1
|
Wang Y, Ding Q, Ma G, Zhang Z, Wang J, Lu C, Xiang C, Qian K, Zheng J, Shan Y, Zhang P, Cheng Z, Gong P, Zhao Q. Mucus-Penetrable Biomimetic Nanoantibiotics for Pathogen-Induced Pneumonia Treatment. ACS NANO 2024; 18:31349-31359. [PMID: 39485232 DOI: 10.1021/acsnano.4c10837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Bacterial pneumonia has garnered significant attention in the realm of infectious diseases owing to a surge in the incidence of severe infections coupled with the growing scarcity of efficacious therapeutic modalities. Antibiotic treatment is still an irreplaceable method for bacterial pneumonia because of its strong bactericidal activity and good clinical efficacy. However, the mucus layer forming after a bacterial infection in the lungs has been considered as the "Achilles' heels" facing the clinical application of such treatment. Herein, traceable biomimetic nanoantibiotics (BioNanoCFPs) were developed by loading indacenodithieno[3,2-b]thiophene (ITIC) and cefoperazone (CFP) in nanoplatforms coated with natural killer (NK) cell membranes. The BioNanoCFP exhibited excellent demonstrated mucus-penetrating abilities, facilitating their arrival at the infection site. The presence of Toll-like receptors in the NK cell membrane rendered the BioNanoCFP with the capability to recognize pathogen-associated molecular patterns within bacteria, allowing precise targeting of bacterial colonization sites and achieving substantial therapeutic efficacy. Overall, our findings demonstrate the viability and desirability of using NK cell membrane-mediated drug delivery as a promising strategy for precision treatment.
Collapse
Affiliation(s)
- Yue Wang
- Cancer Center, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qihang Ding
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Gongcheng Ma
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhiwei Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jiaqi Wang
- Cancer Center, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Chang Lu
- Cancer Center, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Chunbai Xiang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kun Qian
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jun Zheng
- Cancer Center, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Pengfei Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro Center of Biomedicine and Health, Luohu, Shenzhen 518024, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Ping Gong
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro Center of Biomedicine and Health, Luohu, Shenzhen 518024, China
| | - Qi Zhao
- Cancer Center, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| |
Collapse
|
2
|
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
|
3
|
Thim EA, Fox T, Deering T, Vass LR, Sheybani ND, Kester M, Price RJ. Solid tumor treatment via augmentation of bioactive C6 ceramide levels with thermally ablative focused ultrasound. Drug Deliv Transl Res 2023; 13:3145-3153. [PMID: 37335416 PMCID: PMC11423265 DOI: 10.1007/s13346-023-01377-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2023] [Indexed: 06/21/2023]
Abstract
Sparse scan partial thermal ablation (TA) with focused ultrasound (FUS) may be deployed to treat solid tumors and increase delivery of systemically administered therapeutics. Furthermore, C6-ceramide-loaded nanoliposomes (CNLs), which rely upon the enhanced-permeation and retention (EPR) effect for delivery, have shown promise for treating solid tumors and are being tested in clinical trials. Here, our objective was to determine whether CNLs synergize with TA in the control of 4T1 breast tumors. CNL monotherapy of 4T1 tumors yielded significant intratumoral bioactive C6 accumulation by the EPR effect, but tumor growth was not controlled. TA increased bioactive C6 accumulation by ~ 12.5-fold over the EPR effect. In addition, TA + CNL caused shifts in long-chain to very-long-chain ceramide ratios (i.e., C16/24 and C18/C24) that could potentially contribute to tumor control. Nonetheless, these changes in intratumoral ceramide levels were still insufficient to confer tumor growth control beyond that achieved when combining with TA with control "ghost" nanoliposomes (GNL). While this lack of synergy could be due to increased "pro-tumor" sphingosine-1-phosphate (S1P) levels, this is unlikely because S1P levels exhibited only a moderate and statistically insignificant increase with TA + CNL. In vitro studies showed that 4T1 cells are highly resistant to C6, offering the most likely explanation for the inability of TA to synergize with CNL. Thus, while our results show that sparse scan TA is a powerful approach for markedly enhancing CNL delivery and generating "anti-tumor" shifts in long-chain to very-long-chain ceramide ratios, resistance of the tumor to C6 can still be a rate-limiting factor for some solid tumor types.
Collapse
Affiliation(s)
- E Andrew Thim
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, VA, 22908, USA
| | - Todd Fox
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Tye Deering
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Luke R Vass
- Department of Pathology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Natasha D Sheybani
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, VA, 22908, USA
| | - Mark Kester
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, VA, 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, VA, 22908, USA.
| |
Collapse
|
4
|
Yue Y, Li H, Wang X, Zhang B, Li Y, Liu Y, Ma X, Liu G, Zhao X, Shi J. Intelligent Responsive Nanoparticles with Multilevel Triggered Drug Penetration for Tumor Photochemotherapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:44175-44185. [PMID: 37669460 DOI: 10.1021/acsami.3c06674] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Nanomedicines have contradictory size requirements to overcome systemic barriers and penetrate the tumor extracellular matrix (ECM). Larger-sized nanoparticles (50-200 nm) exhibit prolonged blood circulation half-life and improved tumor enrichment, while small-sized nanoparticles (4-20 nm) easily penetrate deep tumor tissues. Therefore, the development of intelligent responsive nanomedicine systems can not only increase nanodrug tumor accumulation but also improve their penetration into the ECM. Herein, we propose an intelligent responsive nanoparticle triggered by near-infrared light (NIR). The nanoparticle was constructed by a temperature-sensitive liposome (TSL) encapsulating ultrasmall melanin nanoparticles (MNPs) loaded with doxorubicin (MNP/doxorubicin (DOX)@TSL). When exposed to NIR irradiation, the tailor-made nanoparticles not only effectively ablated the tumor cells around blood vessels but also destroyed the structural integrity and released loaded ultrasmall MNP/DOX (<10 nm) to promote deep tumor penetration and enhance interior tumor cell killing. This NIR-triggered intelligent nanoparticle successfully integrated photothermal therapy (PTT) for perivascular tumor cells and chemotherapy for deep tumor cell inhibition. The in vivo results showed remarkable tumor regression in 4T1 breast tumor-bearing mice by 74.2%. This controllable size switchable nanosystem with efficient tumor accumulation and penetration has shown great potential in improving synergistic antitumor effects of photochemotherapy.
Collapse
Affiliation(s)
- Yale Yue
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Hejia Li
- Third Clinical Division, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Xinwei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Baohua Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Yiting Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaotu Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
5
|
Duncan B, Al-Kassas R, Zhang G, Hughes D, Qiu Y. Ultrasound-Mediated Ocular Drug Delivery: From Physics and Instrumentation to Future Directions. MICROMACHINES 2023; 14:1575. [PMID: 37630111 PMCID: PMC10456754 DOI: 10.3390/mi14081575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023]
Abstract
Drug delivery to the anterior and posterior segments of the eye is impeded by anatomical and physiological barriers. Increasingly, the bioeffects produced by ultrasound are being proven effective for mitigating the impact of these barriers on ocular drug delivery, though there does not appear to be a consensus on the most appropriate system configuration and operating parameters for this application. In this review, the fundamental aspects of ultrasound physics most pertinent to drug delivery are presented; the primary phenomena responsible for increased drug delivery efficacy under ultrasound sonication are discussed; an overview of common ocular drug administration routes and the associated ocular barriers is also given before reviewing the current state of the art of ultrasound-mediated ocular drug delivery and its potential future directions.
Collapse
Affiliation(s)
- Blair Duncan
- School of Engineering, Faculty of Engineering & Technology, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, UK
| | - Raida Al-Kassas
- School of Pharmacy & Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, UK
| | - Guangming Zhang
- School of Engineering, Faculty of Engineering & Technology, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, UK
| | - Dave Hughes
- Novosound Ltd., Biocity, BoNess Road, Newhouse, Glasgow ML1 5UH, UK
| | - Yongqiang Qiu
- School of Engineering, Faculty of Engineering & Technology, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, UK
| |
Collapse
|
6
|
Rafik ST, Vaidya JS, MacRobert AJ, Yaghini E. Organic Nanodelivery Systems as a New Platform in the Management of Breast Cancer: A Comprehensive Review from Preclinical to Clinical Studies. J Clin Med 2023; 12:jcm12072648. [PMID: 37048731 PMCID: PMC10095028 DOI: 10.3390/jcm12072648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/05/2023] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
Breast cancer accounts for approximately 25% of cancer cases and 16.5% of cancer deaths in women, and the World Health Organization predicts that the number of new cases will increase by almost 70% over the next two decades, mainly due to an ageing population. Effective diagnostic and treatment strategies are, therefore, urgently required for improving cure rates among patients since current therapeutic modalities have many limitations and side effects. Nanomedicine is evolving as a promising approach for cancer management, including breast cancer, and various types of organic and inorganic nanomaterials have been investigated for their role in breast cancer diagnosis and treatment. Following an overview on breast cancer characteristics and pathogenesis and challenges of the current treatment strategies, the therapeutic potential of biocompatible organic-based nanoparticles such as liposomes and polymeric micelles that have been tested in breast cancer models are reviewed. The efficacies of different drug delivery and targeting strategies are documented, ranging from synthetic to cell-derived nanoformulations together with a summary of the interaction of nanoparticles with externally applied energy such as radiotherapy. The clinical translation of nanoformulations for breast cancer treatment is summarized including those undergoing clinical trials.
Collapse
Affiliation(s)
- Salma T. Rafik
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London (UCL), London W1W 7TY, UK
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria 21516, Egypt
| | - Jayant S. Vaidya
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London (UCL), London W1W 7TY, UK
| | - Alexander J. MacRobert
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London (UCL), London W1W 7TY, UK
| | - Elnaz Yaghini
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London (UCL), London W1W 7TY, UK
| |
Collapse
|
7
|
Regenold M, Wang X, Kaneko K, Bannigan P, Allen C. Harnessing immunotherapy to enhance the systemic anti-tumor effects of thermosensitive liposomes. Drug Deliv Transl Res 2023; 13:1059-1073. [PMID: 36577832 DOI: 10.1007/s13346-022-01272-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2022] [Indexed: 12/29/2022]
Abstract
Chemotherapy plays an important role in debulking tumors in advance of surgery and/or radiotherapy, tackling residual disease, and treating metastatic disease. In recent years many promising advanced drug delivery strategies have emerged that offer more targeted delivery approaches to chemotherapy treatment. For example, thermosensitive liposome-mediated drug delivery in combination with localized mild hyperthermia can increase local drug concentrations resulting in a reduction in systemic toxicity and an improvement in local disease control. However, the majority of solid tumor-associated deaths are due to metastatic spread. A therapeutic approach focused on a localized target area harbors the risk of overlooking and undertreating potential metastatic spread. Previous studies reported systemic, albeit limited, anti-tumor effects following treatment with thermosensitive liposomal chemotherapy and localized mild hyperthermia. This work explores the systemic treatment capabilities of a thermosensitive liposome formulation of the vinca alkaloid vinorelbine in combination with mild hyperthermia in an immunocompetent murine model of rhabdomyosarcoma. This treatment approach was found to be highly effective at heated, primary tumor sites. However, it demonstrated limited anti-tumor effects in secondary, distant tumors. As a result, the addition of immune checkpoint inhibition therapy was pursued to further enhance the systemic anti-tumor effect of this treatment approach. Once combined with immune checkpoint inhibition therapy, a significant improvement in systemic treatment capability was achieved. We believe this is one of the first studies to demonstrate that a triple combination of thermosensitive liposomes, localized mild hyperthermia, and immune checkpoint inhibition therapy can enhance the systemic treatment capabilities of thermosensitive liposomes.
Collapse
Affiliation(s)
- Maximilian Regenold
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada
| | - Xuehan Wang
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada
| | - Kan Kaneko
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada
| | - Pauric Bannigan
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada.
| |
Collapse
|
8
|
Thim EA, Fox T, Deering T, Vass LR, Sheybani ND, Kester M, Price RJ. Solid Tumor Treatment via Augmentation of Bioactive C6 Ceramide Levels with Thermally Ablative Focused Ultrasound. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.23.532394. [PMID: 36993445 PMCID: PMC10055354 DOI: 10.1101/2023.03.23.532394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Sparse scan partial thermal ablation (TA) with focused ultrasound (FUS) may be deployed to treat solid tumors and increase delivery of systemically administered therapeutics. Further, C6-ceramide-loaded nanoliposomes (CNLs), which rely upon the enhanced permeation and retention (EPR) effect for delivery, have shown promise for treating solid tumors and are being tested in clinical trials. Here, our objective was to determine whether CNLs synergize with TA in the control of 4T1 breast tumors. CNL-monotherapy of 4T1 tumors yielded significant intratumoral bioactive C6 accumulation by the EPR effect, but tumor growth was not controlled. TA increased bioactive C6 accumulation by ∼12.5-fold over the EPR effect. In addition, TA+CNL caused shifts in long-chain to very-long-chain ceramide ratios (i.e., C16/24 and C18/C24) that could potentially contribute to tumor control. Nonetheless, these changes in intratumoral ceramide levels were still insufficient to confer tumor growth control beyond that achieved when combining with TA with control "ghost" nanoliposomes (GNL). While this lack of synergy could be due to increased "pro-tumor" sphingosine-1-phosphate (S1P) levels, this is unlikely because S1P levels exhibited only a moderate and statistically insignificant increase with TA+CNL. In vitro studies showed that 4T1 cells are highly resistant to C6, offering the most likely explanation for the inability of TA to synergize with CNL. Thus, while our results show that sparse scan TA is a powerful approach for markedly enhancing CNL delivery and generating "anti-tumor" shifts in long-chain to very-long-chain ceramide ratios, resistance of the tumor to C6 can still be a rate-limiting factor for some solid tumor types.
Collapse
Affiliation(s)
- E. Andrew Thim
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| | - Todd Fox
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
| | - Tye Deering
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
| | - Luke R. Vass
- Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Natasha D. Sheybani
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| | - Mark Kester
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
| | - Richard J. Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
9
|
Kumar D, Moghiseh M, Chitcholtan K, Mutreja I, Lowe C, Kaushik A, Butler A, Sykes P, Anderson N, Raja A. LHRH conjugated gold nanoparticles assisted efficient ovarian cancer targeting evaluated via spectral photon-counting CT imaging: a proof-of-concept research. J Mater Chem B 2023; 11:1916-1928. [PMID: 36744575 DOI: 10.1039/d2tb02416k] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Emerging multifunctional nanoparticulate formulations take advantage of nano-meter scale size and surface chemistry to work as a therapeutic delivery agent and a diagnostic tool for non-invasive real-time monitoring using imaging technologies. Here, we evaluate the selective uptake of 18 nm and 80 nm sized gold nanoparticles (AuNPs) by SKOV3 (4 times higher) ovarian cancer (OC) cells (compared to OVCAR5) in vitro, quantified by inductively coupled plasma (ICP) and MARS spectral photon-counting CT imaging (MARS SPCCT). Based on in vitro analysis, pristine AuNPs (18 nm) and surface modified AuNPs (18 nm) were chosen as a contrast agent for MARS SPCCT. The chemical analysis by FTIR spectroscopy confirmed the luteinizing hormone-releasing hormone (LHRH) conjugation to the AuNPs surface. For the first time, LHRH conjugated AuNPs were used for in vitro and selective in vivo OC targeting. The ICP-MS analysis confirmed preferential uptake of LHRH modified AuNPs by organs residing in the abdominal cavity with OC nodules (pancreas: 0.46 ng mg-1, mesentery: 0.89 ng mg-1, ovary: 1.43 ng mg-1, and abdominal wall: 2.12 ng mg-1) whereas the MARS SPCCT analysis suggested scattered accumulation of metal around the abdominal cavity. Therefore, the study showed the exciting potential of LHRH conjugated AuNPs to target ovarian cancer and also as a potential contrast agent for novel SPCCT imaging technology.
Collapse
Affiliation(s)
- Dhiraj Kumar
- Division of Pediatrics Dentistry, School of Dentistry, University of Minnesota, 515 Delaware St SE, Minneapolis, Minnesota, 55455, USA. .,Department of Obstetrics and Gynaecology, Christchurch Women Hospital, University of Otago Christchurch, 2 Riccarton Ave, School of Medicine, Christchurch, New Zealand
| | - Mahdieh Moghiseh
- Department of Radiology, University of Otago Christchurch, 2 Riccarton Ave, School of Medicine, Christchurch, New Zealand.,MARS Bioimaging Limited, Christchurch, New Zealand
| | - Kenny Chitcholtan
- Department of Obstetrics and Gynaecology, Christchurch Women Hospital, University of Otago Christchurch, 2 Riccarton Ave, School of Medicine, Christchurch, New Zealand
| | - Isha Mutreja
- Minnesota Dental Research Center for Biomaterials and Biomechanics (MDRCBB), School of Dentistry, University of Minnesota, 515 Delaware St SE, Minneapolis, Minnesota, 55455, USA
| | - Chiara Lowe
- Department of Radiology, University of Otago Christchurch, 2 Riccarton Ave, School of Medicine, Christchurch, New Zealand.,MARS Bioimaging Limited, Christchurch, New Zealand
| | - Ajeet Kaushik
- NanoBiotech Laboratory, Department of Environmental Engineering, Florida Polytechnic University, FL, 33805, USA
| | - Anthony Butler
- Department of Radiology, University of Otago Christchurch, 2 Riccarton Ave, School of Medicine, Christchurch, New Zealand.,MARS Bioimaging Limited, Christchurch, New Zealand
| | - Peter Sykes
- Department of Obstetrics and Gynaecology, Christchurch Women Hospital, University of Otago Christchurch, 2 Riccarton Ave, School of Medicine, Christchurch, New Zealand
| | - Nigel Anderson
- Department of Radiology, University of Otago Christchurch, 2 Riccarton Ave, School of Medicine, Christchurch, New Zealand
| | - Aamir Raja
- Department of Radiology, University of Otago Christchurch, 2 Riccarton Ave, School of Medicine, Christchurch, New Zealand.,Department of Physics, Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
10
|
Moradi Kashkooli F, Jakhmola A, Hornsby TK, Tavakkoli JJ, Kolios MC. Ultrasound-mediated nano drug delivery for treating cancer: Fundamental physics to future directions. J Control Release 2023; 355:552-578. [PMID: 36773959 DOI: 10.1016/j.jconrel.2023.02.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/13/2023]
Abstract
The application of biocompatible nanocarriers in medicine has provided several benefits over conventional treatment methods. However, achieving high treatment efficacy and deep penetration of nanocarriers in tumor tissue is still challenging. To address this, stimuli-responsive nano-sized drug delivery systems (DDSs) are an active area of investigation in delivering anticancer drugs. While ultrasound is mainly used for diagnostic purposes, it can also be applied to affect cellular function and the delivery/release of anticancer drugs. Therapeutic ultrasound (TUS) has shown potential as both a stand-alone anticancer treatment and a method to induce targeted drug release from nanocarrier systems. TUS approaches have been used to overcome various physiological obstacles, including endothelial barriers, the tumor microenvironment (TME), and immunological hurdles. Combining nanomedicine and ultrasound as a smart DDS can increase in situ drug delivery and improve access to impermeable tissues. Furthermore, smart DDSs can perform targeted drug release in response to distinctive TMEs, external triggers, or dual/multi-stimulus. This results in enhanced treatment efficacy and reduced damage to surrounding healthy tissue or organs at risk. Integrating DDSs and ultrasound is still in its early stages. More research and clinical trials are required to fully understand ultrasound's underlying physical mechanisms and interactions with various types of nanocarriers and different types of cells and tissues. In the present review, ultrasound-mediated nano-sized DDS, specifically focused on cancer treatment, is presented and discussed. Ultrasound interaction with nanoparticles (NPs), drug release mechanisms, and various types of ultrasound-sensitive NPs are examined. Additionally, in vitro, in vivo, and clinical applications of TUS are reviewed in light of the critical challenges that need to be considered to advance TUS toward an efficient, secure, straightforward, and accessible cancer treatment. This study also presents effective TUS parameters and safety considerations for this treatment modality and gives recommendations about system design and operation. Finally, future perspectives are considered, and different TUS approaches are examined and discussed in detail. This review investigates drug release and delivery through ultrasound-mediated nano-sized cancer treatment, both pre-clinically and clinically.
Collapse
Affiliation(s)
| | - Anshuman Jakhmola
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Tyler K Hornsby
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Jahangir Jahan Tavakkoli
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
11
|
Haemmerich D, Ramajayam KK, Newton DA. Review of the Delivery Kinetics of Thermosensitive Liposomes. Cancers (Basel) 2023; 15:cancers15020398. [PMID: 36672347 PMCID: PMC9856714 DOI: 10.3390/cancers15020398] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/10/2023] Open
Abstract
Thermosensitive liposomes (TSL) are triggered nanoparticles that release the encapsulated drug in response to hyperthermia. Combined with localized hyperthermia, TSL enabled loco-regional drug delivery to tumors with reduced systemic toxicities. More recent TSL formulations are based on intravascular triggered release, where drug release occurs within the microvasculature. Thus, this delivery strategy does not require enhanced permeability and retention (EPR). Compared to traditional nanoparticle drug delivery systems based on EPR with passive or active tumor targeting (typically <5%ID/g tumor), TSL can achieve superior tumor drug uptake (>10%ID/g tumor). Numerous TSL formulations have been combined with various drugs and hyperthermia devices in preclinical and clinical studies over the last four decades. Here, we review how the properties of TSL dictate delivery and discuss the advantages of rapid drug release from TSL. We show the benefits of selecting a drug with rapid extraction by tissue, and with quick cellular uptake. Furthermore, the optimal characteristics of hyperthermia devices are reviewed, and impact of tumor biology and cancer cell characteristics are discussed. Thus, this review provides guidelines on how to improve drug delivery with TSL by optimizing the combination of TSL, drug, and hyperthermia method. Many of the concepts discussed are applicable to a variety of other triggered drug delivery systems.
Collapse
Affiliation(s)
- Dieter Haemmerich
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
- Correspondence:
| | - Krishna K. Ramajayam
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Danforth A. Newton
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
12
|
Li C, Li Y, Li G, Wu S. Functional Nanoparticles for Enhanced Cancer Therapy. Pharmaceutics 2022; 14:pharmaceutics14081682. [PMID: 36015307 PMCID: PMC9412412 DOI: 10.3390/pharmaceutics14081682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/03/2022] [Accepted: 08/10/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer is the leading cause of death in people worldwide. The conventional therapeutic approach is mainly based on chemotherapy, which has a series of side effects. Compared with traditional chemotherapy drugs, nanoparticle-based delivery of anti-cancer drugs possesses a few attractive features. The application of nanotechnology in an interdisciplinary manner in the biomedical field has led to functional nanoparticles achieving much progress in cancer therapy. Nanoparticles have been involved in the diagnosis and targeted and personalized treatment of cancer. For example, different nano-drug strategies, including endogenous and exogenous stimuli-responsive, surface conjugation, and macromolecular encapsulation for nano-drug systems, have successfully prevented tumor procession. The future for functional nanoparticles is bright and promising due to the fast development of nanotechnology. However, there are still some challenges and limitations that need to be considered. Based on the above contents, the present article analyzes the progress in developing functional nanoparticles in cancer therapy. Research gaps and promising strategies for the clinical application are discussed.
Collapse
Affiliation(s)
- Chenchen Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China
| | - Yuqing Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China
| | - Guangzhi Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China
- Correspondence: (G.L.); (S.W.)
| | - Song Wu
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China
- Correspondence: (G.L.); (S.W.)
| |
Collapse
|
13
|
Wang Z, Li J, Lin G, He Z, Wang Y. Metal complex-based liposomes: Applications and prospects in cancer diagnostics and therapeutics. J Control Release 2022; 348:1066-1088. [PMID: 35718211 DOI: 10.1016/j.jconrel.2022.06.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 06/09/2022] [Indexed: 12/17/2022]
Abstract
Metal complexes are of increasing interest as pharmaceutical agents in cancer diagnostics and therapeutics, while some of them suffer from issues such as limited water solubility and severe systemic toxicity. These drawbacks severely hampered their efficacy and clinical applications. Liposomes hold promise as delivery vehicles for constructing metal complex-based liposomes to maximize the therapeutic efficacy and minimize the side effects of metal complexes. This review provides an overview on the latest advances of metal complex-based liposomal delivery systems. First, the development of metal complex-mediated liposomal encapsulation is briefly introduced. Next, applications of metal complex-based liposomes in a variety of fields are overviewed, where drug delivery, cancer imaging (single photon emission computed tomography (SPECT), positron emission tomography (PET), and magnetic resonance imaging (MRI)), and cancer therapy (chemotherapy, phototherapy, and radiotherapy) were involved. Moreover, the potential toxicity, action of toxic mechanisms, immunological effects of metal complexes as well as the advantages of metal complex-liposomes in this content are also discussed. In the end, the future expectations and challenges of metal complex-based liposomes in clinical cancer therapy are tentatively proposed.
Collapse
Affiliation(s)
- Zhaomeng Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Jinbo Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Guimei Lin
- School of Pharmacy, Shandong University, Jinan 250000, PR China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
14
|
He X, Chen S, Mao X. Utilization of metal or non-metal-based functional materials as efficient composites in cancer therapies. RSC Adv 2022; 12:6540-6551. [PMID: 35424648 PMCID: PMC8982229 DOI: 10.1039/d1ra08335j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/30/2022] [Indexed: 12/03/2022] Open
Abstract
There has been great progress in cancer treatment through traditional approaches, even though some of them are still trapped in relative complications such as certain side effects and prospective chances of full recovery. As a conventional method, the immunotherapy approach is regarded as an effective approach to cure cancer. It is mainly promoted by immune checkpoint blocking and adoptive cell therapy, which can utilize the human immune system to attack tumor cells and make them necrose completely or stop proliferating cancer cells. Currently however, immunotherapy shows limited success due to the limitation of real applicable cases of targeted tumor environments and immune systems. Considering the urgent need to construct suitable strategies towards cancer therapy, metallic materials can be used as delivery systems for immunotherapeutic agents in the human body. Metallic materials exhibit a high degree of specificity, effectiveness, diagnostic ability, imaging ability and therapeutic effects with different biomolecules or polymers, which is an effective option for cancer treatment. In addition, these modified metallic materials contain immune-modulators, which can activate immune cells to regulate tumor microenvironments and enhance anti-cancer immunity. Additionally, they can be used as adjuvants with immunomodulatory activities, or as carriers for molecular transport to specific targets, which results in the loading of specific ligands to facilitate specific uptake. Here, we provide an overview of the different types of metallic materials used as efficient composites in cancer immunotherapy. We elaborate on the advancements using metallic materials with functional agents as effective composites in synergistic cancer treatment. Some nonmetallic functional composites also appear as a common phenomenon. Ascribed to the design of the composites themselves, the materials' surface structural characteristics are introduced as the drug-loading substrate. The physical and chemical properties of the functional materials emphasize that further research is required to fully characterize their mechanism, showing appropriate relevance for material toxicology and biomedical applications.
Collapse
Affiliation(s)
- Xiaoxiao He
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
| | - Shiyue Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
| | - Xiang Mao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
- Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 P. R. China
| |
Collapse
|
15
|
Yu Y, Wang B, Sun M, Zhang Y, Hou L, Wang S, Chen T, Yang F, Ma Z. Lysosomal activable Vorinostat carrier-prodrug self-assembling with BPQDs enables photothermal oncotherapy to reverse tumor thermotolerance and metastasis. Int J Pharm 2022; 617:121580. [PMID: 35202725 DOI: 10.1016/j.ijpharm.2022.121580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/27/2022] [Accepted: 02/10/2022] [Indexed: 12/16/2022]
Abstract
Photothermal therapy (PTT) is becoming increasing prevalent in clinic for eradicating the primary tumor and improving cancer patients' compliance. However, photothermal resistance and distal metastasis still haunt the tumor treatment with PTT. Herein, on the basis that histone deacetylase acetylase inhibitor (HDACis) could activate the expression of anti-tumor gene and accelerate the differentiation and apoptosis of tumor cells, we propose that HDACis supplementing PTT could overcome those obstacles with appropriate drug-controlled release strategy. Thus, we fabricated a nano-complex of lysosomal activable vorinostat (SAHA) carrier-prodrug encapsulating black phosphorus quantum dots (BPQDs@PPS) to counter those challenges in PTT. With spherical morphology and favorable bio-safety, BPQDs@PPS could release BPQDs and Vorinostat spontaneously in lysosome, not only effectively inhibiting tumor growth, but also reversing tumor thermotolerance and metastasis within a PTT procedure. Especially, both western blot and immunofluorescence analysis validate that Vorinostat enables PTT to reverse tumor thermotolerance and distal metastasis by down-regulation of HSP70 and up-regulation of H3. Therefore, this research not only reveals the mechanism how HDACis supplement PTT in reversing tumor thermotolerance and metastasis, but also provides a promising prospect to upgrade clinical photothermal therapy.
Collapse
Affiliation(s)
- Yingjie Yu
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Bingkai Wang
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Miao Sun
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Yunchang Zhang
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Lei Hou
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Sizhen Wang
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Tianheng Chen
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Feng Yang
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China
| | - Zhiqiang Ma
- School of Pharmacy, Naval Medical University, Shanghai, People's Republic of China.
| |
Collapse
|
16
|
Sebeke L, Gómez JDC, Heijman E, Rademann P, Maul AC, Ekdawi S, Vlachakis S, Toker D, Mink BL, Schubert-Quecke C, Yeo SY, Schmidt P, Lucas C, Brodesser S, Hossann M, Lindner LH, Grüll H. Hyperthermia-induced doxorubicin delivery from thermosensitive liposomes via MR-HIFU in a pig model. J Control Release 2022; 343:798-812. [DOI: 10.1016/j.jconrel.2022.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 12/17/2022]
|
17
|
D'Angelo NA, Noronha MA, Câmara MCC, Kurnik IS, Feng C, Araujo VHS, Santos JHPM, Feitosa V, Molino JVD, Rangel-Yagui CO, Chorilli M, Ho EA, Lopes AM. Doxorubicin nanoformulations on therapy against cancer: An overview from the last 10 years. BIOMATERIALS ADVANCES 2022; 133:112623. [PMID: 35525766 DOI: 10.1016/j.msec.2021.112623] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 12/10/2021] [Accepted: 12/16/2021] [Indexed: 06/14/2023]
Abstract
Doxorubicin (DOX) is a natural antibiotic with antineoplastic activity. It has been used for over 40 years and remains one of the most used drugs in chemotherapy for a variety of cancers. However, cardiotoxicity limits its use for long periods. To overcome this limitation, encapsulation in smart drug delivery systems (DDS) brings advantages in comparison with free drug administration (i.e., conventional anticancer drug therapy). In this review, we present the most relevant nanostructures used for DOX encapsulation over the last 10 years, such as liposomes, micelles and polymeric vesicles (i.e., polymersomes), micro/nanoemulsions, different types of polymeric nanoparticles and hydrogel nanoparticles, as well as novel approaches for DOX encapsulation. The studies highlighted here show these nanoformulations achieved higher solubility, improved tumor cytotoxicity, prolonged DOX release, as well as reduced side effects, among other interesting advantages.
Collapse
Affiliation(s)
- Natália A D'Angelo
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Mariana A Noronha
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Mayra C C Câmara
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Isabelle S Kurnik
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Chuying Feng
- Laboratory for Drug Delivery and Biomaterials, School of Pharmacy, University of Waterloo, 10 Victoria St S, Kitchener, Ontario N2G1C5, Canada
| | - Victor H S Araujo
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - João H P M Santos
- Department of Biochemical and Pharmaceutical Technology, University of São Paulo (USP), São Paulo, Brazil; Micromanufacturing Laboratory, Center for Bionanomanufacturing, Institute for Technological Research (IPT), São Paulo, Brazil
| | - Valker Feitosa
- Micromanufacturing Laboratory, Center for Bionanomanufacturing, Institute for Technological Research (IPT), São Paulo, Brazil
| | | | - Carlota O Rangel-Yagui
- Department of Biochemical and Pharmaceutical Technology, University of São Paulo (USP), São Paulo, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Emmanuel A Ho
- Laboratory for Drug Delivery and Biomaterials, School of Pharmacy, University of Waterloo, 10 Victoria St S, Kitchener, Ontario N2G1C5, Canada
| | - André M Lopes
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas, Brazil.
| |
Collapse
|
18
|
Fodor M, Fodor L, Bota O. The role of nanomaterials and nanostructured surfaces for improvement of biomaterial peculiarities in vascular surgery: a review. PARTICULATE SCIENCE AND TECHNOLOGY 2021. [DOI: 10.1080/02726351.2021.1871692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Marius Fodor
- Department of Vascular Surgery, First Surgical Clinic, Emergency District Hospital, Cluj-Napoca, Romania, Cluj-Napoca, Romania
| | - Lucian Fodor
- Department of Plastic Surgery, First Surgical Clinic, Emergency District Hospital, Cluj-Napoca, Romania, Cluj-Napoca, Romania
| | - Olimpiu Bota
- University Center of Orthopaedic, Trauma and Plastic Surgery, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| |
Collapse
|
19
|
Popov AS, Efimova AA, Kazantsev AV, Erzunov DA, Lukashev NV, Grozdova ID, Melik-Nubarov NS, Yaroslavov AA. pH-Sensitive liposomes with embedded ampholytic derivatives of cholan-24-oic acid. MENDELEEV COMMUNICATIONS 2021. [DOI: 10.1016/j.mencom.2021.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
20
|
Priester MI, Curto S, van Rhoon GC, ten Hagen TLM. External Basic Hyperthermia Devices for Preclinical Studies in Small Animals. Cancers (Basel) 2021; 13:cancers13184628. [PMID: 34572855 PMCID: PMC8470307 DOI: 10.3390/cancers13184628] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The application of mild hyperthermia can be beneficial for solid tumor treatment by induction of sublethal effects on a tissue- and cellular level. When designing a hyperthermia experiment, several factors should be taken into consideration. In this review, multiple elementary hyperthermia devices are described in detail to aid standardization of treatment design. Abstract Preclinical studies have shown that application of mild hyperthermia (40–43 °C) is a promising adjuvant to solid tumor treatment. To improve preclinical testing, enhance reproducibility, and allow comparison of the obtained results, it is crucial to have standardization of the available methods. Reproducibility of methods in and between research groups on the same techniques is crucial to have a better prediction of the clinical outcome and to improve new treatment strategies (for instance with heat-sensitive nanoparticles). Here we provide a preclinically oriented review on the use and applicability of basic hyperthermia systems available for solid tumor thermal treatment in small animals. The complexity of these techniques ranges from a simple, low-cost water bath approach, irradiation with light or lasers, to advanced ultrasound and capacitive heating devices.
Collapse
Affiliation(s)
- Marjolein I. Priester
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.C.); (G.C.v.R.)
| | - Sergio Curto
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.C.); (G.C.v.R.)
| | - Gerard C. van Rhoon
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.C.); (G.C.v.R.)
| | - Timo L. M. ten Hagen
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Correspondence:
| |
Collapse
|
21
|
Drug Delivery by Ultrasound-Responsive Nanocarriers for Cancer Treatment. Pharmaceutics 2021; 13:pharmaceutics13081135. [PMID: 34452096 PMCID: PMC8397943 DOI: 10.3390/pharmaceutics13081135] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022] Open
Abstract
Conventional cancer chemotherapies often exhibit insufficient therapeutic outcomes and dose-limiting toxicity. Therefore, there is a need for novel therapeutics and formulations with higher efficacy, improved safety, and more favorable toxicological profiles. This has promoted the development of nanomedicines, including systems for drug delivery, but also for imaging and diagnostics. Nanoparticles loaded with drugs can be designed to overcome several biological barriers to improving efficiency and reducing toxicity. In addition, stimuli-responsive nanocarriers are able to release their payload on demand at the tumor tissue site, preventing premature drug loss. This review focuses on ultrasound-triggered drug delivery by nanocarriers as a versatile, cost-efficient, non-invasive technique for improving tissue specificity and tissue penetration, and for achieving high drug concentrations at their intended site of action. It highlights aspects relevant for ultrasound-mediated drug delivery, including ultrasound parameters and resulting biological effects. Then, concepts in ultrasound-mediated drug delivery are introduced and a comprehensive overview of several types of nanoparticles used for this purpose is given. This includes an in-depth compilation of the literature on the various in vivo ultrasound-responsive drug delivery systems. Finally, toxicological and safety considerations regarding ultrasound-mediated drug delivery with nanocarriers are discussed.
Collapse
|
22
|
Yang B, Song BP, Shankar S, Guller A, Deng W. Recent advances in liposome formulations for breast cancer therapeutics. Cell Mol Life Sci 2021; 78:5225-5243. [PMID: 33974093 PMCID: PMC11071878 DOI: 10.1007/s00018-021-03850-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/31/2021] [Accepted: 04/30/2021] [Indexed: 12/18/2022]
Abstract
Among many nanoparticle-based delivery platforms, liposomes have been particularly successful with many formulations passed into clinical applications. They are well-established and effective gene and/or drug delivery systems, widely used in cancer therapy including breast cancer. In this review we discuss liposome design with the targeting feature and triggering functions. We also summarise the recent progress (since 2014) in liposome-based therapeutics for breast cancer including chemotherapy and gene therapy. We finally identify some challenges on the liposome technology development for the future clinical translation.
Collapse
Affiliation(s)
- Biyao Yang
- ARC Centre of Excellence for Nanoscale Biophotonics, the Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Bo-Ping Song
- ARC Centre of Excellence for Nanoscale Biophotonics, the Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- School of Mechatronic Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Shaina Shankar
- ARC Centre of Excellence for Nanoscale Biophotonics, the Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Anna Guller
- ARC Centre of Excellence for Nanoscale Biophotonics, the Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia
| | - Wei Deng
- ARC Centre of Excellence for Nanoscale Biophotonics, the Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
23
|
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: 60] [Impact Index Per Article: 15.0] [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
|
24
|
Zhang H, Tang WL, Kheirolomoom A, Fite BZ, Wu B, Lau K, Baikoghli M, Raie MN, Tumbale SK, Foiret J, Ingham ES, Mahakian LM, Tam SM, Cheng RH, Borowsky AD, Ferrara KW. Development of thermosensitive resiquimod-loaded liposomes for enhanced cancer immunotherapy. J Control Release 2021; 330:1080-1094. [PMID: 33189786 PMCID: PMC7906914 DOI: 10.1016/j.jconrel.2020.11.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 11/01/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022]
Abstract
Resiquimod (R848) is a toll-like receptor 7 and 8 (TLR7/8) agonist with potent antitumor and immunostimulatory activity. However, systemic delivery of R848 is poorly tolerated because of its poor solubility in water and systemic immune activation. In order to address these limitations, we developed an intravenously-injectable formulation with R848 using thermosensitive liposomes (TSLs) as a delivery vehicle. R848 was remotely loaded into TSLs composed of DPPC: DSPC: DSPE-PEG2K (85:10:5, mol%) with 100 mM FeSO4 as the trapping agent inside. The final R848 to lipid ratio of the optimized R848-loaded TSLs (R848-TSLs) was 0.09 (w/w), 10-fold higher than the previously-reported values. R848-TSLs released 80% of R848 within 5 min at 42 °C. These TSLs were then combined with αPD-1, an immune checkpoint inhibitor, and ultrasound-mediated hyperthermia in a neu deletion (NDL) mouse mammary carcinoma model (Her2+, ER/PR negative). Combined with αPD-1, local injection of R848-TSLs showed superior efficacy with complete NDL tumor regression in both treated and abscopal sites achieved in 8 of 11 tumor bearing mice over 100 days. Immunohistochemistry confirmed enhanced CD8+ T cell infiltration and accumulation by R848-TSLs. Systemic delivery of R848-TSLs, combined with local hyperthermia and αPD-1, inhibited tumor growth and extended median survival from 28 days (non-treatment control) to 94 days. Upon re-challenge with reinjection of tumor cells, none of the previously cured mice developed tumors, as compared with 100% of age-matched control mice. The dose of R848 (10 μg for intra-tumoral injection or 6 mg/kg for intravenous injection delivered up to 4 times) was well-tolerated without weight loss or organ hypertrophy. In summary, we developed R848-TSLs that can be administered locally or systematically, resulting in tumor regression and enhanced survival when combined with αPD-1 in mouse models of breast cancer.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Wei-Lun Tang
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Azadeh Kheirolomoom
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Brett Z Fite
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Bo Wu
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Kenneth Lau
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Mo Baikoghli
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - Marina Nura Raie
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Spencer K Tumbale
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Josquin Foiret
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Lisa M Mahakian
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Sarah M Tam
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - R Holland Cheng
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | | | - Katherine W Ferrara
- Molecular Imaging Program, Department of Radiology, Stanford University, 3165 Porter Drive, Palo Alto, CA 94304, USA.
| |
Collapse
|
25
|
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: 30] [Impact Index Per Article: 6.0] [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
|
26
|
Remote loading paclitaxel-doxorubicin prodrug into liposomes for cancer combination therapy. Acta Pharm Sin B 2020; 10:1730-1740. [PMID: 33088692 PMCID: PMC7564015 DOI: 10.1016/j.apsb.2020.04.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/20/2022] Open
Abstract
The combination of paclitaxel (PTX) and doxorubicin (DOX) has been widely used in the clinic. However, it remains unsatisfied due to the generation of severe toxicity. Previously, we have successfully synthesized a prodrug PTX-S-DOX (PSD). The prodrug displayed comparable in vitro cytotoxicity compared with the mixture of free PTX and DOX. Thus, we speculated that it could be promising to improve the anti-cancer effect and reduce adverse effects by improving the pharmacokinetics behavior of PSD and enhancing tumor accumulation. Due to the fact that copper ions (Cu2+) could coordinate with the anthracene nucleus of DOX, we speculate that the prodrug PSD could be actively loaded into liposomes by Cu2+ gradient. Hence, we designed a remote loading liposomal formulation of PSD (PSD LPs) for combination chemotherapy. The prepared PSD LPs displayed extended blood circulation, improved tumor accumulation, and more significant anti-tumor efficacy compared with PSD NPs. Furthermore, PSD LPs exhibited reduced cardiotoxicity and kidney damage compared with the physical mixture of Taxol and Doxil, indicating better safety. Therefore, this novel nano-platform provides a strategy to deliver doxorubicin with other poorly soluble antineoplastic drugs for combination therapy with high efficacy and low toxicity.
Collapse
Key Words
- ALT, alanine transaminase
- AST, aspartate transaminase
- AUC, area under the curve
- BUN, blood urea nitrogen
- CHO, cholesterol
- CO2, carbon dioxide
- CR, creatinine
- Combination therapy
- Cu2+, copper ions
- DL, drug loading
- DLS, dynamic light scattering
- DMSO, dimethyl sulfoxide
- DNA, deoxyribonucleic acid
- DOX, doxorubicin
- DSPE-PEG2000, 2-distearoyl-snglycero-3-phosphoethanolamine-N-[methyl(polyethylene glycol)-2000
- DTT, d,l-dithiothreitol
- EDTA, ethylene diamine tetraacetic acid
- EE, encapsulation efficacy
- FBS, fetal bovine serum
- GSH, glutathione
- H&E, hematoxylin and eosin
- H2O2, hydrogen peroxide
- HEPES, 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid
- HPLC, high-performance liquid chromatography
- HSPC, hydrogenated soybean phospholipids
- IC50, half maximal inhibitory concentration
- IVIS, in vivo imaging system
- MLVs, multilamellar vesicles
- MRT, mean residence time
- MTD, maximum tolerated dose
- MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- Nanoparticles
- PBS, phosphate buffer saline
- PDI, polydispersity index
- PSD LPs, PTX-S-DOX liposomes
- PSD NPs, PTX-S-DOX self-assembled nanoparticles
- PSD, PTX-S-DOX
- PTX, paclitaxel
- Paclitaxel–doxorubicin prodrug
- Prodrug
- ROS, reactive oxygen species
- Remote loading liposomes
- SD, standard deviation
- Safety
- TEM, transmission electron microscopy
- UV, ultraviolet
Collapse
|
27
|
Ramajayam KK, Wolfe AM, Motamarry A, Yost J, Yost MJ, Haemmerich D. Computer Modeling of Drug Delivery with Thermosensitive Liposomes in a Realistic Three-Dimensional Geometry. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2020:5021-5024. [PMID: 33019114 DOI: 10.1109/embc44109.2020.9176829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Thermosensitive liposomes (TSL) are nanoparticles that can encapsulate therapeutic drugs, and release those drugs when exposed to hyperthermic temperatures (>40 °C). Combined with localized hyperthermia, TSL enable focused drug delivery. In this study, we created a three-dimensional (3D) computer model for simulating delivery with TSL-encapsulated doxorubicin (TSL-Dox) to mouse tumors. A mouse hind limb was scanned by a 3D scanner and the resulting geometry was imported into finite element modeling software, with a virtual tumor added. Then, heating by a surface probe was simulated. Further, a drug delivery model was coupled to the heat transfer model to simulate drug delivery kinetics. For comparison, experimental studies in gel phantoms and in vivo fluorescence imaging studies in mice carrying lung tumor xenografts were performed. We report the tissue temperature profile, drug concentration profile and compare the experimental studies with the computer model. The thermistor produced very localized heating that resulted in highest drug delivery to regions near the probe. The average tumor temperature was 38.2˚C (range 34.4-43.4˚C), and produced an average tumor drug concentration of 11.8 µg/g (0.3-28.1 µg/g) after 15 min heating, and 25.6 µg/g (0.3-52 µg/g), after 60 min heating. The computer model reproduced the temperature profile compared to phantom experiments (mean error 0.71 °C, range 0.59-1.25 °C), as well as drug delivery profile as compared to in vivo studies. Our results suggest feasibility of using this approach to model drug delivery in preclinical studies with accurate model geometry.
Collapse
|
28
|
Yaroslavov A, Efimova A, Smirnova N, Erzunov D, Lukashev N, Grozdova I, Melik-Nubarov N. A novel approach to a controlled opening of liposomes. Colloids Surf B Biointerfaces 2020; 190:110906. [DOI: 10.1016/j.colsurfb.2020.110906] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 01/09/2023]
|
29
|
Theranostic MRI liposomes for magnetic targeting and ultrasound triggered release of the antivascular CA4P. J Control Release 2020; 322:137-148. [PMID: 32145266 DOI: 10.1016/j.jconrel.2020.03.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/05/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023]
Abstract
Theranostic nanocarriers of antivascular drug encapsulated in thermosensitive ultramagnetic liposomes can be advantageously designed to provide a locally high concentration and an active delivery, with image-guided Magnetic Resonance Imaging (MRI) so as to reliably cure tumor. We propose a novel therapeutic strategy consisting of the magnetic accumulation of Ultra Magnetic Liposomes (UML) followed by High-Intensity Focused Ultrasound (HIFU) to trigger the release of an antivascular agent monitored by MRI. For this purpose, we co-encapsulated Combretastatin A4 phosphate (CA4P), a vascular disrupting agent, in the core of UML to obtain CA4P-loaded thermosensitive Ultra Magnetic Liposomes (CA4P-UML). To assess the HIFU parameters, the CA4P release has been triggered in vitro by local heating HIFU at the lipids transition temperature. Morphology of endothelial cells was assessed to evaluate the effect of encapsulated versus non-encapsulated CA4P. The efficiency of a treatment combining the magnetic targeting of CA4P-UML with the CA4P release triggered by HIFU was studied in CT26 murine tumors. Tumor perfusion and volume regression parameters were monitored by multiparametric quantitative anatomical and dynamic in vivo MRI at 7 T. Additionally, vascularization and cellularity were evaluated ex-vivo by histology. This thorough investigation showed that the combined treatment exhibited a full benefit. A 150-fold improvement compared with the chemotherapy alone was obtained using a magnetic targeting of CA4P-UML triggered by HIFU, and was consistent with an expected effect on vascularization 24 h after treatment.
Collapse
|
30
|
Seynhaeve A, Amin M, Haemmerich D, van Rhoon G, ten Hagen T. Hyperthermia and smart drug delivery systems for solid tumor therapy. Adv Drug Deliv Rev 2020; 163-164:125-144. [PMID: 32092379 DOI: 10.1016/j.addr.2020.02.004] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 12/31/2022]
Abstract
Chemotherapy is a cornerstone of cancer therapy. Irrespective of the administered drug, it is crucial that adequate drug amounts reach all cancer cells. To achieve this, drugs first need to be absorbed, then enter the blood circulation, diffuse into the tumor interstitial space and finally reach the tumor cells. Next to chemoresistance, one of the most important factors for effective chemotherapy is adequate tumor drug uptake and penetration. Unfortunately, most chemotherapeutic agents do not have favorable properties. These compounds are cleared rapidly, distribute throughout all tissues in the body, with only low tumor drug uptake that is heterogeneously distributed within the tumor. Moreover, the typical microenvironment of solid cancers provides additional hurdles for drug delivery, such as heterogeneous vascular density and perfusion, high interstitial fluid pressure, and abundant stroma. The hope was that nanotechnology will solve most, if not all, of these drug delivery barriers. However, in spite of advances and decades of nanoparticle development, results are unsatisfactory. One promising recent development are nanoparticles which can be steered, and release content triggered by internal or external signals. Here we discuss these so-called smart drug delivery systems in cancer therapy with emphasis on mild hyperthermia as a trigger signal for drug delivery.
Collapse
|
31
|
Increased Temperature Facilitates Adeno-Associated Virus Vector Transduction of Colorectal Cancer Cell Lines in a Manner Dependent on Heat Shock Protein Signature. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9107140. [PMID: 32090115 PMCID: PMC7031720 DOI: 10.1155/2020/9107140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/30/2019] [Accepted: 08/10/2019] [Indexed: 12/24/2022]
Abstract
Colorectal cancer (CRC) is one of the most common cancers in human population. A great achievement in the treatment of CRC was the introduction of targeted biological drugs and solutions of chemotherapy, combined with hyperthermia. Cytoreductive surgery and HIPEC (hyperthermic intraperitoneal chemotherapy) extends the patients' survival with CRC. Recently, gene therapy approaches are also postulated. The studies indicate the possibility of enhancing the gene transfer to cells by recombinant adeno-associated vectors (rAAV) at hyperthermia. The rAAV vectors arouse a lot of attention in the field of cancer treatment due to many advantages. In this study, the effect of elevated temperature on the transduction efficiency of rAAV vectors on CRC cells with different origin and gene profile was examined. The effect of heat shock on the penetration of rAAV vectors into CRC cells in relation with the expression of HSP and AAV receptor genes was tested. It was found that the examined cells under hyperthermia (43°C, 1 h) are transduced at a higher level than in normal conditions (37°C). The results also indicate that studied RKO, HT-29, and LS411N cell lines express HSP genes at different levels under both 37°C and 43°C. Moreover, the results showed that the expression of AAV receptors increases in response to elevated temperature. The study suggests that increased rAAV transfer to CRC can be achieved under elevated temperature conditions. The obtained results provide information relevant to the design of new solutions in CRC therapy based on the combination of hyperthermia, chemotherapy, and gene therapy.
Collapse
|
32
|
Motamarry A, Negussie AH, Rossmann C, Small J, Wolfe AM, Wood BJ, Haemmerich D. Real-time fluorescence imaging for visualization and drug uptake prediction during drug delivery by thermosensitive liposomes. Int J Hyperthermia 2020; 36:817-826. [PMID: 31451077 DOI: 10.1080/02656736.2019.1642521] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Objective: Thermosensitive liposomal doxorubicin (TSL-Dox) is a promising stimuli-responsive nanoparticle drug delivery system that rapidly releases the contained drug in response to hyperthermia (HT) (>40 °C). Combined with localized heating, TSL-Dox allows highly localized delivery. The goals of this study were to demonstrate that real-time fluorescence imaging can visualize drug uptake during delivery, and can predict tumor drug uptake. Methods: Nude mice carrying subcutaneous tumors (Lewis lung carcinoma) were anesthetized and injected with TSL-Dox (5 mg/kg dose). Localized HT was induced by heating tumors for 15, 30 or 60 min via a custom-designed HT probe placed superficially at the tumor location. In vivo fluorescence imaging (excitation 523 nm, emission 610 nm) was performed before, during, and for 5 min following HT. After imaging, tumors were extracted, drug uptake was quantified by high-performance liquid chromatography, and correlated with in vivo fluorescence. Plasma samples were obtained before and after HT to measure TSL-Dox pharmacokinetics. Results: Local drug uptake could be visualized in real-time during HT. Compared to unheated control tumors, fluorescence of heated tumors increased by 4.6-fold (15 min HT), 9.3-fold (30 min HT), and 13.2-fold (60 min HT). HT duration predicted tumor drug uptake (p = .02), with tumor drug concentrations of 4.2 ± 1.3 µg/g (no HT), 7.1 ± 5.9 µg/g (15 min HT), 14.1 ± 6.7 µg/g (30 min HT) and 21.4 ± 12.6 µg/g (60 min HT). There was good correlation (R2 = 0.67) between fluorescence of the tumor region and tumor drug uptake. Conclusions: Real-time in vivo fluorescence imaging can visualize drug uptake during delivery, and can predict tumor drug uptake.
Collapse
Affiliation(s)
- Anjan Motamarry
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina , Charleston , SC , USA.,Department of Pediatrics, Medical University of South Carolina , Charleston , SC , USA
| | - Ayele H Negussie
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health , Bethesda , MD , USA
| | - Christian Rossmann
- Department of Pediatrics, Medical University of South Carolina , Charleston , SC , USA
| | - James Small
- Department of Public Health Sciences, Medical University of South Carolina , Charleston , SC , USA
| | - A Marissa Wolfe
- Department of Comparative Medicine, Medical University of South Carolina , Charleston , SC , USA
| | - Bradford J Wood
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health , Bethesda , MD , USA
| | - Dieter Haemmerich
- Department of Pediatrics, Medical University of South Carolina , Charleston , SC , USA.,Department of Bioengineering, Clemson University , Clemson , SC , USA
| |
Collapse
|
33
|
Norouzi M, Amerian M, Amerian M, Atyabi F. Clinical applications of nanomedicine in cancer therapy. Drug Discov Today 2020; 25:107-125. [DOI: 10.1016/j.drudis.2019.09.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/03/2019] [Accepted: 09/24/2019] [Indexed: 12/23/2022]
|
34
|
Cheng CA, Chen W, Zhang L, Wu HH, Zink JI. A Responsive Mesoporous Silica Nanoparticle Platform for Magnetic Resonance Imaging-Guided High-Intensity Focused Ultrasound-Stimulated Cargo Delivery with Controllable Location, Time, and Dose. J Am Chem Soc 2019; 141:17670-17684. [PMID: 31604010 DOI: 10.1021/jacs.9b07591] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Magnetic resonance imaging (MRI) is an essential modality for clinical diagnosis, and MRI-guided high-intensity focused ultrasound (MRgHIFU) is a powerful technology for targeted therapy. Clinical applications of MRgHIFU primarily utilize hyperthermia and ablation to treat cancerous tissue, but for drug delivery applications thermal damage is undesirable. A biofriendly MRgHIFU-responsive mesoporous silica nanoparticle (MSN) platform that is stimulated within a physiological safe temperature range has been developed, reducing the possibility of thermal damage to the surrounding healthy tissues. Biocompatible polyethylene glycol (PEG) was employed to cap the pores of MSNs, and the release of cargo molecules by HIFU occurs without substantial temperature increase (∼4 °C). To visualize by MRI and measure the stimulated delivery in situ, a U.S. Food and Drug Administration (FDA)-approved gadolinium-based contrast agent, gadopentetate dimeglumine (Gd(DTPA)2-), was used as the imageable cargo. Taking advantage of the three-dimensional (3-D) imaging and targeting capabilities of MRgHIFU, the release of Gd(DTPA)2- stimulated by HIFU was pinpointed at the HIFU focal point in 3-D space in a tissue-mimicking gel phantom. The amount of Gd(DTPA)2- released was controlled by HIFU stimulation times and power levels. A positive correlation between the amount of Gd(DTPA)2- released and T1 was found. The MRgHIFU-stimulated cargo release was further imaged in a sample of ex vivo animal tissue. With this technology, the biodistribution of the nanocarriers can be tracked and the MRgHIFU-stimulated cargo release can be pinpointed, opening up an opportunity for future image-guided theranostic applications.
Collapse
Affiliation(s)
- Chi-An Cheng
- Department of Bioengineering , University of California Los Angeles , Los Angeles , California 90095 , United States.,California NanoSystems Institute , University of California Los Angeles , Los Angeles 90095 , California , United States
| | - Wei Chen
- Department of Chemistry & Biochemistry , University of California Los Angeles , Los Angeles , California 90095 , United States.,California NanoSystems Institute , University of California Los Angeles , Los Angeles 90095 , California , United States
| | - Le Zhang
- Department of Radiological Sciences, David Geffen School of Medicine , University of California Los Angeles , Los Angeles , California 90095 , United States
| | - Holden H Wu
- Department of Bioengineering , University of California Los Angeles , Los Angeles , California 90095 , United States.,Department of Radiological Sciences, David Geffen School of Medicine , University of California Los Angeles , Los Angeles , California 90095 , United States
| | - Jeffrey I Zink
- Department of Chemistry & Biochemistry , University of California Los Angeles , Los Angeles , California 90095 , United States.,California NanoSystems Institute , University of California Los Angeles , Los Angeles 90095 , California , United States
| |
Collapse
|
35
|
Tucci ST, Kheirolomoom A, Ingham ES, Mahakian LM, Tam SM, Foiret J, Hubbard NE, Borowsky AD, Baikoghli M, Cheng RH, Ferrara KW. Tumor-specific delivery of gemcitabine with activatable liposomes. J Control Release 2019; 309:277-288. [PMID: 31301340 DOI: 10.1016/j.jconrel.2019.07.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 07/05/2019] [Accepted: 07/10/2019] [Indexed: 12/18/2022]
Abstract
Gemcitabine delivery to pancreatic ductal adenocarcinoma is limited by poor pharmacokinetics, dense fibrosis and hypo-vascularization. Activatable liposomes, with drug release resulting from local heating, enhance serum stability and circulation, and the released drug retains the ability to diffuse within the tumor. A limitation of liposomal gemcitabine has been the low loading efficiency. To address this limitation, we used the superior solubilizing potential of copper (II) gluconate to form a complex with gemcitabine at copper:gemcitabine (1:4). Thermosensitive liposomes composed of DPPC:DSPC:DSPE-PEG2k (80:15:5, mole%) then reached 12 wt% loading, 4-fold greater than previously reported values. Cryo transmission electron microscopy confirmed the presence of a liquid crystalline gemcitabine‑copper mixture. The optimized gemcitabine liposomes released 60% and 80% of the gemcitabine within 1 and 5 min, respectively, at 42 °C. Liposomal encapsulation resulted in a circulation half-life of ~2 h in vivo (compared to reported circulation of 16 min for free gemcitabine in mice), and free drug was not detected within the plasma. The resulting gemcitabine liposomes were efficacious against both murine breast cancer and pancreatic cancer in vitro. Three repeated treatments of activatable gemcitabine liposomes plus ultrasound hyperthermia regressed or eliminated tumors in the neu deletion model of murine breast cancer with limited toxicity, enhancing survival when compared to treatment with gemcitabine alone. With 5% of the free gemcitabine dose (5 rather than 100 mg/kg), tumor growth was suppressed to the same degree as gemcitabine. Additionally, in a more aggressive tumor model of murine pancreatic cancer, liposomal gemcitabine combined with local hyperthermia induced cell death and regions of apoptosis and necrosis.
Collapse
Affiliation(s)
- Samantha T Tucci
- University of California, Davis, Department of Biomedical Engineering, Davis, CA 95616, USA
| | - Azadeh Kheirolomoom
- University of California, Davis, Department of Biomedical Engineering, Davis, CA 95616, USA; Stanford University, Department of Radiology, 3165 Porter Drive, Palo Alto, CA 94304, USA
| | - Elizabeth S Ingham
- University of California, Davis, Department of Biomedical Engineering, Davis, CA 95616, USA
| | - Lisa M Mahakian
- University of California, Davis, Department of Biomedical Engineering, Davis, CA 95616, USA
| | - Sarah M Tam
- University of California, Davis, Department of Biomedical Engineering, Davis, CA 95616, USA
| | - Josquin Foiret
- Stanford University, Department of Radiology, 3165 Porter Drive, Palo Alto, CA 94304, USA
| | - Neil E Hubbard
- University of California, Davis, Center for Comparative Medicine, Davis, CA 95616, USA
| | - Alexander D Borowsky
- University of California, Davis, Center for Comparative Medicine, Davis, CA 95616, USA
| | - Mo Baikoghli
- University of California, Davis, Department of Molecular and Cellular Biology, Davis, CA 95616, USA
| | - R Holland Cheng
- University of California, Davis, Department of Molecular and Cellular Biology, Davis, CA 95616, USA
| | - Katherine W Ferrara
- Stanford University, Department of Radiology, 3165 Porter Drive, Palo Alto, CA 94304, USA.
| |
Collapse
|
36
|
Bi H, Xue J, Jiang H, Gao S, Yang D, Fang Y, Shi K. Current developments in drug delivery with thermosensitive liposomes. Asian J Pharm Sci 2019; 14:365-379. [PMID: 32104466 PMCID: PMC7032122 DOI: 10.1016/j.ajps.2018.07.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 06/26/2018] [Accepted: 07/25/2018] [Indexed: 12/19/2022] Open
Abstract
Thermosensitive liposomes (TSLs) have been an important research area in the field of tumor targeted chemotherapy. Since the first TSLs appeared that using 1,2-dipalmitoyl-sn-glyce-ro-3-phosphocholine (DPPC) as the primary liposomal lipid, many studies have been done using this type of liposome from basic and practical aspects. While TSLs composed of DPPC enhance the cargo release near the phase transition temperature, it has been shown that many factors affect their temperature sensitivity. Thus numerous attempts have been undertaken to develop new TSLs for improving their thermal response performance. The main objective of this review is to introduce the development and recent update of innovative TSLs formulations, including combination of radiofrequency ablation (RFA), high-intensity focused ultrasound (HIFU), magnetic resonance imaging (MRI) and alternating magnetic field (AMF). In addition, various factors affecting the design of TSLs, such as lipid composition, surfactant, size and serum components are also discussed.
Collapse
Key Words
- (DPPC), 1,2-dipalmitoyl-sn-glycero-3-phosphatidylcholine
- (DPPGOG), 1,2-dipalmitoyl-sn-glycero-3-phosphoglyceroglycerol
- (DSPC), 1,2-distearoyl-sn-glycero-3-phosphocholine
- (DSPE-mPEG2000), 1,2-distearoyl-sn-glycero-3-phosphatiylethanol-amine-N-[methoxy(polyethyleneglycol)-2000]
- (LTSLs), lyso-lipid temperature sensitive liposomes
- (MPPC), 1-myristoyl-2-palmitoyl-sn-glycero-3-phosphatidylcholine
- (MSPC), 1-stearoyl-2-hydroxy-sn-glycero-3-phosphatidylcholine
- (P-lyso-PC), lysophosphatidylcholine
- (P188), 1-palmitoyl-2-stearoyl-sn-glycero-3-phosphatidylcholinex
- (P188), HO-(C2H4O)a-(C3H6O)b-(C2H4O)c-H, a=80, b=27, c=80
- Content release rate
- Drug delivery
- Hyperthermia
- Smart liposomes
- Thermosensitive liposomes
- Tumor chemotherapy
- fTSLs, fast release TSLs
- sTSLs, slow release TSLs
Collapse
Affiliation(s)
- Hongshu Bi
- Institute of New Drug Development, Liaoning Yaolian Pharmaceutical Co., Ltd., Benxi, Liaoning 117004, China
| | - Jianxiu Xue
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Hong Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Shan Gao
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Dongjuan Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Yan Fang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Kai Shi
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| |
Collapse
|
37
|
Kheirolomoom A, Silvestrini MT, Ingham ES, Mahakian LM, Tam SM, Tumbale SK, Foiret J, Hubbard NE, Borowsky AD, Ferrara KW. Combining activatable nanodelivery with immunotherapy in a murine breast cancer model. J Control Release 2019; 303:42-54. [PMID: 30978432 DOI: 10.1016/j.jconrel.2019.04.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/11/2019] [Accepted: 04/07/2019] [Indexed: 12/20/2022]
Abstract
A successful chemotherapy-immunotherapy solid-tumor protocol should accomplish the following goals: debulk large tumors, release tumor antigen for cross-presentation and cross-priming, release cancer-suppressive cytokines and enhance anti-tumor immune cell populations. Thermally-activated drug delivery particles have the potential to synergize with immunotherapeutics to accomplish these goals; activation can release chemotherapy within bulky solid tumors and can enhance response when combined with immunotherapy. We set out to determine whether a single protocol, combining locally-activated chemotherapy and agonist immunotherapy, could accomplish these goals and yield a potentially translational therapy. For effective delivery of free doxorubicin to tumors with minimal toxicity, we stabilized doxorubicin with copper in temperature-sensitive liposomes that rapidly release free drug in the vasculature of cancer lesions upon exposure to ultrasound-mediated hyperthermia. We found that in vitro exposure of tumor cells to hyperthermia and doxorubicin resulted in immunogenic cell death and the local release of type I interferons across murine cancer cell lines. Following intravenous injection, local activation of the liposomes within a single tumor released doxorubicin and enhanced cross-presentation of a model antigen at distant tumor sites. While a variety of protocols achieved a complete response in >50% of treated mice, the complete response rate was greatest (90%) when 1 week of immunotherapy priming preceded a single activatable chemotherapeutic administration. While repeated chemotherapeutic delivery reduced local viable tumor, the complete response rate and a subset of tumor immune cells were also reduced. Taken together, the results suggest that activatable chemotherapy can enhance adjuvant immunotherapy; however, in a murine model the systemic adaptive immune response was greatest with a single administration of chemotherapy.
Collapse
Affiliation(s)
- Azadeh Kheirolomoom
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA; Stanford University, Department of Radiology, 3165 Porter Drive, Palo Alto, CA 94304, USA
| | - Matthew T Silvestrini
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Elizabeth S Ingham
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Lisa M Mahakian
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Sarah M Tam
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Spencer K Tumbale
- Stanford University, Department of Radiology, 3165 Porter Drive, Palo Alto, CA 94304, USA
| | - Josquin Foiret
- Stanford University, Department of Radiology, 3165 Porter Drive, Palo Alto, CA 94304, USA
| | - Neil E Hubbard
- University of California, Davis, Center for Comparative Medicine, Davis, CA 95616, USA
| | - Alexander D Borowsky
- University of California, Davis, Center for Comparative Medicine, Davis, CA 95616, USA
| | - Katherine W Ferrara
- Stanford University, Department of Radiology, 3165 Porter Drive, Palo Alto, CA 94304, USA.
| |
Collapse
|
38
|
Nonviral ultrasound-mediated gene delivery in small and large animal models. Nat Protoc 2019; 14:1015-1026. [PMID: 30804568 DOI: 10.1038/s41596-019-0125-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 12/18/2018] [Indexed: 12/15/2022]
Abstract
Ultrasound-mediated gene delivery (sonoporation) is a minimally invasive, nonviral and clinically translatable method of gene therapy. This method offers a favorable safety profile over that of viral vectors and is less invasive as compared with other physical gene delivery approaches (e.g., electroporation). We have previously used sonoporation to overexpress transgenes in different skeletal tissues in order to induce tissue regeneration. Here, we provide a protocol that could easily be adapted to address various other targets of tissue regeneration or additional applications, such as cancer and neurodegenerative diseases. This protocol describes how to prepare, conduct and optimize ultrasound-mediated gene delivery in both a murine and a porcine animal model. The protocol includes the preparation of a microbubble-DNA mix and in vivo sonoporation under ultrasound imaging. Ultrasound-mediated gene delivery can be accomplished within 10 min. After DNA delivery, animals can be followed to monitor gene expression, protein secretion and other transgene-specific outcomes, including tissue regeneration. This procedure can be accomplished by a competent graduate student or technician with prior experience in ultrasound imaging or in performing in vivo procedures.
Collapse
|
39
|
Kauscher U, Holme MN, Björnmalm M, Stevens MM. Physical stimuli-responsive vesicles in drug delivery: Beyond liposomes and polymersomes. Adv Drug Deliv Rev 2019; 138:259-275. [PMID: 30947810 PMCID: PMC7180078 DOI: 10.1016/j.addr.2018.10.012] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/30/2018] [Accepted: 10/22/2018] [Indexed: 12/11/2022]
Abstract
Over the past few decades, a range of vesicle-based drug delivery systems have entered clinical practice and several others are in various stages of clinical translation. While most of these vesicle constructs are lipid-based (liposomes), or polymer-based (polymersomes), recently new classes of vesicles have emerged that defy easy classification. Examples include assemblies with small molecule amphiphiles, biologically derived membranes, hybrid vesicles with two or more classes of amphiphiles, or more complex hierarchical structures such as vesicles incorporating gas bubbles or nanoparticulates in the lumen or membrane. In this review, we explore these recent advances and emerging trends at the edge and just beyond the research fields of conventional liposomes and polymersomes. A focus of this review is the distinct behaviors observed for these classes of vesicles when exposed to physical stimuli - such as ultrasound, heat, light and mechanical triggers - and we discuss the resulting potential for new types of drug delivery, with a special emphasis on current challenges and opportunities.
Collapse
Affiliation(s)
- Ulrike Kauscher
- Department of Materials, Imperial College London, London SW7 2AZ, UK; Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Margaret N Holme
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Mattias Björnmalm
- Department of Materials, Imperial College London, London SW7 2AZ, UK; Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Molly M Stevens
- Department of Materials, Imperial College London, London SW7 2AZ, UK; Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK; Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm SE-171 77, Sweden.
| |
Collapse
|
40
|
Evans ER, Bugga P, Asthana V, Drezek R. Metallic Nanoparticles for Cancer Immunotherapy. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2018; 21:673-685. [PMID: 30197553 PMCID: PMC6124314 DOI: 10.1016/j.mattod.2017.11.022] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Cancer immunotherapy, or the utilization of the body's immune system to attack tumor cells, has gained prominence over the past few decades as a viable cancer treatment strategy. Recently approved immunotherapeutics have conferred remission upon patients with previously bleak outcomes and have expanded the number of tools available to treat cancer. Nanoparticles -including polymeric, liposomal, and metallic formulations - naturally traffic to the spleen and lymph organs and the relevant immune cells therein, making them good candidates for delivery of immunotherapeutic agents. Metallic nanoparticle formulations in particular are advantageous because of their potential for dense surface functionalization and their capability for optical or heat based therapeutic methods. Many research groups have investigated the potential of nanoparticle-mediated delivery platforms to improve the efficacy of immunotherapies. Despite the significant preclinical successes demonstrated by many of these platforms over the last twenty years, few metallic nanoparticles have successfully entered clinical trials with none achieving FDA approval for cancer therapy. In this review, we will discuss preclinical research and clinical trials involving metallic nanoparticles (MNPs) for cancer immunotherapy applications and discuss the potential for clinical translation of MNPs.
Collapse
Affiliation(s)
- Emily Reiser Evans
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Pallavi Bugga
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Vishwaratn Asthana
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Rebekah Drezek
- Department of Bioengineering, Rice University, Houston, TX 77005, United States. Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, United States
| |
Collapse
|
41
|
Centelles MN, Wright M, So PW, Amrahli M, Xu XY, Stebbing J, Miller AD, Gedroyc W, Thanou M. Image-guided thermosensitive liposomes for focused ultrasound drug delivery: Using NIRF-labelled lipids and topotecan to visualise the effects of hyperthermia in tumours. J Control Release 2018; 280:87-98. [DOI: 10.1016/j.jconrel.2018.04.047] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 04/25/2018] [Accepted: 04/27/2018] [Indexed: 12/26/2022]
|
42
|
Abstract
Ultrasound is a rapidly advancing field with many emerging diagnostic and therapeutic applications. For diagnostics, new vascular targets are routinely identified and mature technologies are being translated to humans, while other recent innovations may bring about the creation of acoustic reporter genes and micron-scale resolution with ultrasound. As a cancer therapy, ultrasound is being explored as an adjuvant to immune therapies and to deliver acoustically or thermally active drugs to tumor regions. Ultrasound-enhanced delivery across the blood brain barrier (BBB) could potentially be very impactful for brain cancers and neurodegenerative diseases where the BBB often impedes the delivery of therapeutic molecules. In this minireview, we provide an overview of these topics in the field of ultrasound that are especially relevant to the interests of World Molecular Imaging Society.
Collapse
|
43
|
Matsuki D, Adewale O, Horie S, Okajima J, Komiya A, Oluwafemi O, Maruyama S, Mori S, Kodama T. Treatment of tumor in lymph nodes using near-infrared laser light-activated thermosensitive liposome-encapsulated doxorubicin and gold nanorods. JOURNAL OF BIOPHOTONICS 2017; 10:1676-1682. [PMID: 28417560 DOI: 10.1002/jbio.201600241] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 06/07/2023]
Abstract
Tumor metastasis to lymph nodes is an important contributory factor for cancer-related deaths despite recent developments in cancer therapy. In this study, we demonstrate that tumor in the proper axillary lymph node (PALN) of the mouse can be treated by the application of external laser light to trigger the unloading of doxorubicin (DOX) encapsulated in thermosensitive liposomes (TSLs) administered together with gold nanorods (GNRs). GNRs + DOX-TSLs were injected into a mouse lymph node containing cancer cells (malignant fibrous histiocytoma-like cells) and intranodal DOX release was activated using near-infrared (NIR) laser irradiation. The temperature changes arising from the laser-irradiated GNRs triggered the release of DOX from the TSLs. A greater degree of inhibition of tumor growth was found in the co-therapy group compared to the other groups. The treatment effect was achieved by a combination of chemotherapy and NIR-activated hyperthermia. In vivo bioluminescence imaging and histological analysis confirmed tumor necrosis in response to combined treatment. This work presents a theranostic approach with excellent treatment results that has the potential to be developed into an alternative to surgery for the treatment of breast cancer metastasis.
Collapse
Affiliation(s)
- Daisuke Matsuki
- Laboratory of Biomedical Engineering for Cancer, Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
| | - Oladipo Adewale
- Department of Applied Chemistry, University of Johannesburg, Doornfontein Campus P.O. Box 17011, Doornfontein, 2028, South Africa
| | - Sachiko Horie
- Laboratory of Biomedical Engineering for Cancer, Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
| | - Junnosuke Okajima
- Institute of Fluid Science, Tohoku University, 2-1-1 Katahira, Aoba, Sendai, 980-8577, Japan
| | - Atsuki Komiya
- Institute of Fluid Science, Tohoku University, 2-1-1 Katahira, Aoba, Sendai, 980-8577, Japan
| | - Oluwatobi Oluwafemi
- Department of Applied Chemistry, University of Johannesburg, Doornfontein Campus P.O. Box 17011, Doornfontein, 2028, South Africa
| | - Shigenao Maruyama
- Institute of Fluid Science, Tohoku University, 2-1-1 Katahira, Aoba, Sendai, 980-8577, Japan
| | - Shiro Mori
- Laboratory of Biomedical Engineering for Cancer, Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Department of Oral and Maxillofacial Surgery, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai, 980-8575, Japan
| | - Tetsuya Kodama
- Laboratory of Biomedical Engineering for Cancer, Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4-1 Seiryo, Aoba, Sendai, 980-8575, Japan
| |
Collapse
|
44
|
Thermal combination therapies for local drug delivery by magnetic resonance-guided high-intensity focused ultrasound. Proc Natl Acad Sci U S A 2017; 114:E4802-E4811. [PMID: 28566498 DOI: 10.1073/pnas.1700790114] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Several thermal-therapy strategies such as thermal ablation, hyperthermia-triggered drug delivery from temperature-sensitive liposomes (TSLs), and combinations of the above were investigated in a rhabdomyosarcoma rat tumor model (n = 113). Magnetic resonance-guided high-intensity focused ultrasound (MR-HIFU) was used as a noninvasive heating device with precise temperature control for image-guided drug delivery. For the latter, TSLs were prepared, coencapsulating doxorubicin (dox) and [Gd(HPDO3A)(H2O)], and injected in tumor-bearing rats before MR-HIFU treatment. Four treatment groups were defined: hyperthermia, ablation, hyperthermia followed by ablation, or no HIFU. The intratumoral TSL and dox distribution were analyzed by single-photon emission computed tomography (SPECT)/computed tomography (CT), autoradiography, and fluorescence microscopy. Dox biodistribution was quantified and compared with that of nonliposomal dox. Finally, the treatment efficacy of all heating strategies plus additional control groups (saline, free dox, and Caelyx) was assessed by tumor growth measurements. All HIFU heating strategies combined with TSLs resulted in cellular uptake of dox deep into the interstitial space and a significant increase of tumor drug concentrations compared with a treatment with free dox. Ablation after TSL injection showed [Gd(HPDO3A)(H2O)] and dox release along the tumor rim, mirroring the TSL distribution pattern. Hyperthermia either as standalone treatment or before ablation ensured homogeneous TSL, [Gd(HPDO3A)(H2O)], and dox delivery across the tumor. The combination of hyperthermia-triggered drug delivery followed by ablation showed the best therapeutic outcome compared with all other treatment groups due to direct induction of thermal necrosis in the tumor core and efficient drug delivery to the tumor rim.
Collapse
|
45
|
Chaplin V, Caskey CF. Multi-focal HIFU reduces cavitation in mild-hyperthermia. J Ther Ultrasound 2017; 5:12. [PMID: 28413682 PMCID: PMC5390440 DOI: 10.1186/s40349-017-0089-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/07/2017] [Indexed: 12/21/2022] Open
Abstract
Background Mild-hyperthermia therapy (40–45 °C) with high-intensity focused ultrasound (HIFU) is a technique being considered in a number of different treatments such as thermally activated drug delivery, immune-stimulation, and as a chemotherapy adjuvant. Mechanical damage and loss of cell viability associated with HIFU-induced acoustic cavitation may pose a risk during these treatments or may hinder their success. Here we present a method that achieves mild heating and reduces cavitation by using a multi-focused HIFU beam. We quantify cavitation level and temperature rise in multi-focal sonications and compare it to single-focus sonications at the transducer geometric focus. Methods Continuous wave sonications were performed with the Sonalleve V2 transducer in gel phantoms and pork at 5, 10, 20, 40, 60, 80 acoustic watts for 30 s. Cavitation activity was measured with two ultrasound (US) imaging probes, both by computing the raw channel variance and using passive acoustic mapping (PAM). Temperature rise was measured with MR thermometry at 3 T. Cavitation and heating were compared for single- and multi-focal sonication geometries. Multi-focal sonications used four points equally spaced on a ring of either 4 mm or 8 mm diameter. Single-focus sonications were not steered. Results Multi-focal sonication generated distinct foci that were visible in MRI thermal maps in both phantoms and pork, and visible in PAM images in phantoms only. Cavitation activity (measured by channel variance) and mean PAM image value were highly correlated (r > 0.9). In phantoms, cavitation exponentially decreased over the 30-second sonication, consistent with depletion of cavitation nuclei. In pork, sporadic spikes signaling cavitation were observed with single focusing only. In both materials, the widest beam reduced average and peak cavitation level by a factor of two or more at each power tested when compared to a single focus. The widest beam reduced peak temperature by at least 10 °C at powers above 5 W, and created heating that was more spatially diffuse than single focus, resulting in more voxels in the mild heating (3–8 °C) range. Conclusions Multi-focal HIFU can be used to achieve mild temperature elevation and reduce cavitation activity.
Collapse
Affiliation(s)
- Vandiver Chaplin
- Vanderbilt University Institute of Imaging Science, 1161 21st Avenue South, Nashville, TN 37232 USA
| | - Charles F Caskey
- Vanderbilt University Institute of Imaging Science, 1161 21st Avenue South, Nashville, TN 37232 USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232 USA
| |
Collapse
|
46
|
Fite BZ, Kheirolomoom A, Foiret JL, Seo JW, Mahakian LM, Ingham ES, Tam SM, Borowsky AD, Curry FRE, Ferrara KW. Dynamic contrast enhanced MRI detects changes in vascular transport rate constants following treatment with thermally-sensitive liposomal doxorubicin. J Control Release 2017; 256:203-213. [PMID: 28395970 DOI: 10.1016/j.jconrel.2017.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 03/16/2017] [Accepted: 04/05/2017] [Indexed: 01/03/2023]
Abstract
Temperature-sensitive liposomal formulations of chemotherapeutics, such as doxorubicin, can achieve locally high drug concentrations within a tumor and tumor vasculature while maintaining low systemic toxicity. Further, doxorubicin delivery by temperature-sensitive liposomes can reliably cure local cancer in mouse models. Histological sections of treated tumors have detected red blood cell extravasation within tumors treated with temperature-sensitive doxorubicin and ultrasound hyperthermia. We hypothesize that the local release of drug into the tumor vasculature and resulting high drug concentration can alter vascular transport rate constants along with having direct tumoricidal effects. Dynamic contrast enhanced MRI (DCE-MRI) coupled with a pharmacokinetic model can detect and quantify changes in such vascular transport rate constants. Here, we set out to determine whether changes in rate constants resulting from intravascular drug release were detectable by MRI. We found that the accumulation of gadoteridol was enhanced in tumors treated with temperature-sensitive liposomal doxorubicin and ultrasound hyperthermia. While the initial uptake rate of the small molecule tracer was slower (k1=0.0478±0.011s-1 versus 0.116±0.047s-1) in treated compared to untreated tumors, the tracer was retained after treatment due to a larger reduction in the rate of clearance (k2=0.291±0.030s-1 versus 0.747±0.24s-1). While DCE-MRI assesses a combination of blood flow and permeability, ultrasound imaging of microvascular flow rate is sensitive only to changes in vascular flow rate; based on this technique, blood flow was not significantly altered 30min after treatment. In summary, DCE-MRI provides a means to detect changes that are associated with treatment by thermally-activated particles and such changes can be exploited to enhance local delivery.
Collapse
Affiliation(s)
- Brett Z Fite
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Azadeh Kheirolomoom
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Josquin L Foiret
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Jai W Seo
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Lisa M Mahakian
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Sarah M Tam
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Alexander D Borowsky
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA 95616, USA.
| | - Fitz-Roy E Curry
- Department of Physiology and Membrane Biology, University of California, Davis, CA 95616, USA.
| | - Katherine W Ferrara
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| |
Collapse
|
47
|
PBCA-based polymeric microbubbles for molecular imaging and drug delivery. J Control Release 2017; 259:128-135. [PMID: 28279799 DOI: 10.1016/j.jconrel.2017.03.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/23/2017] [Accepted: 03/03/2017] [Indexed: 02/08/2023]
Abstract
Microbubbles (MB) are routinely used as contrast agents for ultrasound (US) imaging. We describe different types of targeted and drug-loaded poly(n-butyl cyanoacrylate) (PBCA) MB, and demonstrate their suitability for multiple biomedical applications, including molecular US imaging and US-mediated drug delivery. Molecular imaging of angiogenic tumor blood vessels and inflamed atherosclerotic endothelium is performed by modifying the surface of PBCA MB with peptides and antibodies recognizing E-selectin and VCAM-1. Stable and inertial cavitation of PBCA MB enables sonoporation and permeabilization of blood vessels in tumors and in the brain, which can be employed for direct and indirect drug delivery. Direct drug delivery is based on US-induced release of (model) drug molecules from the MB shell. Indirect drug delivery refers to US- and MB-mediated enhancement of extravasation and penetration of co-administered drugs and drug delivery systems. These findings are in line with recently reported pioneering proof-of-principle studies showing the usefulness of (phospholipid) MB for molecular US imaging and sonoporation-enhanced drug delivery in patients. They aim to exemplify the potential and the broad applicability of combining MB with US to improve disease diagnosis and therapy.
Collapse
|
48
|
Boissenot T, Bordat A, Fattal E, Tsapis N. Ultrasound-triggered drug delivery for cancer treatment using drug delivery systems: From theoretical considerations to practical applications. J Control Release 2016; 241:144-163. [DOI: 10.1016/j.jconrel.2016.09.026] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/19/2016] [Accepted: 09/21/2016] [Indexed: 12/21/2022]
|
49
|
Kheirolomoom A, Ingham ES, Commisso J, Abushaban N, Ferrara KW. Intracellular trafficking of a pH-responsive drug metal complex. J Control Release 2016; 243:232-242. [PMID: 27746275 DOI: 10.1016/j.jconrel.2016.10.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 09/23/2016] [Accepted: 10/11/2016] [Indexed: 01/18/2023]
Abstract
We previously developed a pH-responsive copper-doxorubicin (CuDox) cargo in lysolipid-based temperature-sensitive liposomes (LTSLs). The CuDox complex is released from the particle by elevated temperature; however, full release of doxorubicin from CuDox requires a reduced pH, such as that expected in lysosomes. The primary goal of this study is to evaluate the cellular uptake and intracellular trafficking of the drug-metal complex in comparison with intact liposomes and free drug. We found that the CuDox complex was efficiently internalized by mammary carcinoma cells after release from LTSLs. Intracellular doxorubicin and copper were 6-fold and 5-fold greater, respectively, after a 0.5h incubation with the released CuDox complex, as compared to incubation with intact liposomes containing the complex. Total cellular doxorubicin fluorescence was similar following CuDox and free doxorubicin incubation. Imaging and mass spectrometry assays indicated that the CuDox complex was initially internalized intact but breaks down over time within cells, with intracellular copper decreasing more rapidly than intracellular doxorubicin. Doxorubicin fluorescence was reduced when complexed with copper, and nuclear fluorescence was reduced when cells were incubated with the CuDox complex as compared with free doxorubicin. Therapeutic efficacy, which typically results from intercalation of doxorubicin with DNA, was equivalent for the CuDox complex and free doxorubicin and was superior to that of liposomal doxorubicin formulations. Taken together, the results suggest that quenched CuDox reaches the nucleus and remains efficacious. In order to design protocols for the use of these temperature-sensitive particles in cancer treatment, the timing of hyperthermia relative to drug administration must be examined. When cells were heated to 42°C prior to the addition of free doxorubicin, nuclear drug accumulation increased by 1.8-fold in cancer cells after 5h, and cytotoxicity increased 1.4-fold in both cancer and endothelial cells. Endothelial cytotoxicity was similarly augmented with mild hyperthermia applied prior to treatment with released CuDox. In summary, we find that the drug-metal complex formed in temperature-sensitive particles can be internalized by cancer and endothelial cells resulting in therapeutic efficacy that is similar to free doxorubicin, and this efficacy can be enhanced by elevated temperature.
Collapse
Affiliation(s)
- Azadeh Kheirolomoom
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Elizabeth S Ingham
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Joel Commisso
- University of California, Davis, Interdisciplinary Center for Plasma Mass Spectrometry, Davis, CA 95616, USA
| | - Neveen Abushaban
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Katherine W Ferrara
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| |
Collapse
|
50
|
Pradhan L, Thakur B, Srivastava R, Ray P, Bahadur D. Assessing Therapeutic Potential of Magnetic Mesoporous Nanoassemblies for Chemo-Resistant Tumors. Theranostics 2016; 6:1557-72. [PMID: 27446490 PMCID: PMC4955055 DOI: 10.7150/thno.15231] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/28/2016] [Indexed: 12/28/2022] Open
Abstract
Smart drug delivery system with strategic drug distribution is the future state-of-the-art treatment for any malignancy. To investigate therapeutic potential of such nanoparticle mediated delivery system, we examined the efficacy of dual drug-loaded, pH and thermo liable lipid coated mesoporous iron oxide-based magnetic nanoassemblies (DOX:TXL-LMMNA) in mice bearing both drug sensitive (A2780(S)) and drug resistant (A2780-CisR) ovarian cancer tumor xenografts. In presence of an external AC magnetic field (ACMF), DOX:TXL-LMMNA particles disintegrate to release encapsulated drug due to hyperthermic temperatures (41-45 ºC). In vivo bio distribution study utilizing the optical and magnetic properties of DOX:TXL-LMMNA particles demonstrated minimum organ specific toxicity. Noninvasive bioluminescence imaging of mice bearing A2780(S) tumors and administered with DOX-TXL-LMMNA followed by the application of ACMF revealed 65% less luminescence signal and 80% mice showed complete tumor regression within eight days. A six months follow-up study revealed absence of relapse in 70% of the mice. Interestingly, the A2780-CisR tumors which did not respond to drug alone (DOX:TXL) showed 80% reduction in luminescence and tumor volume with DOX:TXL-LMMNA after thermo-chemotherapy within eight days. Cytotoxic effect of DOX:TXL-LMMNA particles was more pronounced in A2780-CisR cells than in their sensitive counterpart. Thus these novel stimuli sensitive nanoassemblies hold great promise for therapy resistant malignancies and future clinical applications.
Collapse
Affiliation(s)
- Lina Pradhan
- 1. Centre for Research in Nanotechnology and Sciences, IIT Bombay, Mumbai, 400076,India
- 4. Department of Metallurgical Engineering and Materials Science, IIT Bombay, Mumbai, 400076 India
| | - Bhushan Thakur
- 2. Advance Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Rohit Srivastava
- 3. Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, 400076, India
| | - Pritha Ray
- 2. Advance Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Dhirendra Bahadur
- 4. Department of Metallurgical Engineering and Materials Science, IIT Bombay, Mumbai, 400076 India
| |
Collapse
|