1
|
Zhu X, Shi Z, Mao Y, Lächelt U, Huang R. Cell Membrane Perforation: Patterns, Mechanisms and Functions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310605. [PMID: 38344881 DOI: 10.1002/smll.202310605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/21/2023] [Indexed: 02/21/2024]
Abstract
Cell membrane is crucial for the cellular activities, and any disruption to it may affect the cells. It is demonstrated that cell membrane perforation is associated with some biological processes like programmed cell death (PCD) and infection of pathogens. Specific developments make it a promising technique to perforate the cell membrane controllably and precisely. The pores on the cell membrane provide direct pathways for the entry and exit of substances, and can also cause cell death, which means reasonable utilization of cell membrane perforation is able to assist intracellular delivery, eliminate diseased or cancerous cells, and bring about other benefits. This review classifies the patterns of cell membrane perforation based on the mechanisms into 1) physical patterns, 2) biological patterns, and 3) chemical patterns, introduces the characterization methods and then summarizes the functions according to the characteristics of reversible and irreversible pores, with the aim of providing a comprehensive summary of the knowledge related to cell membrane perforation and enlightening broad applications in biomedical science.
Collapse
Affiliation(s)
- Xinran Zhu
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Huashan Hospital, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 201203, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 201203, China
| | - Ulrich Lächelt
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, 1090, Austria
| | - Rongqin Huang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Huashan Hospital, School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
2
|
Roy M, Alix C, Burlaud-Gaillard J, Fouan D, Raoul W, Bouakaz A, Blanchard E, Lecomte T, Viaud-Massuard MC, Sasaki N, Serrière S, Escoffre JM. Delivery of Anticancer Drugs Using Microbubble-Assisted Ultrasound in a 3D Spheroid Model. Mol Pharm 2024; 21:831-844. [PMID: 38174896 DOI: 10.1021/acs.molpharmaceut.3c00921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Tumor spheroids are promising three-dimensional (3D) in vitro tumor models for the evaluation of drug delivery methods. The design of noninvasive and targeted drug methods is required to improve the intratumoral bioavailability of chemotherapeutic drugs and reduce their adverse off-target effects. Among such methods, microbubble-assisted ultrasound (MB-assisted US) is an innovative modality for noninvasive targeted drug delivery. The aim of the present study is to evaluate the efficacy of this US modality for the delivery of bleomycin, doxorubicin, and irinotecan in colorectal cancer (CRC) spheroids. MB-assisted US permeabilized the CRC spheroids to propidium iodide, which was used as a drug model without affecting their growth and viability. Histological analysis and electron microscopy revealed that MB-assisted US affected only the peripheral layer of the CRC spheroids. The acoustically mediated bleomycin delivery induced a significant decrease in CRC spheroid growth in comparison to spheroids treated with bleomycin alone. However, this US modality did not improve the therapeutic efficacy of doxorubicin and irinotecan on CRC spheroids. In conclusion, this study demonstrates that tumor spheroids are a relevant approach to evaluate the efficacy of MB-assisted US for the delivery of chemotherapeutics.
Collapse
Affiliation(s)
- Marie Roy
- UMR 1253, iBrain, Université de Tours, Inserm, 37032 Tours, France
| | - Corentin Alix
- UMR 1253, iBrain, Université de Tours, Inserm, 37032 Tours, France
| | - Julien Burlaud-Gaillard
- Inserm U1259, Université de Tours et CHRU de Tours & Plateforme IBiSA des Microscopies, PPF ASB, CHRU de Tours, 37032 Tours, France
| | - Damien Fouan
- UMR 1253, iBrain, Université de Tours, Inserm, 37032 Tours, France
| | - William Raoul
- Inserm UMR 1069, Nutrition Croissance et Cancer (N2C), Université de Tours, 37032 Tours, France
| | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, 37032 Tours, France
| | - Emmanuelle Blanchard
- Inserm U1259, Université de Tours et CHRU de Tours & Plateforme IBiSA des Microscopies, PPF ASB, CHRU de Tours, 37032 Tours, France
| | - Thierry Lecomte
- Inserm UMR 1069, Nutrition Croissance et Cancer (N2C), Université de Tours, 37032 Tours, France
- Department of Hepato-Gastroenterology & Digestive Oncology, CHRU de Tours, 37000 Tours, France
| | | | - Noboru Sasaki
- Laboratory of Veterinary Internal Medicine, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, 060-0818 Sapporo, Japan
| | - Sophie Serrière
- UMR 1253, iBrain, Université de Tours, Inserm, 37032 Tours, France
- Département d'Imagerie Préclinique, Plateforme Scientifique et Technique Analyse des Systèmes Biologiques, Université de Tours, 37032 Tours, France
| | | |
Collapse
|
3
|
Ikeda-Motonakano R, Hirabayashi-Nishimuta F, Yada N, Yamasaki R, Nagai-Yoshioka Y, Usui M, Nakazawa K, Yoshiga D, Yoshioka I, Ariyoshi W. Fabrication of a Three-Dimensional Spheroid Culture System for Oral Squamous Cell Carcinomas Using a Microfabricated Device. Cancers (Basel) 2023; 15:5162. [PMID: 37958336 PMCID: PMC10649954 DOI: 10.3390/cancers15215162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Cancer stem cells (CSCs) are considered to be responsible for recurrence, metastasis, and resistance to treatment in many types of cancers; therefore, new treatment strategies targeting CSCs are attracting attention. In this study, we fabricated a polyethylene glycol-tagged microwell device that enabled spheroid formation from human oral squamous carcinoma cells. HSC-3 and Ca9-22 cells cultured in the microwell device aggregated and generated a single spheroid per well within 24-48 h. The circular shape and smooth surface of spheroids were maintained for up to five days, and most cells comprising the spheroids were Calcein AM-positive viable cells. Interestingly, the mRNA expression of CSC markers (Cd44, Oct4, Nanog, and Sox2) were significantly higher in the spheroids than in the monolayer cultures. CSC marker-positive cells were observed throughout the spheroids. Moreover, resistance to cisplatin was enhanced in spheroid-cultured cells compared to that in the monolayer-cultured cells. Furthermore, some CSC marker genes were upregulated in HSC-3 and Ca9-22 cells that were outgrown from spheroids. In xenograft model, the tumor growth in the spheroid implantation group was comparable to that in the monolayer culture group. These results suggest that our spheroid culture system may be a high-throughput tool for producing uniform CSCs in large numbers from oral cancer cells.
Collapse
Affiliation(s)
- Reiko Ikeda-Motonakano
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan; (R.I.-M.); (R.Y.); (Y.N.-Y.)
- Division of Oral Medicine, Department of Science of Physical Function, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan; (F.H.-N.); (D.Y.); (I.Y.)
| | - Fumika Hirabayashi-Nishimuta
- Division of Oral Medicine, Department of Science of Physical Function, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan; (F.H.-N.); (D.Y.); (I.Y.)
| | - Naomi Yada
- Division of Oral Pathology, Department of Health Promotion, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan;
| | - Ryota Yamasaki
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan; (R.I.-M.); (R.Y.); (Y.N.-Y.)
| | - Yoshie Nagai-Yoshioka
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan; (R.I.-M.); (R.Y.); (Y.N.-Y.)
| | - Michihiko Usui
- Division of Periodontology, Department of Oral Function, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan;
| | - Kohji Nakazawa
- Department of Life and Environment Engineering, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka 808-0135, Japan;
| | - Daigo Yoshiga
- Division of Oral Medicine, Department of Science of Physical Function, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan; (F.H.-N.); (D.Y.); (I.Y.)
| | - Izumi Yoshioka
- Division of Oral Medicine, Department of Science of Physical Function, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan; (F.H.-N.); (D.Y.); (I.Y.)
| | - Wataru Ariyoshi
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan; (R.I.-M.); (R.Y.); (Y.N.-Y.)
| |
Collapse
|
4
|
Graceffa V. Intracellular protein delivery: New insights into the therapeutic applications and emerging technologies. Biochimie 2023; 213:82-99. [PMID: 37209808 DOI: 10.1016/j.biochi.2023.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023]
Abstract
The inability to cross the plasma membranes traditionally limited the therapeutic use of recombinant proteins. However, in the last two decades, novel technologies made delivering proteins inside the cells possible. This allowed researchers to unlock intracellular targets, once considered 'undruggable', bringing a new research area to emerge. Protein transfection systems display a large potential in a plethora of applications. However, their modality of action is often unclear, and cytotoxic effects are elevated, whereas experimental conditions to increase transfection efficacy and cell viability still need to be identified. Furthermore, technical complexity often limits in vivo experimentation, while challenging industrial and clinical translation. This review highlights the applications of protein transfection technologies, and then critically discuss the current methodologies and their limitations. Physical membrane perforation systems are compared to systems exploiting cellular endocytosis. Research evidence of the existence of either extracellular vesicles (EVs) or cell-penetrating peptides (CPPs)- based systems, that circumvent the endosomal systems is critically analysed. Commercial systems, novel solid-phase reverse protein transfection systems, and engineered living intracellular bacteria-based mechanisms are finally described. This review ultimately aims at finding new methodologies and possible applications of protein transfection systems, while helping the development of an evidence-based research approach.
Collapse
Affiliation(s)
- Valeria Graceffa
- Cellular Health and Toxicology Research Group (CHAT), Centre for Mathematical Modelling and Intelligent Systems for Health and Environment (MISHE), Atlantic Technological University (ATU), Sligo, Ireland.
| |
Collapse
|
5
|
Stella GM, Lettieri S, Piloni D, Ferrarotti I, Perrotta F, Corsico AG, Bortolotto C. Smart Sensors and Microtechnologies in the Precision Medicine Approach against Lung Cancer. Pharmaceuticals (Basel) 2023; 16:1042. [PMID: 37513953 PMCID: PMC10385174 DOI: 10.3390/ph16071042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/23/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND AND RATIONALE The therapeutic interventions against lung cancer are currently based on a fully personalized approach to the disease with considerable improvement of patients' outcome. Alongside continuous scientific progresses and research investments, massive technologic efforts, innovative challenges, and consolidated achievements together with research investments are at the bases of the engineering and manufacturing revolution that allows a significant gain in clinical setting. AIM AND METHODS The scope of this review is thus to focus, rather than on the biologic traits, on the analysis of the precision sensors and novel generation materials, as semiconductors, which are below the clinical development of personalized diagnosis and treatment. In this perspective, a careful revision and analysis of the state of the art of the literature and experimental knowledge is presented. RESULTS Novel materials are being used in the development of personalized diagnosis and treatment for lung cancer. Among them, semiconductors are used to analyze volatile cancer compounds and allow early disease diagnosis. Moreover, they can be used to generate MEMS which have found an application in advanced imaging techniques as well as in drug delivery devices. CONCLUSIONS Overall, these issues represent critical issues only partially known and generally underestimated by the clinical community. These novel micro-technology-based biosensing devices, based on the use of molecules at atomic concentrations, are crucial for clinical innovation since they have allowed the recent significant advances in cancer biology deciphering as well as in disease detection and therapy. There is an urgent need to create a stronger dialogue between technologists, basic researchers, and clinicians to address all scientific and manufacturing efforts towards a real improvement in patients' outcome. Here, great attention is focused on their application against lung cancer, from their exploitations in translational research to their application in diagnosis and treatment development, to ensure early diagnosis and better clinical outcomes.
Collapse
Affiliation(s)
- Giulia Maria Stella
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Sara Lettieri
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Davide Piloni
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Ilaria Ferrarotti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Fabio Perrotta
- Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", 80131 Napoli, Italy
- U.O.C. Clinica Pneumologica "L. Vanvitelli", A.O. dei Colli, Ospedale Monaldi, 80131 Napoli, Italy
| | - Angelo Guido Corsico
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Chandra Bortolotto
- Department of Clinical-Surgical, Diagnostic and Paediatric Sciences, University of Pavia Medical School, 27100 Pavia, Italy
- Department of Diagnostic Services and Imaging, Unit of Radiology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
6
|
Vyas K, Rathod M, Patel MM. Insight on nano drug delivery systems with targeted therapy in treatment of oral cancer. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 49:102662. [PMID: 36746272 DOI: 10.1016/j.nano.2023.102662] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/18/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Oral cancer is a type of cancer that develops in the mouth and is one of the deadliest malignancies in the world. Currently surgical, radiation therapy, and chemotherapy are most common treatments. Better treatment and early detection strategies are required. Chemotherapeutic drugs fail frequently due to toxicity and poor tumor targeting. There are high chances of failure of chemotherapeutic drugs due to toxicity. Active, passive, and immunity-targeting techniques are devised for tumor-specific activity. Nanotechnology-based drug delivery systems are the best available solution and important for precise targeting. Nanoparticles, liposomes, exosomes, and cyclodextrins are nano-based carriers for drug delivery. Nanotechnology is being used to develop new techniques such as intratumoral injections, microbubble mediated ultrasonic therapy, phototherapies, and site-specific delivery. This systematic review delves into the details of such targeted and nano-based drug delivery systems in order to improve patient health and survival rates in oral cancer.
Collapse
Affiliation(s)
- Kunj Vyas
- Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad 382481, Gujarat, India
| | - Maharshsinh Rathod
- Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad 382481, Gujarat, India
| | - Mayur M Patel
- Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad 382481, Gujarat, India.
| |
Collapse
|
7
|
Tabata H, Koyama D, Matsukawa M, Krafft MP, Yoshida K. Concentration-Dependent Viscoelasticity of Poloxamer-Shelled Microbubbles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:433-441. [PMID: 36580034 DOI: 10.1021/acs.langmuir.2c02690] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The oscillation of shelled microbubbles during exposure to ultrasound is influenced by the mechanical properties of the shell components. The oscillation behavior of bubbles coated with various phospholipids and other amphiphiles has been studied. However, there have been few investigations of how the adsorption conditions of the shell molecules relate to the viscoelastic properties of the shell and influence the oscillation behavior of the bubbles. In the present study, we investigated the oscillation characteristics of microbubbles coated with a poloxamer surfactant, that is, Pluronic F-68, at several concentrations after the adsorption kinetics of the surfactant at the gas-water interface had reached equilibrium. The dilatational viscoelasticity of the shell during exposure to ultrasound was analyzed in the frequency domain from the attenuation characteristics of the acoustic pulses propagated in the bubble suspension. At Pluronic F-68 concentrations lower than 2.0 × 10-2 mol L-1, the attenuation characteristics typically exhibited a sharp peak. At concentrations higher than 2.0 × 10-2 mol L-1, the peak flattened. The dilatational elasticity and viscosity of the shell were estimated by fitting the theoretical model to the experimental values, which revealed that both the elasticity and viscosity increased markedly at approximately 2.0 × 10-2 mol L-1. This suggests that the adsorption properties of Pluronic F-68 strongly affect the oscillation characteristics of microbubbles of a size suitable for medical ultrasound diagnostics.
Collapse
Affiliation(s)
- Hiraku Tabata
- Faculty of Science and Engineering, Doshisha University, 1-3 Tataramiyakodani, Kyotanabe, Kyoto610-0321, Japan
| | - Daisuke Koyama
- Faculty of Science and Engineering, Doshisha University, 1-3 Tataramiyakodani, Kyotanabe, Kyoto610-0321, Japan
| | - Mami Matsukawa
- Faculty of Science and Engineering, Doshisha University, 1-3 Tataramiyakodani, Kyotanabe, Kyoto610-0321, Japan
| | - Marie Pierre Krafft
- Institut Charles Sadron (CNRS), University of Strasbourg, 23 rue du Loess, Strasbourg67034, France
| | - Kenji Yoshida
- Center for Frontier Medical Engineering, Chiba University, 1-33 Yayoicho, Inage-ku, Chiba263-8522, Japan
| |
Collapse
|
8
|
Evaluation of Liposome-Loaded Microbubbles as a Theranostic Tool in a Murine Collagen-Induced Arthritis Model. Sci Pharm 2022. [DOI: 10.3390/scipharm90010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by severe inflammation of the synovial tissue. Here, we assess the feasibility of liposome-loaded microbubbles as theranostic agents in a murine arthritis model. First, contrast-enhanced ultrasound (CEUS) was used to quantify neovascularization in this model since CEUS is well-established for RA diagnosis in humans. Next, the potential of liposome-loaded microbubbles and ultrasound (US) to selectively enhance liposome delivery to the synovium was evaluated with in vivo fluorescence imaging. This procedure is made very challenging by the presence of hard joints and by the limited lifetime of the microbubbles. The inflamed knee joints were exposed to therapeutic US after intravenous injection of liposome-loaded microbubbles. Loaded microbubbles were found to be quickly captured by the liver. This resulted in fast clearance of attached liposomes while free and long-circulating liposomes were able to accumulate over time in the inflamed joints. Our observations show that murine arthritis models are not well-suited for evaluating the potential of microbubble-mediated drug delivery in joints given: (i) restricted microbubble passage in murine synovial vasculature and (ii) limited control over the exact ultrasound conditions in situ given the much shorter length scale of the murine joints as compared to the therapeutic wavelength.
Collapse
|
9
|
SonoVue ® vs. Sonazoid™ vs. Optison™: Which Bubble Is Best for Low-Intensity Sonoporation of Pancreatic Ductal Adenocarcinoma? Pharmaceutics 2022; 14:pharmaceutics14010098. [PMID: 35056994 PMCID: PMC8777813 DOI: 10.3390/pharmaceutics14010098] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/15/2021] [Accepted: 12/28/2021] [Indexed: 02/04/2023] Open
Abstract
The use of ultrasound and microbubbles to enhance therapeutic efficacy (sonoporation) has shown great promise in cancer therapy from in vitro to ongoing clinical studies. The fastest bench-to-bedside translation involves the use of ultrasound contrast agents (microbubbles) and clinical diagnostic scanners. Despite substantial research in this field, it is currently not known which of these microbubbles result in the greatest enhancement of therapy within the applied conditions. Three microbubble formulations-SonoVue®, Sonazoid™, and Optison™-were physiochemically and acoustically characterized. The microbubble response to the ultrasound pulses used in vivo was simulated via a Rayleigh-Plesset type equation. The three formulations were compared in vitro for permeabilization efficacy in three different pancreatic cancer cell lines, and in vivo, using an orthotopic pancreatic cancer (PDAC) murine model. The mice were treated using one of the three formulations exposed to ultrasound from a GE Logiq E9 and C1-5 ultrasound transducer. Characterisation of the microbubbles showed a rapid degradation in concentration, shape, and/or size for both SonoVue® and Optison™ within 30 min of reconstitution/opening. Sonazoid™ showed no degradation after 1 h. Attenuation measurements indicated that SonoVue® was the softest bubble followed by Sonazoid™ then Optison™. Sonazoid™ emitted nonlinear ultrasound at the lowest MIs followed by Optison™, then SonoVue®. Simulations indicated that SonoVue® would be the most effective bubble using the evaluated ultrasound conditions. This was verified in the pre-clinical PDAC model demonstrated by improved survival and largest tumor growth inhibition. In vitro results indicated that the best microbubble formulation depends on the ultrasound parameters and concentration used, with SonoVue® being best at lower intensities and Sonazoid™ at higher intensities.
Collapse
|
10
|
V B, Femina T A, Iyengar D, K A, Ravi M. Approaches for Head and Neck Cancer Research - Current Status and the Way Forward. Cancer Invest 2021; 40:151-172. [PMID: 34806936 DOI: 10.1080/07357907.2021.2009850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Head and neck cancers (HNCs) are seeing an increasing trend in their prevalence among both genders and are the seventh most common cancer type occurring at the global level. Studies addressing both the cancer cell physiology and individual differences in response to a specific treatment modality should be understood for arriving at effective treatment and management of the HNCs. In this article, we discuss the trends in HNC research and their various approaches starting from 2D in vitro models, which are the traditional experimental materials to recently established Cancer-Tissue Originated Spheroids (CTOS) distinctly contributing towards personalized or precision medicine.
Collapse
Affiliation(s)
- Barghavi V
- Department of Human Genetics, Faculty of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Arokia Femina T
- Department of Human Genetics, Faculty of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - DivyaSowrirajan Iyengar
- Department of Human Genetics, Faculty of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Archana K
- Department of Human Genetics, Faculty of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Maddaly Ravi
- Department of Human Genetics, Faculty of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| |
Collapse
|
11
|
de Maar JS, Rousou C, van Elburg B, Vos HJ, Lajoinie GPR, Bos C, Moonen CTW, Deckers R. Ultrasound-Mediated Drug Delivery With a Clinical Ultrasound System: In Vitro Evaluation. Front Pharmacol 2021; 12:768436. [PMID: 34737709 PMCID: PMC8560689 DOI: 10.3389/fphar.2021.768436] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/05/2021] [Indexed: 12/22/2022] Open
Abstract
Chemotherapy efficacy is often reduced by insufficient drug uptake in tumor cells. The combination of ultrasound and microbubbles (USMB) has been shown to improve drug delivery and to enhance the efficacy of several drugs in vitro and in vivo, through effects collectively known as sonopermeation. However, clinical translation of USMB therapy is hampered by the large variety of (non-clinical) US set-ups and US parameters that are used in these studies, which are not easily translated to clinical practice. In order to facilitate clinical translation, the aim of this study was to prove that USMB therapy using a clinical ultrasound system (Philips iU22) in combination with clinically approved microbubbles (SonoVue) leads to efficient in vitro sonopermeation. To this end, we measured the efficacy of USMB therapy for different US probes (S5-1, C5-1 and C9-4) and US parameters in FaDu cells. The US probe with the lowest central frequency (i.e. 1.6 MHz for S5-1) showed the highest USMB-induced intracellular uptake of the fluorescent dye SYTOX™ Green (SG). These SG uptake levels were comparable to or even higher than those obtained with a custom-built US system with optimized US parameters. Moreover, USMB therapy with both the clinical and the custom-built US system increased the cytotoxicity of the hydrophilic drug bleomycin. Our results demonstrate that a clinical US system can be used to perform USMB therapy as efficiently as a single-element transducer set-up with optimized US parameters. Therefore, future trials could be based on these clinical US systems, including validated US parameters, in order to accelerate successful translation of USMB therapy.
Collapse
Affiliation(s)
- Josanne S de Maar
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Charis Rousou
- Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Benjamin van Elburg
- Physics of Fluids Group, MIRA Institute of Biomedical Technology and Technical Medicine, University of Twente, Enschede, Netherlands
| | - Hendrik J Vos
- Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, Netherlands
| | - Guillaume P R Lajoinie
- Physics of Fluids Group, MIRA Institute of Biomedical Technology and Technical Medicine, University of Twente, Enschede, Netherlands
| | - Clemens Bos
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Chrit T W Moonen
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Roel Deckers
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
12
|
Omata D, Munakata L, Maruyama K, Suzuki R. Enhanced Vascular Permeability by Microbubbles and Ultrasound in Drug Delivery. Biol Pharm Bull 2021; 44:1391-1398. [PMID: 34602547 DOI: 10.1248/bpb.b21-00453] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ultrasound and microbubbles, an ultrasound contrast agent, have recently increased attention to developing novel drug delivery systems. Ultrasound exposure can induce mechanical effects derived from microbubbles behaviors such as an expansion, contraction, and collapse depending on ultrasound conditions. These mechanical effects induce several biological effects, including enhancement of vascular permeability. For drug delivery, one promising approach is enhancing vascular permeability using ultrasound and microbubbles, resulting in improved drug transport to targeted tissues. This approach is applied to several tissues and drugs to cure diseases. This review describes the enhancement of vascular permeability by ultrasound and microbubbles and its therapeutic application, including our recent study. We also discuss the current situation of the field and its potential future perspectives.
Collapse
Affiliation(s)
- Daiki Omata
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University
| | - Lisa Munakata
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University
| | - Kazuo Maruyama
- Laboratory of Theranostics, Faculty of Pharma-Science, Teikyo University.,Advanced Comprehensive Research Organization (ACRO), Teikyo University
| | - Ryo Suzuki
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University.,Advanced Comprehensive Research Organization (ACRO), Teikyo University
| |
Collapse
|
13
|
Deprez J, Lajoinie G, Engelen Y, De Smedt SC, Lentacker I. Opening doors with ultrasound and microbubbles: Beating biological barriers to promote drug delivery. Adv Drug Deliv Rev 2021; 172:9-36. [PMID: 33705877 DOI: 10.1016/j.addr.2021.02.015] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Apart from its clinical use in imaging, ultrasound has been thoroughly investigated as a tool to enhance drug delivery in a wide variety of applications. Therapeutic ultrasound, as such or combined with cavitating nuclei or microbubbles, has been explored to cross or permeabilize different biological barriers. This ability to access otherwise impermeable tissues in the body makes the combination of ultrasound and therapeutics very appealing to enhance drug delivery in situ. This review gives an overview of the most important biological barriers that can be tackled using ultrasound and aims to provide insight on how ultrasound has shown to improve accessibility as well as the biggest hurdles. In addition, we discuss the clinical applicability of therapeutic ultrasound with respect to the main challenges that must be addressed to enable the further progression of therapeutic ultrasound towards an effective, safe and easy-to-use treatment tailored for drug delivery in patients.
Collapse
Affiliation(s)
- J Deprez
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - G Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, P.O. Box 217, 7500 AE Enschede, Netherlands
| | - Y Engelen
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - S C De Smedt
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - I Lentacker
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
14
|
Choromańska A, Chwiłkowska A, Kulbacka J, Baczyńska D, Rembiałkowska N, Szewczyk A, Michel O, Gajewska-Naryniecka A, Przystupski D, Saczko J. Modifications of Plasma Membrane Organization in Cancer Cells for Targeted Therapy. Molecules 2021; 26:1850. [PMID: 33806009 PMCID: PMC8037978 DOI: 10.3390/molecules26071850] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/18/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Modifications of the composition or organization of the cancer cell membrane seem to be a promising targeted therapy. This approach can significantly enhance drug uptake or intensify the response of cancer cells to chemotherapeutics. There are several methods enabling lipid bilayer modifications, e.g., pharmacological, physical, and mechanical. It is crucial to keep in mind the significance of drug resistance phenomenon, ion channel and specific receptor impact, and lipid bilayer organization in planning the cell membrane-targeted treatment. In this review, strategies based on cell membrane modulation or reorganization are presented as an alternative tool for future therapeutic protocols.
Collapse
Affiliation(s)
- Anna Choromańska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (J.K.); (D.B.); (N.R.); (A.S.); (O.M.); (A.G.-N.); (J.S.)
| | - Agnieszka Chwiłkowska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (J.K.); (D.B.); (N.R.); (A.S.); (O.M.); (A.G.-N.); (J.S.)
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (J.K.); (D.B.); (N.R.); (A.S.); (O.M.); (A.G.-N.); (J.S.)
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (J.K.); (D.B.); (N.R.); (A.S.); (O.M.); (A.G.-N.); (J.S.)
| | - Nina Rembiałkowska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (J.K.); (D.B.); (N.R.); (A.S.); (O.M.); (A.G.-N.); (J.S.)
| | - Anna Szewczyk
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (J.K.); (D.B.); (N.R.); (A.S.); (O.M.); (A.G.-N.); (J.S.)
| | - Olga Michel
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (J.K.); (D.B.); (N.R.); (A.S.); (O.M.); (A.G.-N.); (J.S.)
| | - Agnieszka Gajewska-Naryniecka
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (J.K.); (D.B.); (N.R.); (A.S.); (O.M.); (A.G.-N.); (J.S.)
| | - Dawid Przystupski
- Department of Paediatric Bone Marrow Transplantation, Oncology and Haematology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (J.K.); (D.B.); (N.R.); (A.S.); (O.M.); (A.G.-N.); (J.S.)
| |
Collapse
|
15
|
The prospects of nanotherapeutic approaches for targeting tumor-associated macrophages in oral cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 34:102371. [PMID: 33662592 DOI: 10.1016/j.nano.2021.102371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 11/23/2022]
Abstract
OSCC (oral squamous cell carcinoma) is currently one of the most formidable cancers plagued by challenges like low overall survivability, lymph node associated metastasis, drug resistance, and poor diagnostics. The tumor microenvironment (TME) and its constituent stromal elements are crucial modulators of tumor growth and treatment response, more specifically so with regards to resident tumor associated macrophages (TAMs) and their liaison with the different stromal elements in the tumor niche (Figure 1). Interestingly, there isn't much information on TAM-targeted nanotherapy in OSCC where the first line of therapeutics for oral cancer is surgery with other therapeutics such as chemo- and radiotherapy acting only as adjuvant therapy for oral cancer. In the face of this real time situation, there have been some successful attempts at targeted therapy for OSCC cells and we believe they might elicit favorable responses against TAMs as well. Demanding our immediate attention, this review intends to provide a glimpse of the prevailing anti-TAM treatment strategies, which present great prospect for an uncharted territory like OSCC.
Collapse
|
16
|
Ultrasound and microbubbles (USMB) potentiated doxorubicin penetration and distribution in 3D breast tumour spheroids. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
17
|
Haugse R, Langer A, Murvold ET, Costea DE, Gjertsen BT, Gilja OH, Kotopoulis S, Ruiz de Garibay G, McCormack E. Low-Intensity Sonoporation-Induced Intracellular Signalling of Pancreatic Cancer Cells, Fibroblasts and Endothelial Cells. Pharmaceutics 2020; 12:pharmaceutics12111058. [PMID: 33171947 PMCID: PMC7694645 DOI: 10.3390/pharmaceutics12111058] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
The use of ultrasound (US) and microbubbles (MB), usually referred to as sonoporation, has great potential to increase the efficacy of chemotherapy. However, the molecular mechanisms that mediate sonoporation response are not well-known, and recent research suggests that cell stress induced by US + MBs may contribute to the treatment benefit. Furthermore, there is a growing understanding that the effects of US + MBs are beyond only the cancer cells and involves the tumour vasculature and microenvironment. We treated pancreatic cancer cells (MIA PaCa-2) and stromal cells, fibroblasts (BJ) and human umbilical vein endothelial cells (HUVECs), with US ± MB, and investigated the extent of uptake of cell impermeable dye (calcein, by flow cytometry), viability (cell count, Annexin/PI and WST-1 assays) and activation of a number of key proteins in important intracellular signalling pathways immediately and 2 h after sonoporation (phospho flow cytometry). Different cell types responded differently to US ± MBs in all these aspects. In general, sonoporation induces immediate, transient activation of MAP-kinases (p38, ERK1/2), and an increase in phosphorylation of ribosomal protein S6 together with dephosphorylation of 4E-BP1. The sonoporation stress-response resembles cellular responses to electroporation and pore-forming toxins in membrane repair and restoring cellular homeostasis, and may be exploited therapeutically. The stromal cells were more sensitive to sonoporation than tumoural cells, and further efforts in optimising sonoporation-enhanced therapy should be targeted at the microenvironment.
Collapse
Affiliation(s)
- Ragnhild Haugse
- Centre for Pharmacy, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway;
- Department of Quality and Development, Hospital Pharmacies Enterprise in Western Norway, Møllendalsbakken 9, 5021 Bergen, Norway
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway; (A.L.); (D.E.C.); (B.T.G.); (G.R.d.G.)
| | - Anika Langer
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway; (A.L.); (D.E.C.); (B.T.G.); (G.R.d.G.)
| | - Elisa Thodesen Murvold
- KinN Therapeutics AS, Jonas Lies vei 91B, 5021 Bergen, Norway;
- Department of Clinical Medicine, The University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway; (O.H.G.); (S.K.)
| | - Daniela Elena Costea
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway; (A.L.); (D.E.C.); (B.T.G.); (G.R.d.G.)
- Department of Clinical Medicine, The University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway; (O.H.G.); (S.K.)
| | - Bjørn Tore Gjertsen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway; (A.L.); (D.E.C.); (B.T.G.); (G.R.d.G.)
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Jonas Lies vei 65, 5021 Bergen, Norway
| | - Odd Helge Gilja
- Department of Clinical Medicine, The University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway; (O.H.G.); (S.K.)
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Jonas Lies vei 65, 5021 Bergen, Norway
| | - Spiros Kotopoulis
- Department of Clinical Medicine, The University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway; (O.H.G.); (S.K.)
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Jonas Lies vei 65, 5021 Bergen, Norway
- EXACT Therapeutics AS, Ullernchausseen 64, 0379 Oslo, Norway
| | - Gorka Ruiz de Garibay
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway; (A.L.); (D.E.C.); (B.T.G.); (G.R.d.G.)
| | - Emmet McCormack
- Centre for Pharmacy, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway;
- Department of Quality and Development, Hospital Pharmacies Enterprise in Western Norway, Møllendalsbakken 9, 5021 Bergen, Norway
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway; (A.L.); (D.E.C.); (B.T.G.); (G.R.d.G.)
- KinN Therapeutics AS, Jonas Lies vei 91B, 5021 Bergen, Norway;
- Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway
- Correspondence:
| |
Collapse
|
18
|
Kukkar D, Kukkar P, Kumar V, Hong J, Kim KH, Deep A. Recent advances in nanoscale materials for antibody-based cancer theranostics. Biosens Bioelectron 2020; 173:112787. [PMID: 33190049 DOI: 10.1016/j.bios.2020.112787] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/08/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
The quest for advanced management tools or options of various cancers has been on the rise to efficiently reduce their risks of mortality without the demerits of conventional treatments (e.g., undesirable side effects of the medications on non-target tissues, non-targeted distribution, slow clearance of the administered drugs, and the development of drug resistance over the duration of therapy). In this context, nanomaterials-antibody conjugates can offer numerous advantages in the development of cancer theranostics over conventional delivery systems (e.g., highly specific and enhanced biodistribution of the drug in targeted tissues, prolonged systemic circulation, low toxicity, and minimally invasive molecular imaging). This review comprehensively discusses and evaluates recent advances in the application of nanomaterial-antibody bioconjugates for cancer theranostics for the further advancement in the control of diverse cancerous diseases. Further, discussion is expanded to cover the various challenges and limitations associated with the design and development of nanomaterial-antibody conjugates applicable towards better management of cancer.
Collapse
Affiliation(s)
- Deepak Kukkar
- Department of Nanotechnology, Sri Guru Granth Sahib World University, Fatehgarh Sahib, Punjab, 140406, India
| | - Preeti Kukkar
- Department of Chemistry, Mata Gujri College, Fatehgarh Sahib, Punjab, 140406, India
| | - Vanish Kumar
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Punjab, 140306, India
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, Seoul, 04763 Republic of Korea.
| | - Akash Deep
- Central Scientific Instruments Organization (CSIR-CSIO), Sector 30 C, Chandigarh, 160030, India.
| |
Collapse
|
19
|
Escoffre JM, Campomanes P, Tarek M, Bouakaz A. New insights on the role of ROS in the mechanisms of sonoporation-mediated gene delivery. ULTRASONICS SONOCHEMISTRY 2020; 64:104998. [PMID: 32062534 DOI: 10.1016/j.ultsonch.2020.104998] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 01/13/2020] [Accepted: 02/03/2020] [Indexed: 06/10/2023]
Abstract
Reactive oxygen species (ROS) are hypothesized to play a role in the sonoporation mechanisms. Nevertheless, the acoustical phenomenon behind the ROS production as well as the exact mechanisms of ROS action involved in the increased cell membrane permeability are still not fully understood. Therefore, we investigated the key processes occurring at the molecular level in and around microbubbles subjected to ultrasound using computational chemistry methods. To confirm the molecular simulation predictions, we measured the ROS production by exposing SonoVue® microbubbles (MBs) to ultrasound using biological assays. To investigate the role of ROS in cell membrane permeabilization, cells were subjected to ultrasound in presence of MBs and plasmid encoding reporter gene, and the transfection level was assessed using flow cytometry. The molecular simulations showed that under sonoporation conditions, ROS can form inside the MBs. These radicals could easily diffuse through the MB shell toward the surrounding aqueous phase and participate in the permeabilization of nearby cell membranes. Experimental data confirmed that MBs favor spontaneous formation of a host of free radicals where HO was the main ROS species after US exposure. The presence of ROS scavengers/inhibitors during the sonoporation process decreased both the production of ROS and the subsequent transfection level without significant loss of cell viability. In conclusion, the exposure of MBs to ultrasound might be the origin of chemical effects, which play a role in the cell membrane permeabilization and in the in vitro gene delivery when generated in its proximity.
Collapse
Affiliation(s)
| | - Pablo Campomanes
- Laboratoire de Physique et Chimie Théoriques, UMR 7019, Université de Lorraine, CNRS, Nancy F-54000, France
| | - Mounir Tarek
- Laboratoire de Physique et Chimie Théoriques, UMR 7019, Université de Lorraine, CNRS, Nancy F-54000, France.
| | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
| |
Collapse
|
20
|
Tharkar P, Varanasi R, Wong WSF, Jin CT, Chrzanowski W. Nano-Enhanced Drug Delivery and Therapeutic Ultrasound for Cancer Treatment and Beyond. Front Bioeng Biotechnol 2019; 7:324. [PMID: 31824930 PMCID: PMC6883936 DOI: 10.3389/fbioe.2019.00324] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/28/2019] [Indexed: 12/24/2022] Open
Abstract
While ultrasound is most widely known for its use in diagnostic imaging, the energy carried by ultrasound waves can be utilized to influence cell function and drug delivery. Consequently, our ability to use ultrasound energy at a given intensity unlocks the opportunity to use the ultrasound for therapeutic applications. Indeed, in the last decade ultrasound-based therapies have emerged with promising treatment modalities for several medical conditions. More recently, ultrasound in combination with nanomedicines, i.e., nanoparticles, has been shown to have substantial potential to enhance the efficacy of many treatments including cancer, Alzheimer disease or osteoarthritis. The concept of ultrasound combined with drug delivery is still in its infancy and more research is needed to unfold the mechanisms and interactions of ultrasound with different nanoparticles types and with various cell types. Here we present the state-of-art in ultrasound and ultrasound-assisted drug delivery with a particular focus on cancer treatments. Notably, this review discusses the application of high intensity focus ultrasound for non-invasive tumor ablation and immunomodulatory effects of ultrasound, as well as the efficacy of nanoparticle-enhanced ultrasound therapies for different medical conditions. Furthermore, this review presents safety considerations related to ultrasound technology and gives recommendations in the context of system design and operation.
Collapse
Affiliation(s)
- Priyanka Tharkar
- Faculty of Medicine and Health, Sydney School of Pharmacy, Sydney Nano Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Ramya Varanasi
- Faculty of Medicine and Health, Sydney School of Pharmacy, Sydney Nano Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Wu Shun Felix Wong
- School of Women's and Children's Health, University of New South Wales, Sydney, NSW, Australia
| | - Craig T Jin
- Faculty of Engineering, School of Electrical and Information Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Wojciech Chrzanowski
- Faculty of Medicine and Health, Sydney School of Pharmacy, Sydney Nano Institute, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
21
|
Wang L, Lu H, Gao Q, Yuan C, Ding F, Li J, Zhang D, Ou X. A multifunctional theranostic contrast agent for ultrasound/near infrared fluorescence imaging-based tumor diagnosis and ultrasound-triggered combined photothermal and gene therapy. Acta Biomater 2019; 99:373-386. [PMID: 31525534 DOI: 10.1016/j.actbio.2019.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 08/23/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Encapsulated microbubbles (MBs) have been reported as new theranostic carriers for simultaneous imaging and ultrasound (US)-triggered therapy. Here, we designed a dual-modality US/NIRF contrast agent and extended its applications from image contrast enhancement to combined diagnosis and therapy with US-directed and site-specific targeting. METHODS Gold nanorods (AuNRs) resonant at 880 nm together with the NIR797 dye were first encapsulated in lipid-shelled MBs to construct fluorescent gold microbubbles (NIR797/AuMBs) via thin film hydration and mechanical shaking in the presence of sulfur hexafluoride (SF6) gas. Then, polyethylenimine (PEI)-DNA complexes were electrostatically conjugated onto the surface of the NIR797/AuMBs, forming theranostic encapsulated MBs (PEI-DNA/NIR797/AuMBs). The potential of the PEI-DNA/NIR797/AuMBs for use as a dual-modality contrast enhancement agent was evaluated in vitro and in vivo. The antitumor effect of US/NIR laser irradiation mediating double-fusion suicide gene and photothermal therapy was also investigated using Bel-7402 cells and xenografts. RESULTS The developed theranostic AuMB complexes could not only provide excellent US and NIRF imaging to detect tumors but also serve as an efficient US-triggered carrier for gene delivery and photothermal ablation of tumors in xenografted nude mice. And US + laser exposure group showed a much higher rate of cell inhibition, apoptosis and necrosis as well as a higher Bel-7402 xenograft inhibition rate than the single gene therapy or single exposure (US or laser) group. CONCLUSIONS PEI-DNA/NIR797/AuMBs would be of great value for providing more comprehensive diagnostic information and to guide more accurate and effective synergistic cancer therapy. STATEMENT OF SIGNIFICANCE This is an original paper focusing on developing a dual-modality US/NIRF contrast agent and extended its applications from image contrast enhancement to combined diagnosis and therapy with US-directed and site-specific targeting. The developed theranostic AuMB complexes could not only provide excellent US and NIRF imaging to detect tumors but also serve as an efficient US-triggered carrier for gene delivery and photothermal ablation of tumors in xenografted nude mice. PEI-DNA/NIR797/AuMBs would be of great value for providing more comprehensive diagnostic information and to guide more accurate and effective synergistic cancer therapy.
Collapse
|
22
|
Bressand D, Novell A, Girault A, Raoul W, Fromont-Hankard G, Escoffre JM, Lecomte T, Bouakaz A. Enhancing Nab-Paclitaxel Delivery Using Microbubble-Assisted Ultrasound in a Pancreatic Cancer Model. Mol Pharm 2019; 16:3814-3822. [DOI: 10.1021/acs.molpharmaceut.9b00416] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Diane Bressand
- UMR 1253, iBrain, Université de Tours, Inserm, 10 boulevard Tonnellé, 37032 Tours, France
- Department of Hepato-Gastroenterology and Digestive Cancerology, Université de Tours, EA7501 GICC, Team PATCH, CHRU de Tours, 10 boulevard Tonnellé, 37032 Tours, France
| | - Anthony Novell
- UMR 1253, iBrain, Université de Tours, Inserm, 10 boulevard Tonnellé, 37032 Tours, France
| | - Alban Girault
- Department of Hepato-Gastroenterology and Digestive Cancerology, Université de Tours, EA7501 GICC, Team PATCH, CHRU de Tours, 10 boulevard Tonnellé, 37032 Tours, France
| | - William Raoul
- Department of Hepato-Gastroenterology and Digestive Cancerology, Université de Tours, EA7501 GICC, Team PATCH, CHRU de Tours, 10 boulevard Tonnellé, 37032 Tours, France
| | - Gaëlle Fromont-Hankard
- Department of Pathological Anatomy and Cytology, Université de Tours, Inserm, UMR 1069, Nutrition, Croissance, Cancer, CHRU de Tours, 37032 Tours, France
| | - Jean-Michel Escoffre
- UMR 1253, iBrain, Université de Tours, Inserm, 10 boulevard Tonnellé, 37032 Tours, France
| | - Thierry Lecomte
- Department of Hepato-Gastroenterology and Digestive Cancerology, Université de Tours, EA7501 GICC, Team PATCH, CHRU de Tours, 10 boulevard Tonnellé, 37032 Tours, France
| | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, 10 boulevard Tonnellé, 37032 Tours, France
| |
Collapse
|
23
|
Ketabat F, Pundir M, Mohabatpour F, Lobanova L, Koutsopoulos S, Hadjiiski L, Chen X, Papagerakis P, Papagerakis S. Controlled Drug Delivery Systems for Oral Cancer Treatment-Current Status and Future Perspectives. Pharmaceutics 2019; 11:E302. [PMID: 31262096 PMCID: PMC6680655 DOI: 10.3390/pharmaceutics11070302] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 12/18/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC), which encompasses the oral cavity-derived malignancies, is a devastating disease causing substantial morbidity and mortality in both men and women. It is the most common subtype of the head and neck squamous cell carcinoma (HNSCC), which is ranked the sixth most common malignancy worldwide. Despite promising advancements in the conventional therapeutic approaches currently available for patients with oral cancer, many drawbacks are still to be addressed; surgical resection leads to permanent disfigurement, altered sense of self and debilitating physiological consequences, while chemo- and radio-therapies result in significant toxicities, all affecting patient wellbeing and quality of life. Thus, the development of novel therapeutic approaches or modifications of current strategies is paramount to improve individual health outcomes and survival, while early tumour detection remains a priority and significant challenge. In recent years, drug delivery systems and chronotherapy have been developed as alternative methods aiming to enhance the benefits of the current anticancer therapies, while minimizing their undesirable toxic effects on the healthy non-cancerous cells. Targeted drug delivery systems have the potential to increase drug bioavailability and bio-distribution at the site of the primary tumour. This review confers current knowledge on the diverse drug delivery methods, potential carriers (e.g., polymeric, inorganic, and combinational nanoparticles; nanolipids; hydrogels; exosomes) and anticancer targeted approaches for oral squamous cell carcinoma treatment, with an emphasis on their clinical relevance in the era of precision medicine, circadian chronobiology and patient-centred health care.
Collapse
Affiliation(s)
- Farinaz Ketabat
- Laboratory of Oral, Head and Neck Cancer - Personalized Diagnostics and Therapeutics, Department of Surgery - Division of Head and Neck Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
- Laboratory of Precision Oral Health and Chronobiology, College of Dentistry, University of Saskatchewan, Saskatoon, SK S7N 5E4, Canada
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7K 5A9, Canada
| | - Meenakshi Pundir
- Laboratory of Oral, Head and Neck Cancer - Personalized Diagnostics and Therapeutics, Department of Surgery - Division of Head and Neck Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
- Laboratory of Precision Oral Health and Chronobiology, College of Dentistry, University of Saskatchewan, Saskatoon, SK S7N 5E4, Canada
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7K 5A9, Canada
| | - Fatemeh Mohabatpour
- Laboratory of Oral, Head and Neck Cancer - Personalized Diagnostics and Therapeutics, Department of Surgery - Division of Head and Neck Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
- Laboratory of Precision Oral Health and Chronobiology, College of Dentistry, University of Saskatchewan, Saskatoon, SK S7N 5E4, Canada
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7K 5A9, Canada
| | - Liubov Lobanova
- Laboratory of Precision Oral Health and Chronobiology, College of Dentistry, University of Saskatchewan, Saskatoon, SK S7N 5E4, Canada
| | - Sotirios Koutsopoulos
- Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lubomir Hadjiiski
- Departmnet of Radiology, School of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiongbiao Chen
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7K 5A9, Canada
- Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK S7K 5A9, Canada
| | - Petros Papagerakis
- Laboratory of Precision Oral Health and Chronobiology, College of Dentistry, University of Saskatchewan, Saskatoon, SK S7N 5E4, Canada
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7K 5A9, Canada
| | - Silvana Papagerakis
- Laboratory of Oral, Head and Neck Cancer - Personalized Diagnostics and Therapeutics, Department of Surgery - Division of Head and Neck Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7K 5A9, Canada.
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
24
|
Development of Antibody-Modified Nanobubbles Using Fc-Region-Binding Polypeptides for Ultrasound Imaging. Pharmaceutics 2019; 11:pharmaceutics11060283. [PMID: 31208098 PMCID: PMC6631014 DOI: 10.3390/pharmaceutics11060283] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 02/01/2023] Open
Abstract
Ultrasound (US) imaging is a widely used imaging technique. The use of US contrast agents such as microbubbles, which consist of phospholipids and are filled with perfluorocarbon gases, has become an indispensable component of clinical US imaging, while molecular US imaging has recently attracted significant attention in combination with efficient diagnostics. The avidin–biotin interaction method is frequently used to tether antibodies to microbubbles, leading to the development of a molecular targeting US imaging agent. However, avidin still has limitations such as immunogenicity. We previously reported that lipid-based nanobubbles (NBs) containing perfluorocarbon gas are suitable for US imaging and gene delivery. In this paper, we report on the development of a novel antibody modification method for NBs using Fc-region-binding polypeptides derived from protein A/G. First, we prepared anti-CD146 antibody-modified NBs using this polypeptide, resulting in high levels of attachment to human umbilical vein endothelial cells expressing CD146. To examine their targeting ability and US imaging capability, the NBs were administered to tumor-bearing mice. The contrast imaging of antibody-modified NBs was shown to be prolonged compared with that of non-labeled NBs. Thus, this antibody modification method using an Fc-binding polypeptide may be a feasible tool for developing a next-generation antibody-modified US imaging agent.
Collapse
|
25
|
Tanaka M, Okinaga T, Iwanaga K, Matsuo K, Toyono T, Sasaguri M, Ariyoshi W, Tominaga K, Enomoto Y, Matsumura Y, Nishihara T. Anticancer effect of novel platinum nanocomposite beads on oral squamous cell carcinoma cells. J Biomed Mater Res B Appl Biomater 2019; 107:2281-2287. [PMID: 30689290 DOI: 10.1002/jbm.b.34320] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 12/17/2018] [Accepted: 12/27/2018] [Indexed: 01/19/2023]
Abstract
Nanoparticles are used in industry and medicine, because of their physiochemical properties, such as size, charge, large surface area and surface reactivity. Recently, metal nanoparticles were reported to show cell toxicity on cancer cells. In this study, we focused novel platinum nanoparticles-conjugated latex beads (P2VPs), platinum nanocomposite (PtNCP) beads, and investigated the possibility to incorporate novel anti-cancer effect of these combined nanoparticles. Oral squamous cell carcinoma cell lines, HSC-3-M3 cells were injected subcutaneously into the back of nude mice to produce a xenograft model. PtNCP beads were injected locally and examined by measuring tumor volume and comparing pathological histology. PtNCP beads treatment suppressed tumor growth and identified increasing pathological necrotic areas, in vivo. PtNCP beads inhibited the cell viability of HSC-3-M3 cells in dose-dependent manner and induced the cytotoxicity with extracellular LDH value, in vitro. Furthermore, SEM images were morphologically observed in PtNCP beads-treated HSC-3-M3 cells. The aggregation of the PtNCP beads on the cell membrane, the destructions of the cell membrane and globular structures were observed in the SEM image. Our results indicated that a potential anti-cancer effect of the PtNCP beads, suggesting the possibility as a therapeutic tool for cancer cell-targeted therapy. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 2281-2287, 2019.
Collapse
Affiliation(s)
- Mai Tanaka
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan.,Division of Oral and Maxillofacial Surgery, Department of Science of physical Functions, Kyushu Dental University, Kitakyushu, Japan
| | | | - Kenjiro Iwanaga
- Division of Preventive Dentistry, Department of Oral Health and Development Sciences, Tohoku University, Sendai, Japan
| | - Kou Matsuo
- Division of Oral Pathology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan
| | - Takashi Toyono
- Division of Oral Histology and Neurobiology, Kyushu Dental College, Kitakyushu, Japan
| | - Masaaki Sasaguri
- Division of Oral and Maxillofacial Surgery, Department of Science of physical Functions, Kyushu Dental University, Kitakyushu, Japan
| | - Wataru Ariyoshi
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan
| | - Kazuhiro Tominaga
- Division of Oral and Maxillofacial Surgery, Department of Science of physical Functions, Kyushu Dental University, Kitakyushu, Japan
| | - Yasushi Enomoto
- New Materials Development Center, Nippon Steel & Sumikin Chemical Co., Ltd, Chiba, Japan
| | - Yasufumi Matsumura
- New Materials Development Center, Nippon Steel & Sumikin Chemical Co., Ltd, Chiba, Japan
| | - Tatsuji Nishihara
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan
| |
Collapse
|
26
|
Snipstad S, Sulheim E, de Lange Davies C, Moonen C, Storm G, Kiessling F, Schmid R, Lammers T. Sonopermeation to improve drug delivery to tumors: from fundamental understanding to clinical translation. Expert Opin Drug Deliv 2018; 15:1249-1261. [PMID: 30415585 DOI: 10.1080/17425247.2018.1547279] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Ultrasound in combination with microbubbles can make cells and tissues more accessible for drugs, thereby achieving improved therapeutic outcomes. In this review, we introduce the term 'sonopermeation', covering mechanisms such as pore formation (traditional sonoporation), as well as the opening of intercellular junctions, stimulated endocytosis/transcytosis, improved blood vessel perfusion and changes in the (tumor) microenvironment. Sonopermeation has gained a lot of interest in recent years, especially for delivering drugs through the otherwise impermeable blood-brain barrier, but also to tumors. AREAS COVERED In this review, we summarize various in vitro assays and in vivo setups that have been employed to unravel the fundamental mechanisms involved in ultrasound-enhanced drug delivery, as well as clinical trials that are ongoing in patients with brain, pancreatic, liver and breast cancer. We summarize the basic principles of sonopermeation, describe recent findings obtained in (pre-) clinical trials, and discuss future directions. EXPERT OPINION We suggest that an improved mechanistic understanding, and microbubbles and ultrasound equipment specialized for drug delivery (and not for imaging) are key aspects to create more effective treatment regimens by sonopermeation. Real-time feedback and tools to predict therapeutic outcome and which tumors/patients will benefit from sonopermeation-based interventions will be important to promote clinical translation.
Collapse
Affiliation(s)
- Sofie Snipstad
- a Department of Physics , Norwegian University of Science and Technology (NTNU) , Trondheim , Norway.,b Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway.,c Cancer Clinic , St. Olavs Hospital , Trondheim , Norway
| | - Einar Sulheim
- a Department of Physics , Norwegian University of Science and Technology (NTNU) , Trondheim , Norway.,b Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway.,c Cancer Clinic , St. Olavs Hospital , Trondheim , Norway
| | - Catharina de Lange Davies
- a Department of Physics , Norwegian University of Science and Technology (NTNU) , Trondheim , Norway
| | - Chrit Moonen
- d Imaging Division , University Medical Center , Utrecht , The Netherlands
| | - Gert Storm
- e Department of Pharmaceutics , Utrecht University , Utrecht , The Netherlands.,f Department of Targeted Therapeutics , University of Twente , Enschede , The Netherlands
| | - Fabian Kiessling
- g Institute for Experimental Molecular Imaging , RWTH Aachen University , Aachen , Germany
| | - Ruth Schmid
- b Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway
| | - Twan Lammers
- e Department of Pharmaceutics , Utrecht University , Utrecht , The Netherlands.,f Department of Targeted Therapeutics , University of Twente , Enschede , The Netherlands.,g Institute for Experimental Molecular Imaging , RWTH Aachen University , Aachen , Germany
| |
Collapse
|