayer polymer membranes. Janus or Cerberus emulsions contain droplets composed of two or three phases: immiscible oils in O/W emulsions and aqueous polymer or salt solutions that are separated into two or three phases and form connected droplets in W/O emulsions. In more cases, the external surface of engulfed droplets in Janus or Cerberus emulsions is covered with a lipid or polymer monolayer. eLiposomes with emulsion droplets encapsulated into a bilayer shell have been given little attention so far, but they have very great prospects. In addition to nanoemulsion droplets, solid lipid nanoparticles, nanostructured lipid carriers and inorganic nanoparticles can be loaded into eLiposomes. Molecular engineering of the external membrane allows the creation of ligand-targeted and stimuli-responsive multifunctional systems. As a result, the efficacy of drug delivery can be significantly enhanced.
Collapse
Affiliation(s)
- Marina Koroleva
- Mendeleev University of Chemical Technology, Miusskaya sq. 9, Moscow 125047.
| |
Collapse
|
8
|
Edwards IA, De Carlo F, Sitta J, Varner W, Howard CM, Claudio PP. Enhancing Targeted Therapy in Breast Cancer by Ultrasound-Responsive Nanocarriers. Int J Mol Sci 2023; 24:ijms24065474. [PMID: 36982548 PMCID: PMC10053544 DOI: 10.3390/ijms24065474] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023] Open
Abstract
Currently, the response to cancer treatments is highly variable, and severe side effects and toxicity are experienced by patients receiving high doses of chemotherapy, such as those diagnosed with triple-negative breast cancer. The main goal of researchers and clinicians is to develop new effective treatments that will be able to specifically target and kill tumor cells by employing the minimum doses of drugs exerting a therapeutic effect. Despite the development of new formulations that overall can increase the drugs’ pharmacokinetics, and that are specifically designed to bind overexpressed molecules on cancer cells and achieve active targeting of the tumor, the desired clinical outcome has not been reached yet. In this review, we will discuss the current classification and standard of care for breast cancer, the application of nanomedicine, and ultrasound-responsive biocompatible carriers (micro/nanobubbles, liposomes, micelles, polymeric nanoparticles, and nanodroplets/nanoemulsions) employed in preclinical studies to target and enhance the delivery of drugs and genes to breast cancer.
Collapse
Affiliation(s)
- Isaiah A. Edwards
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Flavia De Carlo
- Department of Pharmacology and Toxicology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Juliana Sitta
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - William Varner
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Candace M. Howard
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Pier Paolo Claudio
- Department of Pharmacology and Toxicology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Correspondence:
| |
Collapse
|
9
|
Zafar MN, Abuwatfa WH, Husseini GA. Acoustically-Activated Liposomal Nanocarriers to Mitigate the Side Effects of Conventional Chemotherapy with a Focus on Emulsion-Liposomes. Pharmaceutics 2023; 15:421. [PMID: 36839744 PMCID: PMC9963571 DOI: 10.3390/pharmaceutics15020421] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/31/2023] Open
Abstract
To improve currently available cancer treatments, nanomaterials are employed as smart drug delivery vehicles that can be engineered to locally target cancer cells and respond to stimuli. Nanocarriers can entrap chemotherapeutic drugs and deliver them to the diseased site, reducing the side effects associated with the systemic administration of conventional anticancer drugs. Upon accumulation in the tumor cells, the nanocarriers need to be potentiated to release their therapeutic cargo. Stimulation can be through endogenous or exogenous modalities, such as temperature, electromagnetic irradiation, ultrasound (US), pH, or enzymes. This review discusses the acoustic stimulation of different sonosensitive liposomal formulations. Emulsion liposomes, or eLiposomes, are liposomes encapsulating phase-changing nanoemulsion droplets, which promote acoustic droplet vaporization (ADV) upon sonication. This gives eLiposomes the advantage of delivering the encapsulated drug at low intensities and short exposure times relative to liposomes. Other formulations integrating microbubbles and nanobubbles are also discussed.
Collapse
Affiliation(s)
- Mah Noor Zafar
- Biomedical Engineering Program, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Waad H. Abuwatfa
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| |
Collapse
|
10
|
NIR-Mediated drug release and tumor theranostics using melanin-loaded liposomes. Biomater Res 2022; 26:22. [PMID: 35659113 PMCID: PMC9164422 DOI: 10.1186/s40824-022-00270-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Heat generation in a drug delivery carrier by exposure to near-infrared (NIR) light with excellent tissue transmittance is an effective strategy for drug release and tumor therapy. Because liposomes have amphiphilic properties, they are useful as drug carriers. Liposomes are also very suitable for drug delivery strategies using heat generation by NIR laser because lipid bilayers are easily broken by heat. Thermally generated bubbles from liposomes not only induce drug release, but also enable ultrasound imaging. METHODS Melanin, perfluorohexane (PFH), and 5-fluorouracil (5-FU)-loaded liposomes (melanin@PFH@5-FU-liposomes) that can generate heat and bubble by NIR laser irradiation were prepared by a thin film method. Conversion of light to heat and bubble generation of melanin@PFH@5-FU-liposomes were evaluated using an infrared (IR) thermal imaging camera and an ultrasound imaging system both in vitro and in vivo. To investigate tumor therapeutic effect, NIR laser of 808 nm was used to irradiate tumor site for 10 min after injecting melanin@PFH@5-FU-liposome into tail veins of CT26-bearing mice. RESULTS Melanin@PFH@5-FU-liposomes showed a spherical shape with a size of 209.6 ± 4.3 nm. Upon NIR laser irradiation, melanin@PFH@5-FU-liposomes exhibited effective temperature increase both in vitro and in vivo. In this regard, temperature increase caused a phase transition of PFH to induce bubble generation dramatically, resulting in effective drug release behavior and ultrasound imaging. The temperature of the tumor site was increased to 52 t and contrast was greatly enhanced during ultrasound imaging due to the generation of bubble. More importantly, tumor growth was effectively inhibited by injection of melanin@PFH@5-FU-liposomes with laser irradiation. CONCLUSIONS Based on intrinsic photothermal properties of melanin and phase transition properties of PFH, melanin@PFH@5-FU-liposomes exhibited effective heat and bubble generation upon NIR laser irradiation. The elevated temperature induced bubble generation, resulting in contrast enhancement of ultrasound imaging. Melanin@PFH@5-FU-liposomes under NIR laser irradiation induced the death of cancer cells, thereby effectively inhibiting tumor growth. These results suggest that melanin@PFH@5-FU-liposomes can be utilized as a promising agent for photothermal tumor therapy and ultrasound imaging.
Collapse
|
11
|
Catania R, Onion D, Russo E, Zelzer M, Mantovani G, Huett A, Stolnik S. A mechanoresponsive nano-sized carrier achieves intracellular release of drug on external ultrasound stimulus. RSC Adv 2022; 12:16561-16569. [PMID: 35754913 PMCID: PMC9169073 DOI: 10.1039/d2ra02307e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/24/2022] [Indexed: 11/21/2022] Open
Abstract
Control over intracellular release of therapeutic compounds incorporated into nano-carriers will open new possibilities for targeted treatments of various diseases including cancer, and viral and bacterial infections. Here we report our study on mechanoresponsive nano-sized liposomes which, following internalization by cells, achieve intracellular delivery of encapsulated cargo on application of external ultrasound stimulus. This is demonstrated in a bespoke cell reporter system designed to assess free drug in cytoplasm. Biophysical analyses show that drug release is attributable to the action of a mechanoresponsive spiropyran-based compound embedded in the liposomal lipid membrane. Exposure to external ultrasound stimulus results in opening of the molecular structure of the embedded spiropyran, a consequent increase in liposomal lipid membrane fluidity, and size-dependent release of encapsulated model drugs, all pointing to lipid bilayer perturbation. The study hence illustrates feasibility of the proposed concept where intracellular drug release from mechanoresponsive liposomes can be triggered on demand by external ultrasound stimulus.
Collapse
Affiliation(s)
- Rosa Catania
- School of Pharmacy, University of Nottingham Nottingham NG7 2RD UK
- School of Life Sciences, University of Nottingham Nottingham NG7 2UH UK
| | - David Onion
- School of Life Sciences, University of Nottingham Nottingham NG7 2UH UK
| | - Emanuele Russo
- School of Pharmacy, University of Nottingham Nottingham NG7 2RD UK
| | - Mischa Zelzer
- School of Pharmacy, University of Nottingham Nottingham NG7 2RD UK
| | | | - Alan Huett
- School of Life Sciences, University of Nottingham Nottingham NG7 2UH UK
| | - Snow Stolnik
- School of Pharmacy, University of Nottingham Nottingham NG7 2RD UK
| |
Collapse
|
12
|
Mukhopadhyay D, Sano C, AlSawaftah N, El-Awady R, Husseini GA, Paul V. Ultrasound-Mediated Cancer Therapeutics Delivery using Micelles and Liposomes: A Review. Recent Pat Anticancer Drug Discov 2021; 16:498-520. [PMID: 34911412 DOI: 10.2174/1574892816666210706155110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/02/2021] [Accepted: 03/21/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Existing cancer treatment methods have many undesirable side effects that greatly reduce the quality of life of cancer patients. OBJECTIVE This review will focus on the use of ultrasound-responsive liposomes and polymeric micelles in cancer therapy. METHODS This review presents a survey of the literature regarding ultrasound-triggered micelles and liposomes using articles recently published in various journals, as well as some new patents in this field. RESULTS Nanoparticles have proven promising as cancer theranostic tools. Nanoparticles are selective in nature, have reduced toxicity, and controllable drug release patterns making them ideal carriers for anticancer drugs. Numerous nanocarriers have been designed to combat malignancies, including liposomes, micelles, dendrimers, solid nanoparticles, quantum dots, gold nanoparticles, and, more recently, metal-organic frameworks. The temporal and spatial release of therapeutic agents from these nanostructures can be controlled using internal and external triggers, including pH, enzymes, redox, temperature, magnetic and electromagnetic waves, and ultrasound. Ultrasound is an attractive modality because it is non-invasive, can be focused on the diseased site, and has a synergistic effect with anticancer drugs. CONCLUSION The functionalization of micellar and liposomal surfaces with targeting moieties and the use of ultrasound as a triggering mechanism can help improve the selectivity and enable the spatiotemporal control of drug release from nanocarriers.
Collapse
Affiliation(s)
- Debasmita Mukhopadhyay
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Catherine Sano
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Nour AlSawaftah
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Raafat El-Awady
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Ghaleb A Husseini
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Vinod Paul
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
13
|
Transformable vesicles for cancer immunotherapy. Adv Drug Deliv Rev 2021; 179:113905. [PMID: 34331988 DOI: 10.1016/j.addr.2021.113905] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/22/2021] [Accepted: 07/24/2021] [Indexed: 02/06/2023]
Abstract
Immunotherapy that utilizes the human immune system to fight cancer represents a revolutionary method for cancer treatment. Immunotherapeutic agents that trigger the immune response should be carefully delivered to the desired site to maximize immunotherapy effectiveness and minimize side effects. Vesicles offer the possibility of encapsulating both hydrophilic and hydrophobic drugs and thus serve as a promising delivery tool. As multiple irreconcilable requirements exist at different transport stages, developing vesicles transformable in response to given stimuli is of great significance. In this review, we first introduced various vesicle types used for immunotherapy. Furthermore, the typical stimuli that trigger vesicle transformation and the usually generated transformation styles were described. Focusing on three aspects of antigen-presenting cell (APC)/T cell activation, tumor microenvironment (TME) amelioration, and immunogenic cell death (ICD)-induced immunotherapy, we reviewed recently reported transformable vesicles for tumor treatment. Finally, we put forward possible directions for future research and clinical translation.
Collapse
|
14
|
Veroniaina H, Pan X, Wu Z, Qi X. Apoferritin: a potential nanocarrier for cancer imaging and drug delivery. Expert Rev Anticancer Ther 2021; 21:901-913. [PMID: 33844625 DOI: 10.1080/14737140.2021.1910027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Introduction: As a protein-based biomaterial for potential cancer targeting delivery, apoferritin has recently attracted interest.Areas covered: In this review, we discuss the development of this cage-like protein as an endogenous nanocarrier that can hold molecules in its cavity. We present the specific characterizations and formulations of apoferritin nanocarriers, and outline the recent progress of the protein as an appropriate tumor-delivery vehicle in different therapeutic strategies to treat solid tumors. Finally, we propose how the application for cancer drug repurposing delivery within apoferritin could expand cancer treatment in the future.Expert opinion: Being a ubiquitous iron storage protein that exists in many living organisms, apoferritin is promising as a cancer tumor-targeting nanocarrier. By exploiting its versatility, apoferritin could be used for cancer repurposed drug delivery and could reduce the high cost of new drug discovery development and shorten the formulation process.
Collapse
Affiliation(s)
| | - Xiuhua Pan
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhenghong Wu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
15
|
Awad N, Paul V, AlSawaftah NM, ter Haar G, Allen TM, Pitt WG, Husseini GA. Ultrasound-Responsive Nanocarriers in Cancer Treatment: A Review. ACS Pharmacol Transl Sci 2021; 4:589-612. [PMID: 33860189 PMCID: PMC8033618 DOI: 10.1021/acsptsci.0c00212] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Indexed: 12/13/2022]
Abstract
The safe and effective delivery of anticancer agents to diseased tissues is one of the significant challenges in cancer therapy. Conventional anticancer agents are generally cytotoxins with poor pharmacokinetics and bioavailability. Nanocarriers are nanosized particles designed for the selectivity of anticancer drugs and gene transport to tumors. They are small enough to extravasate into solid tumors, where they slowly release their therapeutic load by passive leakage or biodegradation. Using smart nanocarriers, the rate of release of the entrapped therapeutic(s) can be increased, and greater exposure of the tumor cells to the therapeutics can be achieved when the nanocarriers are exposed to certain internally (enzymes, pH, and temperature) or externally (light, magnetic field, and ultrasound) applied stimuli that trigger the release of their load in a safe and controlled manner, spatially and temporally. This review gives a comprehensive overview of recent research findings on the different types of stimuli-responsive nanocarriers and their application in cancer treatment with a particular focus on ultrasound.
Collapse
Affiliation(s)
- Nahid
S. Awad
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| | - Vinod Paul
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| | - Nour M. AlSawaftah
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| | - Gail ter Haar
- Joint
Department of Physics, The Institute of
Cancer Research and The Royal Marsden NHS Foundation Trust, London SM2 5NG, U.K.
| | - Theresa M. Allen
- Department
of Pharmacology, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - William G. Pitt
- Department
of Chemical Engineering, Brigham Young University, Provo, Utah 84602, United States
| | - Ghaleb A. Husseini
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
16
|
Sun Y, Davis E. Nanoplatforms for Targeted Stimuli-Responsive Drug Delivery: A Review of Platform Materials and Stimuli-Responsive Release and Targeting Mechanisms. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:746. [PMID: 33809633 PMCID: PMC8000772 DOI: 10.3390/nano11030746] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
To achieve the promise of stimuli-responsive drug delivery systems for the treatment of cancer, they should (1) avoid premature clearance; (2) accumulate in tumors and undergo endocytosis by cancer cells; and (3) exhibit appropriate stimuli-responsive release of the payload. It is challenging to address all of these requirements simultaneously. However, the numerous proof-of-concept studies addressing one or more of these requirements reported every year have dramatically expanded the toolbox available for the design of drug delivery systems. This review highlights recent advances in the targeting and stimuli-responsiveness of drug delivery systems. It begins with a discussion of nanocarrier types and an overview of the factors influencing nanocarrier biodistribution. On-demand release strategies and their application to each type of nanocarrier are reviewed, including both endogenous and exogenous stimuli. Recent developments in stimuli-responsive targeting strategies are also discussed. The remaining challenges and prospective solutions in the field are discussed throughout the review, which is intended to assist researchers in overcoming interdisciplinary knowledge barriers and increase the speed of development. This review presents a nanocarrier-based drug delivery systems toolbox that enables the application of techniques across platforms and inspires researchers with interdisciplinary information to boost the development of multifunctional therapeutic nanoplatforms for cancer therapy.
Collapse
Affiliation(s)
| | - Edward Davis
- Materials Engineering Program, Mechanical Engineering Department, Auburn University, 101 Wilmore Drive, Auburn, AL 36830, USA;
| |
Collapse
|
17
|
Sun S, Wang P, Sun S, Liang X. Applications of Micro/Nanotechnology in Ultrasound-based Drug Delivery and Therapy for Tumor. Curr Med Chem 2021; 28:525-547. [PMID: 32048951 DOI: 10.2174/0929867327666200212100257] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/30/2019] [Accepted: 01/13/2020] [Indexed: 11/22/2022]
Abstract
Ultrasound has been broadly used in biomedicine for both tumor diagnosis as well as therapy. The applications of recent developments in micro/nanotechnology promote the development of ultrasound-based biomedicine, especially in the field of ultrasound-based drug delivery and tumor therapy. Ultrasound can activate nano-sized drug delivery systems by different mechanisms for ultrasound- triggered on-demand drug release targeted only at the tumor sites. Ultrasound Targeted Microbubble Destruction (UTMD) technology can not only increase the permeability of vasculature and cell membrane via sonoporation effect but also achieve in situ conversion of microbubbles into nanoparticles to promote cellular uptake and therapeutic efficacy. Furthermore, High Intensity Focused Ultrasound (HIFU), or Sonodynamic Therapy (SDT), is considered to be one of the most promising and representative non-invasive treatment for cancer. However, their application in the treatment process is still limited due to their critical treatment efficiency issues. Fortunately, recently developed micro/nanotechnology offer an opportunity to solve these problems, thus improving the therapeutic effect of cancer. This review summarizes and discusses the recent developments in the design of micro- and nano- materials for ultrasound-based biomedicine applications.
Collapse
Affiliation(s)
- Suhui Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing, China
| | - Ping Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, China
| | - Sujuan Sun
- Ordos Center Hospital, Ordos 017000, Inner Mongolia, China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, China
| |
Collapse
|
18
|
Rahim MA, Jan N, Khan S, Shah H, Madni A, Khan A, Jabar A, Khan S, Elhissi A, Hussain Z, Aziz HC, Sohail M, Khan M, Thu HE. Recent Advancements in Stimuli Responsive Drug Delivery Platforms for Active and Passive Cancer Targeting. Cancers (Basel) 2021; 13:670. [PMID: 33562376 PMCID: PMC7914759 DOI: 10.3390/cancers13040670] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
The tumor-specific targeting of chemotherapeutic agents for specific necrosis of cancer cells without affecting the normal cells poses a great challenge for researchers and scientists. Though extensive research has been carried out to investigate chemotherapy-based targeted drug delivery, the identification of the most promising strategy capable of bypassing non-specific cytotoxicity is still a major concern. Recent advancements in the arena of onco-targeted therapies have enabled safe and effective tumor-specific localization through stimuli-responsive drug delivery systems. Owing to their promising characteristic features, stimuli-responsive drug delivery platforms have revolutionized the chemotherapy-based treatments with added benefits of enhanced bioavailability and selective cytotoxicity of cancer cells compared to the conventional modalities. The insensitivity of stimuli-responsive drug delivery platforms when exposed to normal cells prevents the release of cytotoxic drugs into the normal cells and therefore alleviates the off-target events associated with chemotherapy. Contrastingly, they showed amplified sensitivity and triggered release of chemotherapeutic payload when internalized into the tumor microenvironment causing maximum cytotoxic responses and the induction of cancer cell necrosis. This review focuses on the physical stimuli-responsive drug delivery systems and chemical stimuli-responsive drug delivery systems for triggered cancer chemotherapy through active and/or passive targeting. Moreover, the review also provided a brief insight into the molecular dynamic simulations associated with stimuli-based tumor targeting.
Collapse
Affiliation(s)
- Muhammad Abdur Rahim
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (M.A.R.); (N.J.); (S.K.); (H.S.); (A.K.)
| | - Nasrullah Jan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (M.A.R.); (N.J.); (S.K.); (H.S.); (A.K.)
| | - Safiullah Khan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (M.A.R.); (N.J.); (S.K.); (H.S.); (A.K.)
| | - Hassan Shah
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (M.A.R.); (N.J.); (S.K.); (H.S.); (A.K.)
| | - Asadullah Madni
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (M.A.R.); (N.J.); (S.K.); (H.S.); (A.K.)
| | - Arshad Khan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (M.A.R.); (N.J.); (S.K.); (H.S.); (A.K.)
| | - Abdul Jabar
- College of Pharmacy, University of Sargodha, Sargodha 40100, Punjab, Pakistan;
| | - Shahzeb Khan
- Department of Pharmacy, University of Malakand, Chakdara, Dir Lower 18800, Khyber Pakhtunkhwa, Pakistan;
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Westville 3631, Durban 4000, South Africa
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Abdelbary Elhissi
- College of Pharmacy, QU Health and Office of VP for Research and Graduate Studies, Qatar University, P.O. Box 2713, Doha, Qatar;
| | - Zahid Hussain
- Department of Pharmaceutics & Pharmaceutical Technology, College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates;
- Research Institute for Medical and Health Sciences (SIMHR), University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Heather C Aziz
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Muhammad Sohail
- Department of Pharmacy, COMSATS University Abbottabad Campus, Abbottabad 45550, Khyber Pakhtunkhwa, Pakistan;
| | - Mirazam Khan
- Department of Pharmacy, University of Malakand, Chakdara, Dir Lower 18800, Khyber Pakhtunkhwa, Pakistan;
| | - Hnin Ei Thu
- Research and Innovation Department, Lincolon University College, Petaling Jaya 47301, Selangor, Malaysia;
- Innoscience Research Institute, Skypark, Subang Jaya 47650, Selangor, Malaysia
| |
Collapse
|
19
|
Kim Y, Ding H, Zheng Y. Enhancing Surface Capture and Sensing of Proteins with Low-Power Optothermal Bubbles in a Biphasic Liquid. NANO LETTERS 2020; 20:7020-7027. [PMID: 32667815 PMCID: PMC7572762 DOI: 10.1021/acs.nanolett.0c01969] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Molecular binding in surface-based biosensing is inherently governed by diffusional transport of molecules in solution to surface-immobilized counterparts. Optothermally generated surface microbubbles can quickly accumulate solutes at the bubble-liquid-substrate interface due to high-velocity fluid flows. Despite its potential as a concentrator, however, the incorporation of bubbles into protein-based sensing is limited by high temperatures. Here, we report a biphasic liquid system, capable of generating microbubbles at a low optical power/temperature by formulating PFP as a volatile, water-immiscible component in the aqueous host. We further exploited zwitterionic surface modification to prevent unwanted printing during bubble generation. In a single protein-protein interaction model, surface binding of dispersed proteins to capture proteins was enhanced by 1 order of magnitude within 1 min by bubbles, compared to that from static incubation for 30 min. Our proof-of-concept study exploiting fluid formulation and optothermal add-on paves an effective way toward improving the performances of sensors and spectroscopies.
Collapse
Affiliation(s)
- Youngsun Kim
- Materials Science and Engineering Program and Texas Materials Institute, The University of Texas at Austin, Austin, Texas 78712, USA
| | - Hongru Ding
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, Texas 78712, USA
| | - Yuebing Zheng
- Materials Science and Engineering Program and Texas Materials Institute, The University of Texas at Austin, Austin, Texas 78712, USA
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, Texas 78712, USA
| |
Collapse
|
20
|
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: 31] [Impact Index Per Article: 6.2] [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
|
21
|
Shalaby TI, El-Refaie WM, Shams El-Din RS, Hassanein SA. Smart Ultrasound-Triggered Doxorubicin-Loaded Nanoliposomes With Improved Therapeutic Response: A Comparative Study. J Pharm Sci 2020; 109:2567-2576. [DOI: 10.1016/j.xphs.2020.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/22/2020] [Accepted: 05/11/2020] [Indexed: 12/14/2022]
|
22
|
Stimuli-responsive nano-assemblies for remotely controlled drug delivery. J Control Release 2020; 322:566-592. [DOI: 10.1016/j.jconrel.2020.03.051] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/19/2020] [Accepted: 03/31/2020] [Indexed: 12/30/2022]
|
23
|
Yang W, Veroniaina H, Qi X, Chen P, Li F, Ke PC. Soft and Condensed Nanoparticles and Nanoformulations for Cancer Drug Delivery and Repurpose. ADVANCED THERAPEUTICS 2020; 3:1900102. [PMID: 34291146 PMCID: PMC8291088 DOI: 10.1002/adtp.201900102] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Indexed: 12/24/2022]
Abstract
Drug repurpose or reposition is recently recognized as a high-performance strategy for developing therapeutic agents for cancer treatment. This approach can significantly reduce the risk of failure, shorten R&D time, and minimize cost and regulatory obstacles. On the other hand, nanotechnology-based delivery systems are extensively investigated in cancer therapy due to their remarkable ability to overcome drug delivery challenges, enhance tumor specific targeting, and reduce toxic side effects. With increasing knowledge accumulated over the past decades, nanoparticle formulation and delivery have opened up a new avenue for repurposing drugs and demonstrated promising results in advanced cancer therapy. In this review, recent developments in nano-delivery and formulation systems based on soft (i.e., DNA nanocages, nanogels, and dendrimers) and condensed (i.e., noble metal nanoparticles and metal-organic frameworks) nanomaterials, as well as their theranostic applications in drug repurpose against cancer are summarized.
Collapse
Affiliation(s)
- Wen Yang
- Materials Research and Education Center, Auburn University, Auburn, AL 36849, USA
| | | | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia
| | - Pengyu Chen
- Materials Research and Education Center, Auburn University, Auburn, AL 36849, USA
| | - Feng Li
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn AL 36849, USA
| | - Pu Chun Ke
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia
| |
Collapse
|
24
|
Li J, Wang J, Zhang X, Xia X, Zhang C. Biodegradable reduction-responsive polymeric micelles for enhanced delivery of melphalan to retinoblastoma cells. Int J Biol Macromol 2019; 141:997-1003. [PMID: 31521654 DOI: 10.1016/j.ijbiomac.2019.09.085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/03/2019] [Accepted: 09/11/2019] [Indexed: 02/01/2023]
Abstract
Melphalan (MEL) is an effective chemotherapeutic agent for treatment of retinoblastoma (Rb) which is the most common childhood malignancy. However, the inherent cardiopulmonary toxicity and hazardous integration limit its therapeutic effect on RB. N-Acetylheparosan (AH), a natural heparin-like polysaccharide in mammals with long circulation effect and good biocompatibility, was linked by d-α-tocopherol acid succinate (VES) via and cystamine (CYS) to synthesize reduction-responsive N-acetylheparosan-CYS-Vitamin E succinate (AHV) copolymers. In addition, CYS was replaced by adipic acid dihydrazide (ADH) to obtain a control of non-reduction-responsive polymers N-acetylheparosan-ADH-Vitamin E succinate (ADV). MEL-loaded AHV micelles (MEL/AHV) as well as ADV micelles (MEL/ADV) were prepared with small particle size and high drug loading content. In vitro drug release showed that MEL/AHV micelles presented obvious reduction-triggered release behavior compared with MEL/ADV. In vitro antitumor effects were investigated using WERI-Rb-1 retinoblastoma cells. Cytotoxicity experiments showed that the IC50 of MEL/AHV was significantly lower than that of free MEL and MEL/ADV, suggesting that MEL/AHV enhanced the cytotoxicity against retinoblastoma cells. Furthermore, MEL/AHV micelles were more easily uptaken by multiple pathways compared with MEL/ADV and free MEL. Therefore, MEL/AHV might be a potential delivery system for enhanced delivery of melphalan to Rb cells.
Collapse
Affiliation(s)
- Jia Li
- Department of Ophthalmology, Affiliated Hospital of Jiangnan University, Wuxi 214062, China.
| | - Jihong Wang
- Department of Ophthalmology, Affiliated Hospital of Jiangnan University, Wuxi 214062, China
| | - Xuetong Zhang
- Department of Ophthalmology, Affiliated Hospital of Jiangnan University, Wuxi 214062, China
| | - Xin Xia
- Department of Ophthalmology, Affiliated Hospital of Jiangnan University, Wuxi 214062, China
| | - Chenchen Zhang
- Department of Ophthalmology, Affiliated Hospital of Jiangnan University, Wuxi 214062, China
| |
Collapse
|
25
|
Lin CY, Tsai CH, Feng LY, Chai WY, Lin CJ, Huang CY, Wei KC, Yeh CK, Chen CM, Liu HL. Focused ultrasound-induced blood brain-barrier opening enhanced vascular permeability for GDNF delivery in Huntington's disease mouse model. Brain Stimul 2019; 12:1143-1150. [PMID: 31079989 DOI: 10.1016/j.brs.2019.04.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 01/30/2019] [Accepted: 04/25/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the gene encoding the huntingtin (Htt) protein, which results in a protein containing an abnormally expanded polyglutamine (polyQ) sequence. The expanded polyQ in the Htt protein is toxic to brain cells. No therapy exists to delay disease progression. METHODS This study describes a gene-liposome system that synergistically applied focused ultrasound (FUS)-blood-brain barrier (BBB) opening for rescuing motor and neuropathological impairments when administered from pre to post-symptomatic transgenic mouse models of HD. DPPC liposomes (LPs) are designed to carry glia cell line-derived neurotrophic factor (GDNF) plasmid DNA (GDNFp) to form a GDNFp-liposome (GDNFp-LPs) complex. Pulsed FUS exposure with microbubbles (MBs) was used to induce BBB opening for non-viral, non-invasive, and targeted gene delivery into the central nervous system (CNS) for therapeutic purposes. RESULTS FUS-gene therapy significantly improved motor performance with GDNFp-LPs + FUS treated HD mice equilibrating longer periods in the animal behavior. Reflecting the improvements observed in motor function, GDNF overexpression results in significantly decreased formation of polyglutamine-expanded aggregates, reduced oxidative stress and apoptosis, promoted neurite outgrowth, and improved neuronal survival. Immunoblotting and histological staining further confirmed the neuroprotective effect from delivery of GDNF genes to neuronal cells. CONCLUSIONS This study suggests that the GDNFp-LPs plus FUS sonication can provide an effective gene therapy to achieve local extravasation and triggered gene delivery for non-invasive in vivo treatment of CNS diseases.
Collapse
Affiliation(s)
- Chung-Yin Lin
- Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan; Department of Nephrology and Clinical Poison Center, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - Chih-Hung Tsai
- Department of Electrical Engineering, Chang Gung University, Taoyuan, 333, Taiwan
| | - Li-Ying Feng
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Wen-Yen Chai
- Department of Electrical Engineering, Chang Gung University, Taoyuan, 333, Taiwan
| | - Chia-Jung Lin
- Department of Electrical Engineering, Chang Gung University, Taoyuan, 333, Taiwan
| | - Chiung-Yin Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan.
| | - Hao-Li Liu
- Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan; Department of Electrical Engineering, Chang Gung University, Taoyuan, 333, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan.
| |
Collapse
|
26
|
Hamarat Şanlıer Ş, Ak G, Yılmaz H, Ünal A, Bozkaya ÜF, Tanıyan G, Yıldırım Y, Yıldız Türkyılmaz G. Development of Ultrasound-Triggered and Magnetic-Targeted Nanobubble System for Dual-Drug Delivery. J Pharm Sci 2019; 108:1272-1283. [DOI: 10.1016/j.xphs.2018.10.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/14/2018] [Accepted: 10/16/2018] [Indexed: 01/06/2023]
|
27
|
Zhou X, Guo L, Shi D, Duan S, Li J. Biocompatible Chitosan Nanobubbles for Ultrasound-Mediated Targeted Delivery of Doxorubicin. NANOSCALE RESEARCH LETTERS 2019; 14:24. [PMID: 30649655 PMCID: PMC6335234 DOI: 10.1186/s11671-019-2853-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 01/03/2019] [Indexed: 05/18/2023]
Abstract
Ultrasound-targeted delivery of nanobubbles (NBs) has become a promising strategy for noninvasive drug delivery. The biosafety and drug-transporting ability of NBs have been a research hotspot, especially regarding chitosan NBs due to their biocompatibility and high biosafety. Since the drug-carrying capacity of chitosan NBs and the performance of ultrasound-assisted drug delivery remain unclear, the aim of this study was to synthesize doxorubicin hydrochloride (DOX)-loaded biocompatible chitosan NBs and assess their drug delivery capacity. In this study, the size distribution of chitosan NBs was measured by dynamic light scattering, while their drug-loading capacity and ultrasound-mediated DOX release were determined by a UV spectrophotometer. In addition, a clinical ultrasound imaging system was used to evaluate the ability of chitosan NBs to achieve imaging enhancement, while the biosafety profile of free chitosan NBs was evaluated by a cytotoxicity assay in MCF-7 cells. Furthermore, NB-mediated DOX uptake and the apoptosis of Michigan Cancer Foundation-7 (MCF-7) cells were measured by flow cytometry. The results showed that the DOX-loaded NBs (DOX-NBs) exhibited excellent drug-loading ability as well as the ability to achieve ultrasound enhancement. Ultrasound (US) irradiation promoted the release of DOX from DOX-NBs in vitro. Furthermore, DOX-NBs effectively delivered DOX into mammalian cancer cells. In conclusion, biocompatible chitosan NBs are suitable for ultrasound-targeted DOX delivery and are thus a promising strategy for noninvasive and targeted drug delivery worthy of further investigation.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Department of Ultrasound, Qilu Hospital of Shandong University, West Wenhua Road, Jinan, Shandong China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital of Shandong University, West Wenhua Road, Jinan, Shandong China
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital of Shandong University, West Wenhua Road, Jinan, Shandong China
| | - Sujuan Duan
- Department of Ultrasound, Qilu Hospital of Shandong University, West Wenhua Road, Jinan, Shandong China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, West Wenhua Road, Jinan, Shandong China
| |
Collapse
|
28
|
Zhu B, Wang L, Huang J, Xiang X, Tang Y, Cheng C, Yan F, Ma L, Qiu L. Ultrasound-triggered perfluorocarbon-derived nanobombs for targeted therapies of rheumatoid arthritis. J Mater Chem B 2019. [DOI: 10.1039/c9tb00978g] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The targeted US-triggered PFC-based “nanobombs” with US used to treat the RA in this work would offer a new treatment strategy and have a great potential for the application in the areas of theranostic agent and nanomedicine treatment.
Collapse
Affiliation(s)
- Bihui Zhu
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Liyun Wang
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Jianbo Huang
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Xi Xiang
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Yuanjiao Tang
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Chong Cheng
- College of Polymer Science and Engineering
- State Key Laboratory of Polymer Materials Engineering
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Feng Yan
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Lang Ma
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Li Qiu
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| |
Collapse
|
29
|
Li M, Wu D, Chen Y, Shan G, Liu Y. Apoferritin nanocages with Au nanoshell coating as drug carrier for multistimuli-responsive drug release. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 95:11-18. [PMID: 30573231 DOI: 10.1016/j.msec.2018.10.060] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 09/10/2018] [Accepted: 10/15/2018] [Indexed: 12/13/2022]
Abstract
Cancer is one of the major causes of mortality worldwide. Therefore, it is necessary to provide an effective method of tumor therapy. Herein, we designed a new type of composite particle, apoferritin (APO) encapsulated doxorubicin (DOX), and the surface of APO was modified with Au nanoshell. As a nanocarrier, APO can carry chemotherapy drug DOX (APODOX) and release drug under acidic and high temperature conditions to reduce side effects of anticancer drugs. After covering Au nanoshell (APODOX-Au), the photothermal effect can be produced because of the unique surface plasmon resonance properties of gold nanoshell. This nanoplatform also provides the multi-stimuli responsive drug release system, which can achieve drug release in different conditions and have great potential in biomedical applications. Our investigation has demonstrated that APODOX-Au has good stability, high dispersibility and biocompatibility in vitro. The strong near-infrared absorption and good photothermal effect make sure the quick response to environmental changes (pH, temperature) to achieve drug release. These findings indicate that these nanoparticles have a potential application value in cancer treatment.
Collapse
Affiliation(s)
- Man Li
- Centre for Advanced Optoelectronic Functional Materials Research, Key Laboratory for UV Light-Emitting Materials and Technology of Ministry of Education, Northeast Normal University, Changchun 130024, China
| | - Dan Wu
- Centre for Advanced Optoelectronic Functional Materials Research, Key Laboratory for UV Light-Emitting Materials and Technology of Ministry of Education, Northeast Normal University, Changchun 130024, China
| | - Yanwei Chen
- Centre for Advanced Optoelectronic Functional Materials Research, Key Laboratory for UV Light-Emitting Materials and Technology of Ministry of Education, Northeast Normal University, Changchun 130024, China.
| | - Guiye Shan
- Centre for Advanced Optoelectronic Functional Materials Research, Key Laboratory for UV Light-Emitting Materials and Technology of Ministry of Education, Northeast Normal University, Changchun 130024, China
| | - Yichun Liu
- Centre for Advanced Optoelectronic Functional Materials Research, Key Laboratory for UV Light-Emitting Materials and Technology of Ministry of Education, Northeast Normal University, Changchun 130024, China
| |
Collapse
|
30
|
de Leon A, Perera R, Nittayacharn P, Cooley M, Jung O, Exner AA. Ultrasound Contrast Agents and Delivery Systems in Cancer Detection and Therapy. Adv Cancer Res 2018; 139:57-84. [PMID: 29941107 DOI: 10.1016/bs.acr.2018.04.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ultrasound is the second most utilized imaging modality in the world because it is widely accessible, robust, and safe. Aside from its extensive use in diagnostic imaging, ultrasound has also been frequently utilized in therapeutic applications. Particularly, when combined with appropriate delivery systems, ultrasound provides a flexible platform for simultaneous real-time imaging and triggered release, enabling precise, on-demand drug delivery to target sites. This chapter will discuss the basics of ultrasound including its mechanism of action and how it can be used to trigger the release of encapsulated drug either through thermal or cavitation effects. Fundamentals of ultrasound contrast agents, how they enhance ultrasound signals, and how they can be modified to function as carriers for triggered and targeted release of drugs will also be discussed.
Collapse
Affiliation(s)
- Al de Leon
- Department of Radiology, Case Western Reserve University, Cleveland, OH, United States
| | - Reshani Perera
- Department of Radiology, Case Western Reserve University, Cleveland, OH, United States
| | - Pinunta Nittayacharn
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Michaela Cooley
- Department of Radiology, Case Western Reserve University, Cleveland, OH, United States
| | - Olive Jung
- Department of Radiology, Case Western Reserve University, Cleveland, OH, United States; Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Agata A Exner
- Department of Radiology, Case Western Reserve University, Cleveland, OH, United States; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States.
| |
Collapse
|
31
|
Tang J, Mi J, Huang W, Zhong H, Li Y, Zhou J, Johri AM. Controlled drug release from ultrasound-visualized elastic eccentric microcapsules using different resonant modes. J Mater Chem B 2018; 6:1920-1929. [DOI: 10.1039/c7tb03164e] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The release rate of drug from elastic eccentric microcapsules can be regulated, based on their mode shapes and resonant natural frequencies.
Collapse
Affiliation(s)
- Junyun Tang
- Department of Biomedical Engineering
- School of Engineering
- Sun Yat-sen University
- Guangzhou 510275
- China
| | - Jiaomei Mi
- Department of Biomedical Engineering
- School of Engineering
- Sun Yat-sen University
- Guangzhou 510275
- China
| | - Wenwei Huang
- Department of Biomedical Engineering
- School of Engineering
- Sun Yat-sen University
- Guangzhou 510275
- China
| | - Huixiang Zhong
- Department of Biomedical Engineering
- School of Engineering
- Sun Yat-sen University
- Guangzhou 510275
- China
| | - Yan Li
- Department of Biomedical Engineering
- School of Engineering
- Sun Yat-sen University
- Guangzhou 510275
- China
| | - Jianhua Zhou
- Department of Biomedical Engineering
- School of Engineering
- Sun Yat-sen University
- Guangzhou 510275
- China
| | - Amer M. Johri
- Department of Medicine
- Division of Cardiology
- Cardiovascular Imaging Network at Queen's University
- Kingston
- Canada
| |
Collapse
|
32
|
Lin CY, Li RJ, Huang CY, Wei KC, Chen PY. Controlled release of liposome-encapsulated temozolomide for brain tumour treatment by convection-enhanced delivery. J Drug Target 2017; 26:325-332. [DOI: 10.1080/1061186x.2017.1379526] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Chung-Yin Lin
- Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan, ROC
- Department of Nephrology, Division of Clinical Toxicology, Chang Gung Memorial Hospital, Lin-Kou Medical Center, Taoyuan, ROC
| | - Rui-Jin Li
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan, ROC
| | - Chiung-Yin Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan, ROC
| | - Kuo-Chen Wei
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan, ROC
| | - Pin-Yuan Chen
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan, ROC
| |
Collapse
|
33
|
甘 珍, 武 昊, 吴 昊, 周 美, 严 飞, 刘 红. [Efficacy of internalized RGD-modified echogenic liposomes in diagnosis and treatment in a mouse model of rheumatoid arthritis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:1283-1289. [PMID: 29070455 PMCID: PMC6743967 DOI: 10.3969/j.issn.1673-4254.2017.10.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To prepare internalized RGD (iRGD) modified echogenic liposomes containing methotrexate (MTX) and indocyanine green (ICG) (iRGD MTX ICG ELIP) and evaluate its targeting efficiency and inhibitory effect combined with ultrasound on synovial cells. METHODS iRGD MTX ICG ELIP was prepared by the thin film rehydration and freeze-lyophilization method and its general characteristics and acoustic responsiveness were assessed. The targeting effect of the prepared liposome was observed by assessing its cell uptake in vitro. In a mouse model of rheumatiod arthritis, the targeting effect of the prepared liposome was determined by detecting the fluorescence intensity of the drug in arthrosis. The inhibitory effect of iRGD MTX ICG ELIP combined with ultrasound on synovial MH7A cells in vitro were investigated using CCK8 test. RESULTS The average diameter and zeta potential of iRGD MTX ICG ELIP was 134.4∓17.61 nm and 10.07∓4.28 mV, and the entrapment efficiency of MTX and ICG was (62.56∓0.77)% and (95.13∓0.82)%, respectively. With ultrasound exposure, the release of MTX and ICG from iRGD MTX ICG ELIP increased with the ultrasound intensity and with the exposure time. In HUVECs, the uptake efficiency of iRGD MTX ICG ELIP was 1.89 times higher than that of non targeted MTX ICG ELIP (P<0.05). In vivo imaging of mouse joint with rheumatiod arthritis showed that the fluorescence intensity of iRGD MTX ICG ELIP was significantly stronger than that of the non targeted liposome. CCK8 assay showed that iRGD MTX ICG ELIP combined with ultrasound resulted in a survival rate of MH7A cells of (32.49∓3.04)%, significantly lower than the rate of cells treated with iRGD MTX ICG ELIP but without ultrasound (P<0.05). CONCLUSIONS iRGD MTX ICG ELIP has a suitable particle size and can effectively target HUVECs and the joints with rheumatiod arthritis. With a good drug entrapment efficiency and acoustic responsiveness, the drug loaded liposome shows enhanced inhibitory effect on MH7A cells combined with ultrasound in vitro, suggesting its potential in the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- 珍 甘
- 南方医科大学第三附属医院超声医学科//广东省骨科研究院, 广东 广州 510630Department of Ultrasonography, Third Affiliated Hospital of Southern Medical University/Academy of Orthopedics of Guangdong Province, Guangzhou 510630, China
| | - 昊 武
- 南方医科大学第三附属医院超声医学科//广东省骨科研究院, 广东 广州 510630Department of Ultrasonography, Third Affiliated Hospital of Southern Medical University/Academy of Orthopedics of Guangdong Province, Guangzhou 510630, China
| | - 昊晗 吴
- 南方医科大学第三附属医院超声医学科//广东省骨科研究院, 广东 广州 510630Department of Ultrasonography, Third Affiliated Hospital of Southern Medical University/Academy of Orthopedics of Guangdong Province, Guangzhou 510630, China
| | - 美君 周
- 南方医科大学第三附属医院超声医学科//广东省骨科研究院, 广东 广州 510630Department of Ultrasonography, Third Affiliated Hospital of Southern Medical University/Academy of Orthopedics of Guangdong Province, Guangzhou 510630, China
| | - 飞 严
- 中国科学院深圳先进技术研究院保罗. C. 劳特伯生物医学成像中心, 广东 深圳 518055Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - 红梅 刘
- 南方医科大学第三附属医院超声医学科//广东省骨科研究院, 广东 广州 510630Department of Ultrasonography, Third Affiliated Hospital of Southern Medical University/Academy of Orthopedics of Guangdong Province, Guangzhou 510630, China
- 南方医科大学附属广东省第二人民医院超声医学科, 广东 广州 510317Department of Ultrasonography, Guangdong Second Provincial General Hospital Affiliated to Southern Medical University, Guangzhou, 510317, China
| |
Collapse
|
34
|
Rwei AY, Paris JL, Wang B, Wang W, Axon CD, Vallet-Regí M, Langer R, Kohane DS. Ultrasound-triggered local anaesthesia. Nat Biomed Eng 2017; 1:644-653. [PMID: 29152410 PMCID: PMC5687284 DOI: 10.1038/s41551-017-0117-6] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 06/27/2017] [Indexed: 01/09/2023]
Abstract
On-demand relief of local pain would allow patients to control the timing, intensity and duration of nerve block in a safe and non-invasive manner. Ultrasound would be a suitable trigger for such a system, as it is in common clinical use and can penetrate deeply into the body. Here, we demonstrate that ultrasound-triggered delivery of an anaesthetic from liposomes allows the timing, intensity and duration of nerve block to be controlled by ultrasound parameters. On insonation, the encapsulated sonosensitizer protoporphyrin IX produces reactive oxygen species that react with the liposomal membrane, leading to the release of the potent local anaesthetic tetrodotoxin. We also show repeatable ultrasound-triggered nerve blocks in vivo, with nerve-block duration depending on the extent and intensity of insonation. We did not detect any systemic toxicity, and tissue reaction was benign in all groups. On-demand, personalized local anaesthesia could be beneficial for the managing of relatively localized pain states, and potentially minimize opioid use.
Collapse
Affiliation(s)
- Alina Y Rwei
- Department of Anaesthesiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Juan L Paris
- Dpto. Química Inorgánica y Bioinorgánica, Facultad de Farmacia, UCM, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029, Madrid, Spain
| | - Bruce Wang
- Department of Anaesthesiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Weiping Wang
- Dr Li Dak-Sum Research Centre, The University of Hong Kong-Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Hong Kong, China
| | - Christopher D Axon
- Department of Anaesthesiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - María Vallet-Regí
- Dpto. Química Inorgánica y Bioinorgánica, Facultad de Farmacia, UCM, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029, Madrid, Spain
| | - Robert Langer
- David H. Koch Institutes for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Daniel S Kohane
- Department of Anaesthesiology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Laboratory for Biomaterials and Drug Delivery, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
35
|
Nguyen AT, Sunny Y, Bawiec C, Lewin PA, Wrenn SP. Investigating the spatial extent of acoustically activated echogenic liposomes. ULTRASONICS 2017; 77:176-182. [PMID: 28246036 DOI: 10.1016/j.ultras.2017.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 01/26/2017] [Accepted: 01/26/2017] [Indexed: 06/06/2023]
Abstract
The purpose of this work was to investigate the ability of bubbles entrapped within echogenic liposomes (ELIP) to serve as foci for cavitational events that would cause leakage in neighboring non-echogenic liposomes (NELIP). Previous studies have shown that entrapping bubbles into liposomes increases ultrasound-mediated leakage of hydrophilic components at ultrasound settings known to induce inertial cavitation, specifically 20kHz and 2.2W/cm2. Using tone-burst approach and pulse repetition frequency of 10Hz would bring this intensity level to the one accepted (220mW/cm2) in clinical imaging. Mixed populations of ELIP and NELIP were simultaneously exposed to ultrasound at varying ratios to examine the effect of ELIP concentration on release of a hydrophilic dye, calcein, from NELIP. Calcein release from NELIP was observed to be independent of ELIP concentration, suggesting that the release enhancement from echogenicity is strictly a localized event. Additionally, it was observed that the release mechanisms independent of echogenicity were active for the duration of experiment whereas those associated with echogenicity were active for only the initial 1-2min.
Collapse
Affiliation(s)
- An T Nguyen
- School of Biomedical Engineering, Science and Health Systems, Drexel University, United States
| | - Youhan Sunny
- School of Biomedical Engineering, Science and Health Systems, Drexel University, United States
| | - Christopher Bawiec
- School of Biomedical Engineering, Science and Health Systems, Drexel University, United States
| | - Peter A Lewin
- School of Biomedical Engineering, Science and Health Systems, Drexel University, United States
| | - Steven P Wrenn
- Department of Chemical & Biological Engineering, Drexel University, United States.
| |
Collapse
|
36
|
Affiliation(s)
- Yuqi Zhang
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Center
for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics,
UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department
of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Jicheng Yu
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Center
for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics,
UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Hunter N. Bomba
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Yong Zhu
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Department
of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Zhen Gu
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Center
for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics,
UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department
of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
37
|
Lin CY, Hsieh HY, Chen CM, Wu SR, Tsai CH, Huang CY, Hua MY, Wei KC, Yeh CK, Liu HL. Non-invasive, neuron-specific gene therapy by focused ultrasound-induced blood-brain barrier opening in Parkinson's disease mouse model. J Control Release 2016; 235:72-81. [DOI: 10.1016/j.jconrel.2016.05.052] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/28/2016] [Accepted: 05/24/2016] [Indexed: 02/01/2023]
|
38
|
Wang C, Li W, Hu B. The anti-tumor effect of folate-targeted liposome microbubbles loaded with oridonin as ultrasound-triggered tumor-targeted therapeutic carrier system. J Drug Target 2016; 25:83-91. [DOI: 10.1080/1061186x.2016.1200588] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
39
|
Dostalova S, Cerna T, Hynek D, Koudelkova Z, Vaculovic T, Kopel P, Hrabeta J, Heger Z, Vaculovicova M, Eckschlager T, Stiborova M, Adam V. Site-Directed Conjugation of Antibodies to Apoferritin Nanocarrier for Targeted Drug Delivery to Prostate Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2016; 8:14430-14441. [PMID: 27219717 DOI: 10.1021/acsami.6b04286] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Herein, we describe a novel approach for targeting of ubiquitous protein apoferritin (APO)-encapsulating doxorubicin (DOX) to prostate cancer using antibodies against prostate-specific membrane antigen (PSMA). The conjugation of anti-PSMA antibodies and APO was carried out using HWRGWVC heptapeptide, providing their site-directed orientation. The prostate-cancer-targeted and nontargeted nanocarriers were tested using LNCaP and HUVEC cell lines. A total of 90% of LNCaP cells died after treatment with DOX (0.25 μM) or DOX in nontargeted and prostate-cancer-targeted APO, proving that the encapsulated DOX toxicity for LNCaP cells remained the same. Free DOX showed higher toxicity for nonmalignant cells, whereas the toxicity was lower after treatment with the same dosage of APO-encapsulated DOX (APODOX) and even more in prostate-cancer-targeted APODOX. Hemolytic assay revealed exceptional hemocompatibility of the entire nanocarrier. The APO encapsulation mechanism ensures applicability using a wide variety of chemotherapeutic drugs, and the presented surface modification enables targeting to various tumors.
Collapse
Affiliation(s)
- Simona Dostalova
- Department of Chemistry and Biochemistry, Mendel University in Brno , Zemedelska 1, Brno CZ-613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology , Purkynova 123, Brno CZ-612 00, Czech Republic
| | - Tereza Cerna
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol , V Uvalu 84/1, Prague 5 CZ-150 06, Czech Republic
- Department of Biochemistry, Faculty of Science, Charles University in Prague , Hlavova 2030/8, Prague 2 CZ-128 43, Czech Republic
| | - David Hynek
- Department of Chemistry and Biochemistry, Mendel University in Brno , Zemedelska 1, Brno CZ-613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology , Purkynova 123, Brno CZ-612 00, Czech Republic
| | - Zuzana Koudelkova
- Department of Chemistry and Biochemistry, Mendel University in Brno , Zemedelska 1, Brno CZ-613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology , Purkynova 123, Brno CZ-612 00, Czech Republic
| | - Tomas Vaculovic
- Department of Chemistry, Faculty of Science, Masaryk University , Kotlarska 2, Brno CZ-611 37, Czech Republic
| | - Pavel Kopel
- Department of Chemistry and Biochemistry, Mendel University in Brno , Zemedelska 1, Brno CZ-613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology , Purkynova 123, Brno CZ-612 00, Czech Republic
| | - Jan Hrabeta
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol , V Uvalu 84/1, Prague 5 CZ-150 06, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno , Zemedelska 1, Brno CZ-613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology , Purkynova 123, Brno CZ-612 00, Czech Republic
| | - Marketa Vaculovicova
- Department of Chemistry and Biochemistry, Mendel University in Brno , Zemedelska 1, Brno CZ-613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology , Purkynova 123, Brno CZ-612 00, Czech Republic
| | - Tomas Eckschlager
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol , V Uvalu 84/1, Prague 5 CZ-150 06, Czech Republic
| | - Marie Stiborova
- Department of Biochemistry, Faculty of Science, Charles University in Prague , Hlavova 2030/8, Prague 2 CZ-128 43, Czech Republic
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno , Zemedelska 1, Brno CZ-613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology , Purkynova 123, Brno CZ-612 00, Czech Republic
| |
Collapse
|
40
|
Aw MS, Paniwnyk L, Losic D. The progressive role of acoustic cavitation for non-invasive therapies, contrast imaging and blood-tumor permeability enhancement. Expert Opin Drug Deliv 2016; 13:1383-96. [PMID: 27195384 DOI: 10.1080/17425247.2016.1192123] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Drug delivery pertaining to acoustic cavitation generated from ultrasonic (US) irradiation is advantageous for devising smarter and more advanced therapeutics. The aim is to showcase microbubbles as drug carriers and robust theranostic for non-invasive therapies across diverse biomedical disciplines, highlighting recent technologies in this field for overcoming the blood-brain barrier (BBB) to treat cancers and neurological disorders. AREAS COVERED This article reviews work on the optimized tuning of ultrasonic parameters, sonoporation, transdermal and responsive drug delivery, acoustic cavitation in vasculature and oncology, contrast imaging for real-time magnification of cell-microbubble dynamics and biomolecular targeting. Scholarly literature was sought through database search on key terminology, latest topics, reputable experts and established journals over the last five years. EXPERT OPINION Cavitation offers immense promise in overcoming current diffusion and convection limitations for treating skull/brain/vascular/tissue injuries and ablating tumors to minimize chronic/acute effects. Since stable cavitation facilitates the restoration of US-opened BBB and the modulation of drug concentration, US equipment with programmable imaging modality and sensitivity are envisaged to create safer miniaturized devices for personalized care. Due to differing biomedical protocols with regard to specific medical conditions, quantitative and qualitative controls are mandatory before translation to real-life clinical applications can be accomplished.
Collapse
Affiliation(s)
- Moom Sinn Aw
- a School of Chemical Engineering , The University of Adelaide , Adelaide , Australia.,b Faculty of Health and Life Sciences , Coventry University , West Midlands , UK
| | - Larysa Paniwnyk
- c Faculty of Health and Life Sciences , Coventry University , West Midlands , UK
| | - Dusan Losic
- a School of Chemical Engineering , The University of Adelaide , Adelaide , Australia
| |
Collapse
|
41
|
Xie X, Lin W, Li M, Yang Y, Deng J, Liu H, Chen Y, Fu X, Liu H, Yang Y. Efficient siRNA Delivery Using Novel Cell-Penetrating Peptide-siRNA Conjugate-Loaded Nanobubbles and Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:1362-1374. [PMID: 27012462 DOI: 10.1016/j.ultrasmedbio.2016.01.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 01/13/2016] [Accepted: 01/27/2016] [Indexed: 06/05/2023]
Abstract
Because of the absence of tolerable and effective carriers for in vivo delivery, the applications of small interfering RNA (siRNA) in the clinic for therapeutic purposes have been limited. In this study, development of a novel siRNA delivery system based on ultrasound-sensitive nanobubbles (NBs, nano-sized echogenic liposomes) and cell-permeable peptides (CPPs) is described. A CPP-siRNA conjugate was entrapped in an NB, (CPP-siRNA)-NB, and the penetration of CPP-siRNA was temporally masked; local ultrasound stimulation triggered the release of CPP-siRNA from the NBs and activated its penetration. Subsequent research revealed that the (CPP-siRNA)-NBs had a mean particle size of 201 ± 2.05 nm and a siRNA entrapment efficiency >85%. In vitro release results indicated that >90% of the encapsulated CPP-siRNA was released from NBs in the presence of ultrasound, whereas <1.5% (30 min) was released in the absence of ultrasound. Cell experiments indicated higher cellular CPP-siRNA uptake of (CPP-siRNA)-NBs with ultrasound among the various formulations in human breast adenocarcinoma cells (HT-1080). Additionally, after systemic administration in mice, (CPP-siRNA)-NBs accumulated in the tumor, augmented c-myc silencing and delayed tumor progression. In conclusion, the application of (CPP-siRNA)-NBs with ultrasound may constitute an approach to selective targeted delivery of siRNA.
Collapse
Affiliation(s)
- Xiangyang Xie
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Wen Lin
- Department of Clinical Laboratory, Huangshi Love & Health Hospital of Hubei Province, Huangshi, China
| | - Mingyuan Li
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yang Yang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| | - Jianping Deng
- Department of Clinical Laboratory, Huangshi Love & Health Hospital of Hubei Province, Huangshi, China
| | - Hui Liu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Ying Chen
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Xudong Fu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Hong Liu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
42
|
Kim Y, Lee S, Kim S. Preparation of Fluorous Solvent-Dispersed Fe3O4 Nanocrystals: Role of Oxygen in Ligand Exchange. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:3348-3353. [PMID: 27018461 DOI: 10.1021/acs.langmuir.6b00526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Perfluorinated ligand-passivated Fe3O4 nanocrystals were prepared through a biphasic ligand exchange method. It was found that dissolved oxygen in the reaction media predominantly determined the degree of ligand exchange and the resultant dispersion property of nanocrystals in a fluorous solvent. X-ray photoelectron spectroscopic analyses revealed that dissolved oxygen molecules bind to the surface iron species of nanocrystals in competition with the carboxylate moiety of ligands during the exchange reaction, lowering the degree of ligand exchange and colloidal stability significantly. Reducing the oxygen content of the fluorous phase by N2 bubbling was found to result in a highly stable dispersion of phase-transferred Fe3O4 nanocrystals with a single-particle size distribution maintained for a few months.
Collapse
Affiliation(s)
- Youngsun Kim
- Center for Theragnosis and ‡Center for Bionics, Korea Institute of Science and Technology (KIST) , 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Sangyoup Lee
- Center for Theragnosis and ‡Center for Bionics, Korea Institute of Science and Technology (KIST) , 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Sehoon Kim
- Center for Theragnosis and ‡Center for Bionics, Korea Institute of Science and Technology (KIST) , 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
43
|
Rizzitelli S, Giustetto P, Faletto D, Delli Castelli D, Aime S, Terreno E. The release of Doxorubicin from liposomes monitored by MRI and triggered by a combination of US stimuli led to a complete tumor regression in a breast cancer mouse model. J Control Release 2016; 230:57-63. [PMID: 27049069 DOI: 10.1016/j.jconrel.2016.03.040] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 03/25/2016] [Accepted: 03/26/2016] [Indexed: 01/26/2023]
Abstract
The work aimed at developing a novel MRI-based theranostic protocol for improving the anticancer efficacy of a Doxil-like liposomal formulation. The goal was achieved stimulating the intratumor release of the drug from the nanocarrier and favoring its diffusion in the lesion by the sequential application of low-intensity pulsed ultrasound. The protocol was tested on mice bearing a syngeneic breast cancer model. The combination of acoustic waves with different characteristics allowed for: i) the release of the drug and the co-encapsulated MRI agent (Gadoteridol) from the liposomes in the vessels of the tumor region, and ii) the extravasation of the released material, as well as intact liposomes, in the tumor stroma. The MR-T1 contrast enhancement measured in the tumor reported on the delivery and US-triggered release of Doxorubicin. The developed protocol resulted in a marked increase in the intratumor drug concentration that, in turn, led to the complete regression of the lesion. The protocol has a good clinical translatability because all the components of the theranostic agent (Doxorubicin, liposomes, Gadoteridol) are approved for human use.
Collapse
Affiliation(s)
- S Rizzitelli
- Molecular & Preclinical Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - P Giustetto
- Molecular & Preclinical Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - D Faletto
- Molecular & Preclinical Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - D Delli Castelli
- Molecular & Preclinical Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - S Aime
- Molecular & Preclinical Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - E Terreno
- Molecular & Preclinical Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy.
| |
Collapse
|
44
|
Jeong K, Kang CS, Kim Y, Lee YD, Kwon IC, Kim S. Development of highly efficient nanocarrier-mediated delivery approaches for cancer therapy. Cancer Lett 2016; 374:31-43. [DOI: 10.1016/j.canlet.2016.01.050] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/13/2016] [Accepted: 01/26/2016] [Indexed: 10/22/2022]
|
45
|
Huynh E, Rajora MA, Zheng G. Multimodal micro, nano, and size conversion ultrasound agents for imaging and therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 8:796-813. [PMID: 27006001 DOI: 10.1002/wnan.1398] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 01/30/2016] [Accepted: 02/02/2016] [Indexed: 12/20/2022]
Abstract
Ultrasound (US) is one of the most commonly used clinical imaging techniques. However, the use of US and US-based intravenous agents extends far beyond imaging. In particular, there has been a surge in the fabrication of multimodality US contrast agents and theranostic US agents for cancer imaging and therapy. The unique interaction of US waves with microscale and nanoscale agents has attracted much attention in the development of contrast agents and drug-delivery vehicles. The dimensions of the agent not only dictate how it behaves in vivo, but also how it interacts with US for imaging and drug delivery. Furthermore, these agents are also unique due to their ability to convert from the nanoscale to the microscale and vice versa, having imaging and therapeutic utility in both dimensions. Here, we review multimodality and multifunctional US-based agents, according to their size, and also highlight recent developments in size conversion US agents. WIREs Nanomed Nanobiotechnol 2016, 8:796-813. doi: 10.1002/wnan.1398 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Elizabeth Huynh
- Princess Margaret Cancer Center and Techna Institute, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Maneesha A Rajora
- Princess Margaret Cancer Center and Techna Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Gang Zheng
- Princess Margaret Cancer Center and Techna Institute, University Health Network, Toronto, Ontario, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. .,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
46
|
Hwang JY, Li Z, Loh XJ. Small molecule therapeutic-loaded liposomes as therapeutic carriers: from development to clinical applications. RSC Adv 2016. [DOI: 10.1039/c6ra09854a] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In this review, various methods and mechanisms for encapsulation of small therapeutic molecules in liposomes for targeted delivery and triggered release, as well as their potential in the clinical uses, are discussed.
Collapse
Affiliation(s)
- Jae Yoon Hwang
- Department of Materials Science and Engineering
- National University of Singapore
- Singapore 117576
- Singapore
| | - Zibiao Li
- Institute of Materials Research and Engineering (IMRE)
- Singapore 117602
- Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE)
- Singapore 117602
- Singapore
- Department of Materials Science and Engineering
- National University of Singapore
| |
Collapse
|
47
|
Suzuki R, Omata D, Oda Y, Unga J, Negishi Y, Maruyama K. Cancer Therapy with Nanotechnology-Based Drug Delivery Systems: Applications and Challenges of Liposome Technologies for Advanced Cancer Therapy. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2016. [DOI: 10.1007/978-1-4939-3121-7_23] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
48
|
Suzuki R, Klibanov AL. Co-administration of Microbubbles and Drugs in Ultrasound-Assisted Drug Delivery: Comparison with Drug-Carrying Particles. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 880:205-20. [PMID: 26486340 DOI: 10.1007/978-3-319-22536-4_12] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There are two alternative approaches to ultrasound-assisted drug delivery. First, the drug can be entrapped into or attached onto the ultrasound-responsive particles and administered in the vasculature, to achieve ultrasound-triggered drug release from the particles and localized tissue deposition in response to ultrasound treatment of the target zone. Second, the drug can be co-administered with the microbubbles or other sonosensitive particles. In this case, the action of ultrasound on the particles (which act as cavitation nuclei) results in the transient improvement of permeability of the physiological barriers, so that the circulating drug can exit the bloodstream and get into the target tissues and cells. We discuss and compare both of these approaches, their characteristic advantages and disadvantages for the specific drug delivery scenarios. Clearly, the system based on the off-label use of the existing approved microbubbles and drugs (or drug carriers) will have a chance of getting to clinical trials faster and with lesser resources spent. However, if a superior curative potential of a sonosensitive drug carrier is proven, and formulation stability problems are addressed properly, this approach may find its way to practical use, especially for nucleic acid delivery scenarios.
Collapse
Affiliation(s)
- Ryo Suzuki
- Cardiovascular Division, Robert M Berne Cardiovascular Research Center, University of Virginia, 801394, Charlottesville, VA, 22908, USA.,Department of Drug and Gene Delivery System, Faculty of Pharma-Sciences, Teikyo University, Tokyo, Japan
| | - Alexander L Klibanov
- Cardiovascular Division, Robert M Berne Cardiovascular Research Center, University of Virginia, 801394, Charlottesville, VA, 22908, USA.
| |
Collapse
|
49
|
Ninomiya K, Yamashita T, Tanabe Y, Imai M, Takahashi K, Shimizu N. Targeted and ultrasound-triggered cancer cell injury using perfluorocarbon emulsion-loaded liposomes endowed with cancer cell-targeting and fusogenic capabilities. ULTRASONICS SONOCHEMISTRY 2016; 28:54-61. [PMID: 26384883 DOI: 10.1016/j.ultsonch.2015.06.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 06/30/2015] [Accepted: 06/30/2015] [Indexed: 06/05/2023]
Abstract
This study investigated the targeting and ultrasound-triggered injury of cancer cells using anticancer drug-free liposomes that contained an emulsion of perfluoropentane (ePFC5) and were co-modified with avidin as a targeting ligand for cancer cells and the hemagglutinating virus of Japan (HVJ) envelope to promote liposome fusion with the cells. These liposomes are designated as ePFC5-loaded avidin/HVJ liposomes. ePFC5-loaded liposomes were sensitized to ultrasound irradiation. Liposomes modified with avidin alone (avidin liposomes) showed binding to MCF-7 human breast cancer cells, and liposomes modified with HVJ envelope alone (HVJ liposomes) were found to fuse with MCF-7 cells. The irradiation of MCF-7 cells with 1 MHz ultrasound (30s, 1.2 W/cm(2), duty ratio 30%) combined with ePFC5-loaded avidin/HVJ liposomes resulted in a decrease in cell viability at 1h after irradiation to 43% of that of controls without ultrasound irradiation or liposomes. The cell viability was lower than that of cells treated with ultrasound irradiation with ePFC5-loaded avidin liposomes or ePFC5-loaded HVJ liposomes. This indicates that co-modification of liposome with avidin and HVJ envelope could enhance ultrasound-induced cell injury in the presence of ePFC5-loaded liposomes.
Collapse
Affiliation(s)
- Kazuaki Ninomiya
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Takahiro Yamashita
- Faculty of Natural System, Institute of Science and Engineering, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yamato Tanabe
- Faculty of Natural System, Institute of Science and Engineering, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Miki Imai
- Faculty of Natural System, Institute of Science and Engineering, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Kenji Takahashi
- Faculty of Natural System, Institute of Science and Engineering, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| | - Nobuaki Shimizu
- Institute of Nature and Environmental Technology, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| |
Collapse
|
50
|
Nguyen T, Tekrony A, Yaehne K, Cramb DT. Designing a better theranostic nanocarrier for cancer applications. Nanomedicine (Lond) 2015; 9:2371-86. [PMID: 25413855 DOI: 10.2217/nnm.14.110] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nanocarriers show incredible potential in theranostic applications as they offer diagnostic capabilities along with the ability to encapsulate and protect drugs from degradation, be functionalized with targeting moieties and be designed with controlled release mechanisms. Most clinically approved nanocarrier drugs are liposomal formulations. As such, considerable research has been directed towards designing liposomal carriers that can release their payloads via exogenous or endogenous triggers. For triggered release to effectively increase drug bioavailability, nanocarriers must first accumulate at the tumor site via the enhanced retention and permeability effect. It has been demonstrated in the chicken embryo chorioallantoic membrane and murine xenografted models that nanoparticle surface charge and geometry, with respect to vascular endothelium fenestration size, drive this accumulation in angiogenic tissue.
Collapse
Affiliation(s)
- Trinh Nguyen
- University of Calgary, Department of Chemistry, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | | | | | | |
Collapse
|