1
|
Zhou L, Zhang TJ, Zhang L, Deng QY, Xia ZY, Chen SL, Cheng DB, Qiao ZY, Wang H. Stimuli-responsive peptide-based nanodrug delivery systems for tumor therapy. Chem Commun (Camb) 2025; 61:7384-7407. [PMID: 40293360 DOI: 10.1039/d5cc00950b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Compared to free chemotherapeutic drugs, nano-sized drug delivery systems exhibit enhanced therapeutic effects and reduced in vivo toxicity. Peptide-based drug delivery systems have garnered significant attention due to the advantageous properties of peptides, including their excellent biocompatibility, diverse side-chain functionalities, and ability to form stable secondary structures. Incorporating stimuli-responsive amino acid residues or specific responsive moieties within their side chains endows these peptide-based drug delivery systems with unique stimuli-responsive characteristics. In this review, we summarize recent advancements and mechanisms in peptide-based nanodrug delivery systems that are capable of responding to one or multiple stimuli as well as conclude with a concise overview of the challenges that lie ahead in this field.
Collapse
Affiliation(s)
- Lei Zhou
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, Wuhan 430070, China.
- CAS Center for Excellence in Nanoscience Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Ting-Jie Zhang
- CAS Center for Excellence in Nanoscience Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Lu Zhang
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, Wuhan 430070, China.
| | - Qiu-Ying Deng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, Wuhan 430070, China.
| | - Zhi-Yu Xia
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, Wuhan 430070, China.
| | - Si-Lin Chen
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, Wuhan 430070, China.
- CAS Center for Excellence in Nanoscience Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, Wuhan 430070, China.
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang 441000, Hubei, China
| | - Zeng-Ying Qiao
- CAS Center for Excellence in Nanoscience Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Hao Wang
- CAS Center for Excellence in Nanoscience Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| |
Collapse
|
2
|
Wang Y, Huang R, Feng S, Mo R. Advances in nanocarriers for targeted drug delivery and controlled drug release. Chin J Nat Med 2025; 23:513-528. [PMID: 40383609 DOI: 10.1016/s1875-5364(25)60861-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/13/2024] [Accepted: 12/24/2024] [Indexed: 05/20/2025]
Abstract
Nanocarrier-based drug delivery systems (nDDSs) present significant opportunities for improving disease treatment, offering advantages in drug encapsulation, solubilization, stability enhancement, and optimized pharmacokinetics and biodistribution. nDDSs, comprising lipid, polymeric, protein, and inorganic nanovehicles, can be guided by or respond to biological cues for precise disease treatment and management. Equipping nanocarriers with tissue/cell-targeted ligands enables effective navigation in complex environments, while functionalization with stimuli-responsive moieties facilitates site-specific controlled release. These strategies enhance drug delivery efficiency, augment therapeutic efficacy, and reduce side effects. This article reviews recent strategies and ongoing advancements in nDDSs for targeted drug delivery and controlled release, examining lesion-targeted nanomedicines through surface modification with small molecules, peptides, antibodies, carbohydrates, or cell membranes, and controlled-release nanocarriers responding to endogenous signals such as pH, redox conditions, enzymes, or external triggers like light, temperature, and magnetism. The article also discusses perspectives on future developments.
Collapse
Affiliation(s)
- Yuqian Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Renqi Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Shufan Feng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Ran Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
3
|
Tayyab M, Ahmed N, Al Nasir MH, Ihsan A, Rehman AU, Munawar KS, Garcia DJ, Iglesias de la Cruz MDC, Rehman M. Magnetic hyperthermia-triggered multi-functional thermo-responsive lipid nanoparticles for enhanced paclitaxel release and cytotoxicity. NANOSCALE ADVANCES 2025:d5na00072f. [PMID: 40444139 PMCID: PMC12117578 DOI: 10.1039/d5na00072f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/21/2025] [Indexed: 06/02/2025]
Abstract
The inadequate safety and efficacy of chemotherapy have led cancer medicine to focus on localizing drug delivery to the target. Thermoresponsive nanocarriers (liposomes and polymeric networks) exploit local hyperthermia to trigger targeted payload release; however, their low stability and unpredictable fate in vivo have led to failure in clinical studies. To overcome these challenges, we reported first-of-its-kind thermoresponsive lipid nanoparticles (TLNs) that undergo solid-liquid phase transition under hyperthermia to release the payload. This study enabled TLNs with on-demand drug delivery functionality to breast cancer cells by incorporating magnetically activated iron oxide nanoparticles (γ-Fe2O3) into a lauric and oleic acid-based phase-changing lipid-matrix to synthesize paclitaxel (PTX)- and γ-Fe2O3-loaded TLNs (P-γ-TLN). Critical independent variables were selected and then optimized using a central composite design to obtain the optimized formulation, P-γ-TLN 12, with a size of ∼183 nm, polydispersity of 0.50, zeta potential of -22 mV, and encapsulation efficiencies of 85% for PTX and 60.49% for γ-Fe2O3. Thermoresponsive delivery was confirmed, with TLNs remaining relatively stable at 37 °C for 72 h, releasing only 34.26% of the drug, whereas exposure to 45 °C resulted in more than a two-fold increase, releasing 79.35% over the same period. Under an external alternating magnetic field, γ-Fe2O3 generated hyperthermia and induced a phase transition in P-γ-TLN, leading to abrupt drug release. Both γ-Fe2O3 and TLNs exhibited high biocompatibility, but TLN encapsulation significantly improved uptake in MCF-7 breast cancer cells. Under AMF, P-γ-TLN showed enhanced PTX release, resulting in more potent cytotoxicity against MCF-7 cells. The combination of high payload capacity, stimuli-responsive release, thermotherapy, and enhanced chemotherapeutic response highlights the substantial potential for TLNs in cancer therapy.
Collapse
Affiliation(s)
- Muhammad Tayyab
- Department of Pharmacy, Quaid I Azam University Islamabad 45320 Pakistan
| | - Naveed Ahmed
- Department of Pharmacy, Quaid I Azam University Islamabad 45320 Pakistan
| | | | - Ayesha Ihsan
- National Institute of Biotechnology and Genetic Engineering (NIBGE-C), Pakistan Institute of Engineering and Applied Sciences (PIEAS) Faisalabad Pakistan
| | - Asim Ur Rehman
- Department of Pharmacy, Quaid I Azam University Islamabad 45320 Pakistan
| | | | - Daniel Jaque Garcia
- Nanomaterials for Bioimaging Group (nanoBIG), Facultad de Ciencias, Departamento de Física de Materiales, Universidad Autónoma de Madrid 28049 Spain
| | - Maria Del Carmen Iglesias de la Cruz
- Nanomaterials for Bioimaging Group (nanoBIG), Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid Avda, Arzobispo Morcillo 2 Madrid 28029 Spain
| | - Mubashar Rehman
- Department of Pharmacy, Quaid I Azam University Islamabad 45320 Pakistan
| |
Collapse
|
4
|
Belyaev IB, Griaznova OY, Yaremenko AV, Deyev SM, Zelepukin IV. Beyond the EPR effect: Intravital microscopy analysis of nanoparticle drug delivery to tumors. Adv Drug Deliv Rev 2025; 219:115550. [PMID: 40021012 DOI: 10.1016/j.addr.2025.115550] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
Delivery of nanoparticles (NPs) to solid tumors has long relied on enhanced permeability and retention (EPR) effect, involving permeation of NPs through a leaky vasculature with prolonged retention by reduced lymphatic drainage in tumor. Recent research studies and clinical data challenge EPR concept, revealing alternative pathways and approaches of NP delivery. The area was significantly impacted by the implementation of intravital optical microscopy, unraveling delivery mechanisms at cellular level in vivo. This review presents analysis of the reasons for EPR heterogeneity in tumors and describes non-EPR based concepts for drug delivery, which can supplement the current paradigm. One of the approaches is targeting tumor endothelium by NPs with subsequent intravascular drug release and gradient-driven drug transport to tumor interstitium. Others exploit various immune cells for tumor infiltration and breaking endothelial barriers. Finally, we discuss the involvement of active transcytosis through endothelial cells in NP delivery. This review aims to inspire further understanding of the process of NP extravasation in tumors and provide insights for developing next-generation nanomedicines with improved delivery.
Collapse
Affiliation(s)
- Iaroslav B Belyaev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands
| | - Olga Yu Griaznova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | | | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Ivan V Zelepukin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75123, Sweden.
| |
Collapse
|
5
|
Einolghasi FB, Zeinali B, Vafai K, Mojra A. Development of a prediction model for hyperthermia-enhanced drug delivery using thermosensitive nanoparticles. J Therm Biol 2025; 129:104124. [PMID: 40300402 DOI: 10.1016/j.jtherbio.2025.104124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 05/01/2025]
Abstract
Therapies targeting solid tumors are often hindered by drug resistance mechanisms, which presenting significant challenges. This study investigates the synergistic effect of hyperthermia and chemotherapy by utilizing thermosensitive nanoparticles (NPs) to enhance drug uptake by cancer cells. The thermoresponsive nature of NPs enables precise drug release under hyperthermic conditions, making them ideal for localized treatment. To achieve this, we developed a multi-component numerical model of focused ultrasound-enhanced doxorubicin delivery using tumor-targeting nanoparticles. The model incorporates critical factors such as blood coagulation and non-Fourier heat transfer, and it was validated against experimental results, thereby enhancing the reliability and accuracy of our findings. Ultimately, a prediction model was developed to estimate heat-driven irreversible cell damage in relation to heating power and NP size. The results demonstrated a remarkable 13-fold increase in drug penetration and a 4.6-fold enhancement in the fraction of killed cells with hyperthermia. These findings underscore the importance of accurately controlling the temperature field and heating power for optimal hyperthermia-assisted chemotherapy outcomes.
Collapse
Affiliation(s)
| | - Behnam Zeinali
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Kambiz Vafai
- Department of Mechanical Engineering, University of California, Riverside, CA, 92521, USA.
| | - Afsaneh Mojra
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
| |
Collapse
|
6
|
Wang X, Allen C. Synergistic effects of thermosensitive liposomal doxorubicin, mild hyperthermia, and radiotherapy in breast cancer management: an orthotopic mouse model study. Drug Deliv Transl Res 2025; 15:1011-1022. [PMID: 38977541 DOI: 10.1007/s13346-024-01654-2] [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: 06/13/2024] [Indexed: 07/10/2024]
Abstract
Liposome formulations of the cancer drug doxorubicin have been developed to address the severe side effects that result from administration of this drug in a conventional formulation. Among them, thermosensitive liposomal doxorubicin presents enhanced tumor targeting and efficient drug release when combined with mild hyperthermia localized to the tumor site. Exploiting the radiosensitizing benefits of localized thermal therapy, the integration of radiation therapy with the thermally activated liposomal system is posited to amplify the anti-tumor efficacy. This study explored a synergistic therapeutic strategy that combines thermosensitive liposomal doxorubicin, mild hyperthermia, and radiotherapy, using an orthotopic murine model of breast cancer. The protocol of sequential multi-modal treatment, incorporating low-dose chemotherapy and radiotherapy, substantially postponed the progression of primary tumor growth in comparison to the application of monotherapy at elevated dosages. Improvements in unheated distant lesions were also observed. Furthermore, the toxicity associated with the combination treatment was comparable to that of either thermosensitive liposome treatment or radiation alone at low doses. These outcomes underscore the potential of multi-modal therapeutic strategies to refine treatment efficacy while concurrently diminishing adverse effects in the management of breast cancer, providing valuable insight for the future refinement of thermosensitive liposomal doxorubicin applications.
Collapse
Affiliation(s)
- Xuehan Wang
- 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.
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
7
|
Guo Z, Xiao Y, Wu W, Zhe M, Yu P, Shakya S, Li Z, Xing F. Metal-organic framework-based smart stimuli-responsive drug delivery systems for cancer therapy: advances, challenges, and future perspectives. J Nanobiotechnology 2025; 23:157. [PMID: 40022098 PMCID: PMC11871784 DOI: 10.1186/s12951-025-03252-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
Cancer treatment is currently one of the most critical healthcare issues globally. A well-designed drug delivery system can precisely target tumor tissues, improve efficacy, and reduce damage to normal tissues. Stimuli-responsive drug delivery systems (SRDDSs) have shown promising application prospects. Intelligent nano drug delivery systems responsive to endogenous stimuli such as weak acidity, complex redox characteristics, hypoxia, active energy metabolism, as well as exogenous stimuli like high temperature, light, pressure, and magnetic fields are increasingly being applied in chemotherapy, radiotherapy, photothermal therapy, photodynamic therapy, and various other anticancer approaches. Metal-organic frameworks (MOFs) have become promising candidate materials for constructing SRDDSs due to their large surface area, tunable porosity and structure, ease of synthesis and modification, and good biocompatibility. This paper reviews the application of MOF-based SRDDSs in various modes of cancer therapy. It summarizes the key aspects, including the classification, synthesis, modifications, drug loading modes, stimuli-responsive mechanisms, and their roles in different cancer treatment modalities. Furthermore, we address the current challenges and summarize the potential applications of artificial intelligence in MOF synthesis. Finally, we propose strategies to enhance the efficacy and safety of MOF-based SRDDSs, ultimately aiming at facilitating their clinical translation.
Collapse
Affiliation(s)
- Ziliang Guo
- Division of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuzhen Xiao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Beijing, 100005, China
| | - Wenting Wu
- Department of Pediatric Surgery, Division of Orthopedic Surgery, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Man Zhe
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peiyun Yu
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
| | - Sujan Shakya
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhihui Li
- Division of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Fei Xing
- Department of Pediatric Surgery, Division of Orthopedic Surgery, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
8
|
You S, Wu S, Yang S, Zhao Z, Chen W, Chen X, Wang H, Xia Q, Xiong J, Zhou H, Mo X. Pulsed Ultrasound-Mediated Drug Delivery Enhancement Through Human Sclera. Transl Vis Sci Technol 2025; 14:7. [PMID: 39786737 PMCID: PMC11734544 DOI: 10.1167/tvst.14.1.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 11/21/2024] [Indexed: 01/12/2025] Open
Abstract
Purpose The purpose of this study was to characterize whether pulsed ultrasound (PUS) affects transscleral drug delivery. Methods Fluorescein sodium (NaF, 376 Da) and fluorescein isothiocyanate-conjugated dextran 40 (FD-40, 40 kDa) were used as model drugs. Human sclera grafts were placed in modified Franz diffusion cells and were treated by PUS (1 megahertz [MHz], 0.71 W/cm2, duty cycle 30%, application time 5 minutes) once or repeatedly under various conditions to assess permeation enhancement and reservoir effect. The safety of PUS application was assessed on human sclera grafts ex vivo and rabbit eyes in vivo by histology and temperature measurements. Results Single PUS application yielded a significant increase in FD-40 permeation (P < 0.05). Repeated PUS applications led to a further enhancement in FD-40 permeation and also significantly promoted NaF permeation (more than 8.51-fold, P < 0.05). The human scleral permeability was temporarily modified by PUS, as evidenced by the increased scleral permeability during PUS application and the unchanged permeability coefficients at steady state. The reservoir effect of human sclera was also enhanced by PUS application. Cavitation was detected under PUS. A minor increase in graft temperature rise (<1°C) and no ocular damage was caused by PUS. Conclusions PUS is an efficient and safe method to enhance model drugs to transport across human sclera by increasing the scleral permeability transiently and improving the reservoir effect. The enhancement was correlated with the molecule size and further promoted by the repeated PUS application. Translational Relevance Our study provides proof of concept for using PUS to enhance drug delivery to the posterior eye segment.
Collapse
Affiliation(s)
- Shuqi You
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Department of Ophthalmology, Children's Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Suqian Wu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Shicheng Yang
- Shanghai Acoustics Laboratory, Chinese Academy of Science, Shanghai, China
| | - Zhenyang Zhao
- Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, USA
- Emory Eye Center, Emory University, Atlanta, GA, USA
| | - Wei Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Xiangwu Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Huan Wang
- Shanghai Acoustics Laboratory, Chinese Academy of Science, Shanghai, China
| | - Qing Xia
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jiawei Xiong
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Hongsheng Zhou
- Institute of Advanced Ultrasonic Technology of National Innovation Center Par Excellence, Shanghai, China
| | - Xiaofen Mo
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| |
Collapse
|
9
|
Li T, Zhang L, Qu X, Lei B. Advanced Thermoactive Nanomaterials for Thermomedical Tissue Regeneration: Opportunities and Challenges. SMALL METHODS 2025; 9:e2400510. [PMID: 39588862 DOI: 10.1002/smtd.202400510] [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: 06/04/2024] [Revised: 11/06/2024] [Indexed: 11/27/2024]
Abstract
Nanomaterials usually possess remarkable properties, including excellent biocompatibility, unique physical and chemical characteristics, and bionic attributes, which make them highly promising for applications in tissue regeneration. Thermal therapy has emerged as a versatile approach for wound healing, nerve repair, bone regeneration, tumor therapy, and antibacterial tissue regeneration. By combining nanomaterials with thermal therapy, multifunctional nanomaterials with thermogenic effects and tissue regeneration capabilities can be engineered to achieve enhanced therapeutic outcomes. This study provides a comprehensive review of the effects of thermal stimulation on cellular and tissue regeneration. Furthermore, it highlights the applications of photothermal, magnetothermal, and electrothermal nanomaterials, and thermally responsive drug delivery systems in tissue engineering. In Addition, the bioactivities and biocompatibilities of several representative thermal nanomaterials are discussed. Finally, the challenges facing thermal nanomaterials are outlined, and future prospects in the field are presented with the aim of offering new opportunities and avenues for the utilization of thermal nanomaterials in tissue regeneration.
Collapse
Affiliation(s)
- Ting Li
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Long Zhang
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Xiaoyan Qu
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Bo Lei
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710054, China
| |
Collapse
|
10
|
Li X, Lai Y, Wan G, Zou J, He W, Yang P. Approved natural products-derived nanomedicines for disease treatment. Chin J Nat Med 2024; 22:1100-1116. [PMID: 39725511 DOI: 10.1016/s1875-5364(24)60726-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Indexed: 12/28/2024]
Abstract
In recent years, there has been an increasing emphasis on exploring innovative drug delivery approaches due to the limitations of conventional therapeutic strategies, such as inadequate drug targeting, insufficient therapeutic efficacy, and significant adverse effects. Nanomedicines have emerged as a promising solution with notable advantages, including extended drug circulation, targeted delivery, and improved bioavailability, potentially enhancing the clinical treatment of various diseases. Natural products/materials-derived nanomedicines, characterized by their natural therapeutic efficacy, superior biocompatibility, and safety profile, play a crucial role in nanomedicine-based treatments. This review provides a comprehensive overview of currently approved natural products-derived nanomedicines, emphasizing the essential properties of natural products-derived drug carriers, their applications in clinical diagnosis and treatment, and the current therapeutic potential and challenges. The aim is to offer guidance for the application and further development of these innovative therapeutic approaches.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Yaoyao Lai
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Guanghan Wan
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China.
| | - Pei Yang
- School of Science, China Pharmaceutical University, Nanjing 2111198, China.
| |
Collapse
|
11
|
Hefnawy A, Abdelhamid AS, Abdelaziz MM, Elzoghby AO, Khalil IA. Recent advances in nano-based drug delivery systems for treatment of liver cancer. J Pharm Sci 2024; 113:3145-3172. [PMID: 39151795 DOI: 10.1016/j.xphs.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Liver cancer is one of the aggressive primary tumors as evident by high rate of incidence and mortality. Conventional treatments (e.g. chemotherapy) suffer from various drawbacks including wide drug distribution, low localized drug concentration, and severe off-site toxicity. Therefore, they cannot satisfy the mounting need for safe and efficient cancer therapeutics, and alternative novel strategies are needed. Nano-based drug delivery systems (NDDSs) are among these novel approaches that can improve the overall therapeutic outcomes. NDDSs are designed to encapsulate drug molecules and target them specifically to liver cancer. Thus, NDDSs can selectively deliver therapeutic agents to the tumor cells and avoid distribution to off-target sites which should improve the safety profile of the active agents. Nonetheless, NDDSs should be well designed, in terms of the preparing materials, nanocarriers structure, and the targeting strategy, in order to accomplish these objectives. This review discusses the latest advances of NDDSs for cancer therapy with emphasis on the aforementioned essential design components. The review also entails the challenges associated with the clinical translation of NDDSs, and the future perspectives towards next-generation NDDSs.
Collapse
Affiliation(s)
- Amr Hefnawy
- Smyth Lab, College of Pharmacy, University of Texas at Austin, TX 78712, USA.
| | - Ahmed S Abdelhamid
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| | - Moustafa M Abdelaziz
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66047, USA.
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt; Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt; Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Islam A Khalil
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City 12582, Giza, Egypt.
| |
Collapse
|
12
|
Abtahi MS, Fotouhi A, Rezaei N, Akalin H, Ozkul Y, Hossein-Khannazer N, Vosough M. Nano-based drug delivery systems in hepatocellular carcinoma. J Drug Target 2024; 32:977-995. [PMID: 38847573 DOI: 10.1080/1061186x.2024.2365937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/22/2024] [Accepted: 06/02/2024] [Indexed: 06/19/2024]
Abstract
The high recurrence rate of hepatocellular carcinoma (HCC) and poor prognosis after medical treatment reflects the necessity to improve the current chemotherapy protocols, particularly drug delivery methods. Development of targeted and efficient drug delivery systems (DDSs), in all active, passive and stimuli-responsive forms for selective delivery of therapeutic drugs to the tumour site has been extended to improve efficacy and reduce the severe side effects. Recent advances in nanotechnology offer promising breakthroughs in the diagnosis, treatment and monitoring of cancer cells. In this review, the specific design of DDSs based on the different nano-particles and their surface engineering is discussed. In addition, the innovative clinical studies in which nano-based DDS was used in the treatment of HCC were highlighted.
Collapse
Affiliation(s)
- Maryam Sadat Abtahi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Alireza Fotouhi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Niloufar Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hilal Akalin
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Yusuf Ozkul
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Nikoo Hossein-Khannazer
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
13
|
McCorkell G, Piva T, Highgate D, Nakayama M, Geso M. Ultrasound-stimulated microbubbles to enhance radiotherapy: A scoping review. J Med Imaging Radiat Oncol 2024; 68:740-769. [PMID: 39250692 DOI: 10.1111/1754-9485.13740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 07/22/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Primarily used as ultrasound contrast agents, microbubbles have recently emerged as a versatile therapeutic vector that can be 'burst' to deliver payloads in the presence of suitably optimised ultrasound fields. Ultrasound-stimulated microbubbles (USMB) have recently demonstrated improvements in treatment outcomes across a variety of clinical applications. This scoping review investigates whether this potential translates into the context of radiation therapy by evaluating the application of this technology across all three phases of radiation action. METHODS Primary research articles, excluding poster presentations and conference proceedings, were identified through systematic searches of the PubMed NCBI/Medline, Embase/OVID, Web of Science and CINAHL/EBSCOhost databases, with additional articles identified via manual Google Scholar searching. Articles were dual screened for inclusion using the Covidence systematic review platform and classified against all three phases of radiation action. RESULTS Overall, 57 eligible publications from a total of 1389 identified articles were included in the review, with studies dating back to 2012. Study heterogeneity prevented formal statistical analysis; however, most articles reported improved outcomes using USMB in the presence of radiation compared to that of radiation alone. These improvements appear to result from the use of USMB as either a biovascular disruptor causing tumour cell damage via indirect mechanisms, or as a localised treatment vector that directly increases tumour cell uptake of other therapeutic and physical agents designed to enhance radiation action. CONCLUSIONS USMB demonstrate exciting potential to enhance the effects of radiation treatments due to their versatility and capacity to target all three phases of radiation action.
Collapse
Affiliation(s)
- Giulia McCorkell
- RMIT University, Melbourne, Victoria, Australia
- The University of Melbourne, Melbourne, Victoria, Australia
| | | | | | - Masao Nakayama
- RMIT University, Melbourne, Victoria, Australia
- Kobe University, Kobe, Hyogo, Japan
- Kita-Harima Medical Center, Ono, Hyogo, Japan
| | - Moshi Geso
- RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Campea MA, Macdonald C, Hoare T. Supramolecularly-Cross-Linked Nanogel Assemblies for On-Demand, Ultrasound-Triggered Chemotherapy. Biomacromolecules 2024; 25:4697-4714. [PMID: 38995854 DOI: 10.1021/acs.biomac.3c01432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Stimulating the release of small nanoparticles (NPs) from a larger NP via the application of an exogenous stimulus offers the potential to address the different size requirements for circulation versus penetration that hinder chemotherapeutic drug delivery. Herein, we report a size-switching nanoassembly-based drug delivery system comprised of ultrasmall starch nanoparticles (SNPs, ∼20-50 nm major size fraction) encapsulated in a poly(oligo(ethylene glycol) methyl ether methacrylate) nanogel (POEGMA, ∼150 nm major size fraction) cross-linked via supramolecular PEG/α-cyclodextrin (α-CD) interactions. Upon heating the nanogel using a non-invasive, high-intensity focused ultrasound (HIFU) trigger, the thermoresponsive POEGMA-CD nanoassemblies are locally de-cross-linked, inducing in situ release of the highly penetrative drug-loaded SNPs. HIFU triggering increased the release of nanoassembly-loaded DOX from 17 to 37% after 3 h, a result correlated with significantly more effective tumor killing relative to nanoassemblies in the absence of HIFU or drug alone. Furthermore, 1.5× more total fluorescence was observed inside a tumor spheroid when nanoassemblies prepared with fluorophore-labeled SNPs were triggered with HIFU relative to the absence of HIFU. We anticipate this strategy holds promise for delivering tunable doses of chemotherapeutic drugs both at and within a tumor site using a non-invasive triggering approach.
Collapse
Affiliation(s)
- Matthew A Campea
- Department of Chemical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Cameron Macdonald
- Department of Chemical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
15
|
Aloss K, Hamar P. Augmentation of the EPR effect by mild hyperthermia to improve nanoparticle delivery to the tumor. Biochim Biophys Acta Rev Cancer 2024; 1879:189109. [PMID: 38750699 DOI: 10.1016/j.bbcan.2024.189109] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
The clinical translation of the nanoparticle (NP)-based anticancer therapies is still unsatisfactory due to the heterogeneity of the enhanced permeability and retention (EPR) effect. Despite the promising preclinical outcome of the pharmacological EPR enhancers, their systemic toxicity can limit their clinical application. Hyperthermia (HT) presents an efficient tool to augment the EPR by improving tumor blood flow (TBF) and vascular permeability, lowering interstitial fluid pressure (IFP), and disrupting the structure of the extracellular matrix (ECM). Furthermore, the HT-triggered intravascular release approach can overcome the EPR effect. In contrast to pharmacological approaches, HT is safe and can be focused to cancer tissues. Moreover, HT conveys direct anti-cancer effects, which improve the efficacy of the anti-cancer agents encapsulated in NPs. However, the clinical application of HT is challenging due to the heterogeneous distribution of temperature within the tumor, the length of the treatment and the complexity of monitoring.
Collapse
Affiliation(s)
- Kenan Aloss
- Institute of Translational Medicine - Semmelweis University - 1094, Tűzoltó utca, 37-49, Budapest, Hungary
| | - Péter Hamar
- Institute of Translational Medicine - Semmelweis University - 1094, Tűzoltó utca, 37-49, Budapest, Hungary.
| |
Collapse
|
16
|
Xu Z, Piao X, Wang M, Pichardo S, Cheng B. Microbubble-enhanced transcranial MR-guided focused ultrasound brain hyperthermia: heating mechanism investigation using finite element method. ULTRASONICS SONOCHEMISTRY 2024; 107:106889. [PMID: 38702233 PMCID: PMC11214346 DOI: 10.1016/j.ultsonch.2024.106889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
Recently, our group developed a synergistic brain drug delivery method to achieve simultaneous transcranial hyperthermia and localized blood-brain barrier opening via MR-guided focused ultrasound (MRgFUS). In a rodent model, we demonstrated that the ultrasound power required for transcranial MRgFUS hyperthermia was significantly reduced by injecting microbubbles (MBs). However, the specific mechanisms underlying the power reduction caused by MBs remain unclear. The present study aims to elucidate the mechanisms of MB-enhanced transcranial MRgFUS hyperthermia through numerical studies using the finite element method. The microbubble acoustic emission (MAE) and the viscous dissipation (VD) were hypothesized to be the specific mechanisms. Acoustic wave propagation was used to model the FUS propagation in the brain tissue, and a bubble dynamics equation for describing the dynamics of MBs with small shell thickness was used to model the MB oscillation under FUS exposures. A modified bioheat transfer equation was used to model the temperature in the rodent brain with different heat sources. A theoretical model was used to estimate the bubble shell's surface tension, elasticity, and viscosity losses. The simulation reveals that MAE and VD caused a 40.5% and 52.3% additional temperature rise, respectively. Compared with FUS only, MBs caused a 64.0% temperature increase, which is consistent with our previous animal experiments. Our investigation showed that MAE and VD are the main mechanisms of MB-enhanced transcranial MRgFUS hyperthermia.
Collapse
Affiliation(s)
- Zhouyang Xu
- Translational Research in Ultrasound Theranostics Laboratory, School of Biomedical Engineering, ShanghaiTech University, Shanghai, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China
| | - Xiangkun Piao
- Translational Research in Ultrasound Theranostics Laboratory, School of Biomedical Engineering, ShanghaiTech University, Shanghai, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China
| | - Mingyu Wang
- Translational Research in Ultrasound Theranostics Laboratory, School of Biomedical Engineering, ShanghaiTech University, Shanghai, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China
| | - Samuel Pichardo
- Department of Radiology, University of Calgary, Calgary, Canada; Department of Clinical Neurosciences, University of Calgary, Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Bingbing Cheng
- Translational Research in Ultrasound Theranostics Laboratory, School of Biomedical Engineering, ShanghaiTech University, Shanghai, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
17
|
Peng J, Partanen A, Pichardo S, Staruch R, Perry K, McGuffin M, Huang Y, Chan KK, Wong S, Czarnota G, Hynynen K, Chu W. Mild hyperthermia with magnetic resonance- guided high intensity focused ultrasound combined with salvage chemoradiation for recurrent rectal cancer. Int J Hyperthermia 2024; 41:2365385. [PMID: 38897584 DOI: 10.1080/02656736.2024.2365385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 04/29/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
INTRODUCTION Pelvic recurrences from rectal cancer present a challenging clinical scenario. Hyperthermia represents an innovative treatment option in combination with concurrent chemoradiation to enhance therapeutic effect. We provide the initial results of a prospective single center feasibility study (NCT02528175) for patients undergoing rectal cancer retreatment using concurrent chemoradiation and mild hyperthermia with MR-guided high intensity focused ultrasound (MR-HIFU). METHODS All patients were deemed ineligible for salvage surgery and were evaluated in a multidisciplinary fashion with a surgical oncologist, radiation oncologist and medical oncologist. Radiation was delivered to a dose of 30.6 Gy in 1.8 Gy per fraction with concurrent capecitabine. MR-HIFU was delivered on days 1, 8 and 15 of concurrent chemoradiation. Our primary objective was feasibility and toxicity. RESULTS Six patients (total 11 screened) were treated with concurrent chemoradiation and mild hyperthermia with MR-HIFU. Tumor size varied between 3.1-16.6 cm. Patients spent an average of 228 min in the MRI suite and sonication with the external transducer lasted an average of 35 min. There were no complications on the day of the MR-HIFU procedure and all acute toxicities (no grade >/=3 toxicities) resolved after completion of treatment. There were no late grade >/=3 toxicities. CONCLUSION Mild hyperthermia with MR-HIFU, in combination with concurrent chemoradiation for appropriately selected patients, is safe for localized pelvic recurrences from rectal cancer. The potential for MR-HIFU to be applied in the recurrent setting in rectal cancer treatment requires further technical development and prospective evaluation.
Collapse
Affiliation(s)
- Jonathan Peng
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | | | - Samuel Pichardo
- Cumming School of Medicine, University of Calgary, Calgary, Canada
| | | | - Kaitlyn Perry
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | - Merrylee McGuffin
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | - Yuexi Huang
- Department of Medical Biophysics, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Kelvin Kw Chan
- Department of Medical Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | - Shun Wong
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | - Greg Czarnota
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
- Department of Medical Biophysics, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Kullervo Hynynen
- Department of Medical Biophysics, Sunnybrook Research Institute, Toronto, ON, Canada
| | - William Chu
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| |
Collapse
|
18
|
Lammers T. Nanomedicine Tumor Targeting. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312169. [PMID: 38361435 DOI: 10.1002/adma.202312169] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/24/2024] [Indexed: 02/17/2024]
Abstract
Nanomedicines are extensively explored for cancer therapy. By delivering drug molecules more efficiently to pathological sites and by attenuating their accumulation in healthy organs and tissues, nanomedicine formulations aim to improve the balance between drug efficacy and toxicity. More than 20 cancer nanomedicines are approved for clinical use, and hundreds of formulations are in (pre)clinical development. Over the years, several key pitfalls have been identified as bottlenecks in nanomedicine tumor targeting and translation. These go beyond materials- and production-related issues, and particularly also encompass biological barriers and pathophysiological heterogeneity. In this manuscript, the author describes the most important principles, progress, and products in nanomedicine tumor targeting, delineates key current problems and challenges, and discusses the most promising future prospects to create clinical impact.
Collapse
Affiliation(s)
- Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Center for Biohyhrid Medical Systems, University Hospital RWTH Aachen, Forckenbeckstrasse 55, 52074, Aachen, Germany
| |
Collapse
|
19
|
Kashkooli FM, Jakhmola A, A Ferrier G, Sathiyamoorthy K, Tavakkoli J(J, C Kolios M. Development of an ultrasound-mediated nano-sized drug-delivery system for cancer treatment: from theory to experiment. Nanomedicine (Lond) 2024; 19:1167-1189. [PMID: 38722104 PMCID: PMC11418290 DOI: 10.2217/nnm-2023-0259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 03/06/2024] [Indexed: 09/21/2024] Open
Abstract
Aim: To establish a methodology for understanding how ultrasound (US) induces drug release from nano-sized drug-delivery systems (NSDDSs) and enhances drug penetration and uptake in tumors. This aims to advance cancer treatment strategies.Materials & methods: We developed a multi-physics mathematical model to elucidate and understand the intricate mechanisms governing drug release, transport and delivery. Unique in vitro models (monolayer, multilayer, spheroid) and a tailored US exposure setup were introduced to evaluate drug penetration and uptake.Results: The results highlight the potential advantages of US-mediated NSDDSs over conventional NSDDSs and chemotherapy, notably in enhancing drug release and inducing cell death.Conclusion: Our sophisticated numerical and experimental methods aid in determining and quantifying drug penetration and uptake into solid tumors.
Collapse
Affiliation(s)
| | - Anshuman Jakhmola
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
| | - Graham A Ferrier
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
| | | | - Jahangir (Jahan) Tavakkoli
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science & Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science & Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| |
Collapse
|
20
|
Zhu P, Simon I, Kokalari I, Kohane DS, Rwei AY. Miniaturized therapeutic systems for ultrasound-modulated drug delivery to the central and peripheral nervous system. Adv Drug Deliv Rev 2024; 208:115275. [PMID: 38442747 PMCID: PMC11031353 DOI: 10.1016/j.addr.2024.115275] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/19/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
Ultrasound is a promising technology to address challenges in drug delivery, including limited drug penetration across physiological barriers and ineffective targeting. Here we provide an overview of the significant advances made in recent years in overcoming technical and pharmacological barriers using ultrasound-assisted drug delivery to the central and peripheral nervous system. We commence by exploring the fundamental principles of ultrasound physics and its interaction with tissue. The mechanisms of ultrasonic-enhanced drug delivery are examined, as well as the relevant tissue barriers. We highlight drug transport through such tissue barriers utilizing insonation alone, in combination with ultrasound contrast agents (e.g., microbubbles), and through innovative particulate drug delivery systems. Furthermore, we review advances in systems and devices for providing therapeutic ultrasound, as their practicality and accessibility are crucial for clinical application.
Collapse
Affiliation(s)
- Pancheng Zhu
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands; State Key Laboratory of Mechanics and Control of Aerospace Structures, Nanjing University of Aeronautics & Astronautics, 210016, Nanjing, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Ignasi Simon
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands
| | - Ida Kokalari
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Alina Y Rwei
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands.
| |
Collapse
|
21
|
Paun RA, Jurchuk S, Tabrizian M. A landscape of recent advances in lipid nanoparticles and their translational potential for the treatment of solid tumors. Bioeng Transl Med 2024; 9:e10601. [PMID: 38435821 PMCID: PMC10905562 DOI: 10.1002/btm2.10601] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 03/05/2024] Open
Abstract
Lipid nanoparticles (LNPs) are biocompatible drug delivery systems that have found numerous applications in medicine. Their versatile nature enables the encapsulation and targeting of various types of medically relevant molecular cargo, including oligonucleotides, proteins, and small molecules for the treatment of diseases, such as cancer. Cancers that form solid tumors are particularly relevant for LNP-based therapeutics due to the enhanced permeation and retention effect that allows nanoparticles to accumulate within the tumor tissue. Additionally, LNPs can be formulated for both locoregional and systemic delivery depending on the tumor type and stage. To date, LNPs have been used extensively in the clinic to reduce systemic toxicity and improve outcomes in cancer patients by encapsulating chemotherapeutic drugs. Next-generation lipid nanoparticles are currently being developed to expand their use in gene therapy and immunotherapy, as well as to enable the co-encapsulation of multiple drugs in a single system. Other developments include the design of targeted LNPs to specific cells and tissues, and triggerable release systems to control cargo delivery at the tumor site. This review paper highlights recent developments in LNP drug delivery formulations and focuses on the treatment of solid tumors, while also discussing some of their current translational limitations and potential opportunities in the field.
Collapse
Affiliation(s)
- Radu A. Paun
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Sarah Jurchuk
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Faculty of Dentistry and Oral Health SciencesMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
22
|
Gray MD, Spiers L, Coussios CC. Sound speed and attenuation of human pancreas and pancreatic tumors and their influence on focused ultrasound thermal and mechanical therapies. Med Phys 2024; 51:809-825. [PMID: 37477551 DOI: 10.1002/mp.16622] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 06/06/2023] [Accepted: 06/20/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND There is increasing interest in using ultrasound for thermal ablation, histotripsy, and thermal or cavitational enhancement of drug delivery for the treatment of pancreatic cancer. Ultrasonic and thermal modelling conducted as part of the treatment planning process requires acoustic property values for all constituent tissues, but the literature contains no data for the human pancreas. PURPOSE This study presents the first acoustic property measurements of human pancreatic samples and provides examples of how these properties impact a broad range of ultrasound therapies. METHODS Data were collected on human pancreatic tissue samples at physiological temperature from 23 consented patients in cooperation with a hospital pathology laboratory. Propagation of ultrasound over the 2.1-4.5 MHz frequency range through samples of various thicknesses and pathologies was measured using a set of custom-built ultrasonic calipers, with the data processed to estimate sound speed and attenuation. The results were used in acoustic and thermal simulations to illustrate the impacts on extracorporeal ultrasound therapies for mild hyperthermia, thermal ablation, and histotripsy implemented with a CE-marked clinical system operating at 0.96 MHz. RESULTS The mean sound speed and attenuation coefficient values for human samples were well below the range of values in the literature for non-human pancreata, while the human attenuation power law exponents were substantially higher. The simulated impacts on ultrasound mediated therapies for the pancreas indicated that when using the human data instead of the literature average, there was a 30% reduction in median temperature elevation in the treatment volume for mild hyperthermia and 43% smaller volume within a 60°C contour for thermal ablation, all driven by attenuation. By comparison, impacts on boiling and intrinsic threshold histotripsy were minor, with peak pressures changing by less than 15% (positive) and 1% (negative) as a consequence of the counteracting effects of attenuation and sound speed. CONCLUSION This study provides the most complete set of speed of sound and attenuation data available for the human pancreas, and it reiterates the importance of acoustic material properties in the planning and conduct of ultrasound-mediated procedures, particularly thermal therapies.
Collapse
Affiliation(s)
- Michael D Gray
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Laura Spiers
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | | |
Collapse
|
23
|
Chen J, Hu S, Sun M, Shi J, Zhang H, Yu H, Yang Z. Recent advances and clinical translation of liposomal delivery systems in cancer therapy. Eur J Pharm Sci 2024; 193:106688. [PMID: 38171420 DOI: 10.1016/j.ejps.2023.106688] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/23/2023] [Accepted: 12/31/2023] [Indexed: 01/05/2024]
Abstract
The limitations of conventional cancer treatment are driving the emergence and development of nanomedicines. Research in liposomal nanomedicine for cancer therapy is rapidly increasing, opening up new horizons for cancer treatment. Liposomal nanomedicine, which focuses on targeted drug delivery to improve the therapeutic effect of cancer while reducing damage to normal tissues and cells, has great potential in the field of cancer therapy. This review aims to clarify the advantages of liposomal delivery systems in cancer therapy. We describe the recent understanding of spatiotemporal fate of liposomes in the organism after different routes of drug administration. Meanwhile, various types of liposome-based drug delivery systems that exert their respective advantages in cancer therapy while reducing side effects were discussed. Moreover, the combination of liposomal agents with other therapies (such as photodynamic therapy and photothermal therapy) has demonstrated enhanced tumor-targeting efficiency and therapeutic efficacy. Finally, the opportunities and challenges faced by the field of liposome nanoformulations for entering the clinical treatment of cancer are highlighted.
Collapse
Affiliation(s)
- Jiayi Chen
- School of Life Sciences, Jilin University, Changchun, China
| | - Siyuan Hu
- School of Life Sciences, Jilin University, Changchun, China
| | - Man Sun
- School of Life Sciences, Jilin University, Changchun, China
| | - Jianan Shi
- School of Life Sciences, Jilin University, Changchun, China
| | - Huan Zhang
- School of Life Sciences, Jilin University, Changchun, China
| | - Hongmei Yu
- China-Japan Union Hospital, Jilin University, Changchun, China.
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun, China.
| |
Collapse
|
24
|
Moradi Kashkooli F, Hornsby TK, Kolios MC, Tavakkoli JJ. Ultrasound-mediated nano-sized drug delivery systems for cancer treatment: Multi-scale and multi-physics computational modeling. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1913. [PMID: 37475577 DOI: 10.1002/wnan.1913] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 07/22/2023]
Abstract
Computational modeling enables researchers to study and understand various complex biological phenomena in anticancer drug delivery systems (DDSs), especially nano-sized DDSs (NSDDSs). The combination of NSDDSs and therapeutic ultrasound (TUS), that is, focused ultrasound and low-intensity pulsed ultrasound, has made significant progress in recent years, opening many opportunities for cancer treatment. Multiple parameters require tuning and optimization to develop effective DDSs, such as NSDDSs, in which mathematical modeling can prove advantageous. In silico computational modeling of ultrasound-responsive DDS typically involves a complex framework of acoustic interactions, heat transfer, drug release from nanoparticles, fluid flow, mass transport, and pharmacodynamic governing equations. Owing to the rapid development of computational tools, modeling the different phenomena in multi-scale complex problems involved in drug delivery to tumors has become possible. In the present study, we present an in-depth review of recent advances in the mathematical modeling of TUS-mediated DDSs for cancer treatment. A detailed discussion is also provided on applying these computational models to improve the clinical translation for applications in cancer treatment. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
| | - Tyler K Hornsby
- Department of Physics, Toronto Metropolitan University, 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
| | - 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
| |
Collapse
|
25
|
Scutigliani EM, van Hattum J, Lobo-Cerna F, Kruyswijk J, Myrcha M, Dekkers FEGA, Hoebe RA, Edwards F, Oppelaar JJ, Vogt L, Bootsma S, Bijlsma MF, Picavet DI, Crezee J, Oddens JR, de Reijke TM, Krawczyk PM. Perturbation of Copper Homeostasis Sensitizes Cancer Cells to Elevated Temperature. Int J Mol Sci 2023; 25:423. [PMID: 38203594 PMCID: PMC10779418 DOI: 10.3390/ijms25010423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Temporary elevation of tumor temperature, also known as hyperthermia, is a safe and well-tolerated treatment modality. The efficacy of hyperthermia can be improved by efficient thermosensitizers, and various candidate drugs, including inhibitors of the heat stress response, have been explored in vitro and in animal models, but clinically relevant thermosensitizers are lacking. Here, we employ unbiased in silico approaches to uncover new mechanisms and compounds that could be leveraged to increase the thermosensitivity of cancer cells. We then focus on elesclomol, a well-performing compound, which amplifies cell killing by hyperthermia by 5- to 20-fold in cell lines and outperforms clinically applied chemotherapy when combined with hyperthermia in vitro. Surprisingly, our findings suggest that the thermosensitizing effects of elesclomol are independent of its previously reported modes of action but depend on copper shuttling. Importantly, we show that, like elesclomol, multiple other copper shuttlers can thermosensitize, suggesting that disturbing copper homeostasis could be a general strategy for improving the efficacy of hyperthermia.
Collapse
Affiliation(s)
- Enzo M. Scutigliani
- Department of Medical Biology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (E.M.S.); (F.L.-C.); (J.K.); (M.M.); (F.E.G.A.D.); (F.E.); (D.I.P.)
- Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands; (J.v.H.); (J.C.); (J.R.O.); (T.M.d.R.)
| | - Jons van Hattum
- Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands; (J.v.H.); (J.C.); (J.R.O.); (T.M.d.R.)
- Department of Urology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Fernando Lobo-Cerna
- Department of Medical Biology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (E.M.S.); (F.L.-C.); (J.K.); (M.M.); (F.E.G.A.D.); (F.E.); (D.I.P.)
- Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands; (J.v.H.); (J.C.); (J.R.O.); (T.M.d.R.)
| | - Joanne Kruyswijk
- Department of Medical Biology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (E.M.S.); (F.L.-C.); (J.K.); (M.M.); (F.E.G.A.D.); (F.E.); (D.I.P.)
- Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands; (J.v.H.); (J.C.); (J.R.O.); (T.M.d.R.)
| | - Maja Myrcha
- Department of Medical Biology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (E.M.S.); (F.L.-C.); (J.K.); (M.M.); (F.E.G.A.D.); (F.E.); (D.I.P.)
- Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands; (J.v.H.); (J.C.); (J.R.O.); (T.M.d.R.)
| | - Frederique E. G. A. Dekkers
- Department of Medical Biology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (E.M.S.); (F.L.-C.); (J.K.); (M.M.); (F.E.G.A.D.); (F.E.); (D.I.P.)
- Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands; (J.v.H.); (J.C.); (J.R.O.); (T.M.d.R.)
| | - Ron A. Hoebe
- Department of Medical Biology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (E.M.S.); (F.L.-C.); (J.K.); (M.M.); (F.E.G.A.D.); (F.E.); (D.I.P.)
| | - Finn Edwards
- Department of Medical Biology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (E.M.S.); (F.L.-C.); (J.K.); (M.M.); (F.E.G.A.D.); (F.E.); (D.I.P.)
- Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands; (J.v.H.); (J.C.); (J.R.O.); (T.M.d.R.)
| | - Jetta J. Oppelaar
- Department of Internal Medicine, Section of Nephrology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (J.J.O.); (L.V.)
- Amsterdam Cardiovascular Sciences, Microcirculation, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Liffert Vogt
- Department of Internal Medicine, Section of Nephrology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (J.J.O.); (L.V.)
- Amsterdam Cardiovascular Sciences, Microcirculation, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Sanne Bootsma
- Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (S.B.); (M.F.B.)
- Cancer Center Amsterdam, Cancer Biology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Oncode Institute, Jaarbeursplein 6, 3521 AL Utrecht, The Netherlands
| | - Maarten F. Bijlsma
- Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (S.B.); (M.F.B.)
- Cancer Center Amsterdam, Cancer Biology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Oncode Institute, Jaarbeursplein 6, 3521 AL Utrecht, The Netherlands
| | - Daisy I. Picavet
- Department of Medical Biology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (E.M.S.); (F.L.-C.); (J.K.); (M.M.); (F.E.G.A.D.); (F.E.); (D.I.P.)
| | - Johannes Crezee
- Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands; (J.v.H.); (J.C.); (J.R.O.); (T.M.d.R.)
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Jorg R. Oddens
- Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands; (J.v.H.); (J.C.); (J.R.O.); (T.M.d.R.)
- Department of Urology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Theo M. de Reijke
- Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands; (J.v.H.); (J.C.); (J.R.O.); (T.M.d.R.)
- Department of Urology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Przemek M. Krawczyk
- Department of Medical Biology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (E.M.S.); (F.L.-C.); (J.K.); (M.M.); (F.E.G.A.D.); (F.E.); (D.I.P.)
- Cancer Center Amsterdam, Treatment and Quality of Life, Cancer Biology and Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands; (J.v.H.); (J.C.); (J.R.O.); (T.M.d.R.)
| |
Collapse
|
26
|
Qin M, Xia H, Xu W, Chen B, Wang Y. The spatiotemporal journey of nanomedicines in solid tumors on their therapeutic efficacy. Adv Drug Deliv Rev 2023; 203:115137. [PMID: 37949414 DOI: 10.1016/j.addr.2023.115137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
The rapid development of nanomedicines is revolutionizing the landscape of cancer treatment, while effectively delivering them into solid tumors remains a formidable challenge. Currently, there is a huge disconnect on therapeutic response between regulatory approved nanomedicines and laboratory reported nanoparticles. The discrepancy is mainly resulted from the failure of using the classic overall pharmacokinetics behaviors of nanomedicines in tumors to predict the antitumor efficacy. Increasing evidence has revealed that the therapeutic efficacy predominantly relies on the intratumoral spatiotemporal distribution of nanomedicines. This review focuses on the spatiotemporal distribution of systemically administered chemotherapeutic nanomedicines in solid tumor. Firstly, the intratumoral biological barriers that regulate the spatiotemporal distribution of nanomedicines are described in detail. Next, the influences on antitumor efficacy caused by the spatial distribution and temporal drug release of nanomedicines are emphatically analyzed. Then, current methodologies for evaluating the spatiotemporal distribution of nanomedicines are summarized. Finally, the advanced strategies to positively modulate the spatiotemporal distribution of nanomedicines for an optimal tumor therapy are comprehensively reviewed.
Collapse
Affiliation(s)
- Mengmeng Qin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Heming Xia
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wenhao Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Binlong Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China; Chemical Biology Center, Peking University, Beijing, China.
| |
Collapse
|
27
|
AIUM Official Statement for the Statement on Biological Effects of Therapeutic Ultrasound. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2023; 42:E68-E73. [PMID: 37584480 DOI: 10.1002/jum.16315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/17/2023]
|
28
|
Lyons B, Balkaran JPR, Dunn-Lawless D, Lucian V, Keller SB, O’Reilly CS, Hu L, Rubasingham J, Nair M, Carlisle R, Stride E, Gray M, Coussios C. Sonosensitive Cavitation Nuclei-A Customisable Platform Technology for Enhanced Therapeutic Delivery. Molecules 2023; 28:7733. [PMID: 38067464 PMCID: PMC10708135 DOI: 10.3390/molecules28237733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Ultrasound-mediated cavitation shows great promise for improving targeted drug delivery across a range of clinical applications. Cavitation nuclei-sound-sensitive constructs that enhance cavitation activity at lower pressures-have become a powerful adjuvant to ultrasound-based treatments, and more recently emerged as a drug delivery vehicle in their own right. The unique combination of physical, biological, and chemical effects that occur around these structures, as well as their varied compositions and morphologies, make cavitation nuclei an attractive platform for creating delivery systems tuned to particular therapeutics. In this review, we describe the structure and function of cavitation nuclei, approaches to their functionalization and customization, various clinical applications, progress toward real-world translation, and future directions for the field.
Collapse
Affiliation(s)
- Brian Lyons
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Joel P. R. Balkaran
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Darcy Dunn-Lawless
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Veronica Lucian
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Sara B. Keller
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Colm S. O’Reilly
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford OX1 3PJ, UK;
| | - Luna Hu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Jeffrey Rubasingham
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Malavika Nair
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Constantin Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| |
Collapse
|
29
|
Grandhi S, Al-Tabakha M, Avula PR. Enhancement of Liver Targetability through Statistical Optimization and Surface Modification of Biodegradable Nanocapsules Loaded with Lamivudine. Adv Pharmacol Pharm Sci 2023; 2023:8902963. [PMID: 38029229 PMCID: PMC10676277 DOI: 10.1155/2023/8902963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/29/2023] [Accepted: 11/11/2023] [Indexed: 12/01/2023] Open
Abstract
The intention of the current work was to develop and optimize the formulation of biodegradable polymeric nanocapsules for lamivudine (LMV) in order to obtain desired physical characteristics so as to have improved liver targetability. Nanocapsules were prepared in this study as aqueous-core nanocapsules (ACNs) with poly(lactide-co-glycolide) using a modified multiple emulsion technique. LMV was taken as a model drug to investigate the potential of ACNs developed in this work in achieving the liver targetability. Three formulations factors were chosen and 33 factorial design was adopted. The selected formulation factors were optimized statistically so as to have the anticipated characteristics of the ACNs viz. maximum entrapment efficiency, minimum particle size, and less drug release rate constant. The optimized LMV-ACNs were found to have 71.54 ± 1.93% of entrapment efficiency and 288.36 ± 2.53 nm of particle size with zeta potential of -24.7 ± 1.2 mV and 0.095 ± 0.006 h-1 of release rate constant. This optimized formulation was subjected to surface modification by treating with sodium lauryl sulphate (SLS), which increased the zeta potential to a maximum of -41.6 ± 1.3 mV at a 6 mM concentration of SLS. The results of in vivo pharmacokinetics from blood and liver tissues indicated that hepatic bioavailability of LMV was increased from 13.78 ± 3.48 μg/mL ∗ h for LMV solution to 32.94 ± 5.12 μg/mL ∗ h for the optimized LMV-ACNs and to 54.91 ± 6.68 μg/mL ∗ h for the surface-modified LMV-ACNs.
Collapse
Affiliation(s)
- Srikar Grandhi
- Department of Pharmaceutical Sciences, Vignan's Foundation for Science Technology and Research, Vadlamudi, Guntur 522213, India
| | - Moawia Al-Tabakha
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, P.O. Box 346, Ajman, UAE
- Centre of Medical and Bio-Allied Health Sciences Research Centre, Ajman University, P.O. Box 346, Ajman, UAE
| | - Prameela Rani Avula
- University College of Pharmaceutical Sciences, Acharya Nagarjuna University, Nagarjuna Nagar, Guntur 522510, India
| |
Collapse
|
30
|
Purohit MP, Roy KS, Xiang Y, Yu BJ, Azadian MM, Muwanga G, Hart AR, Taoube AK, Lopez DG, Airan RD. Acoustomechanically activatable liposomes for ultrasonic drug uncaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563690. [PMID: 37961368 PMCID: PMC10634775 DOI: 10.1101/2023.10.23.563690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Ultrasound-activatable drug-loaded nanocarriers enable noninvasive and spatiotemporally-precise on-demand drug delivery throughout the body. However, most systems for ultrasonic drug uncaging utilize cavitation or heating as the drug release mechanism and often incorporate relatively exotic excipients into the formulation that together limit the drug-loading potential, stability, and clinical translatability and applicability of these systems. Here we describe an alternate strategy for the design of such systems in which the acoustic impedance and osmolarity of the internal liquid phase of a drug-loaded particle is tuned to maximize ultrasound-induced drug release. No gas phase, cavitation, or medium heating is necessary for the drug release mechanism. Instead, a non-cavitation-based mechanical response to ultrasound mediates the drug release. Importantly, this strategy can be implemented with relatively common pharmaceutical excipients, as we demonstrate here by implementing this mechanism with the inclusion of a few percent sucrose into the internal buffer of a liposome. Further, the ultrasound protocols sufficient for in vivo drug uncaging with this system are achievable with current clinical therapeutic ultrasound systems and with intensities that are within FDA and society guidelines for safe transcranial ultrasound application. Finally, this current implementation of this mechanism should be versatile and effective for the loading and uncaging of any therapeutic that may be loaded into a liposome, as we demonstrate for four different drugs in vitro, and two in vivo. These acoustomechanically activatable liposomes formulated with common pharmaceutical excipients promise a system with high clinical translational potential for ultrasonic drug uncaging of myriad drugs of clinical interest.
Collapse
Affiliation(s)
| | - Kanchan Sinha Roy
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
| | - Yun Xiang
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
| | - Brenda J. Yu
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Biophysics Program, Stanford University, Stanford, CA, 94305 USA
| | - Matine M. Azadian
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Neurosciences Program, Stanford University, Stanford, CA, 94305 USA
| | - Gabriella Muwanga
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Neurosciences Program, Stanford University, Stanford, CA, 94305 USA
| | - Alex R. Hart
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Department of Chemistry, Stanford University, Stanford, CA, 94305 USA
| | - Ali K. Taoube
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
| | - Diego Gomez Lopez
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Department of Medicine, Health, and Society, Vanderbilt University, Nashville, TN 37235 USA
| | - Raag D. Airan
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305 USA
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305 USA
| |
Collapse
|
31
|
Spiers L, Gray M, Lyon P, Sivakumar S, Bekkali N, Scott S, Collins L, Carlisle R, Wu F, Middleton M, Coussios C. Clinical trial protocol for PanDox: a phase I study of targeted chemotherapy delivery to non-resectable primary pancreatic tumours using thermosensitive liposomal doxorubicin (ThermoDox®) and focused ultrasound. BMC Cancer 2023; 23:896. [PMID: 37741968 PMCID: PMC10517508 DOI: 10.1186/s12885-023-11228-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 07/24/2023] [Indexed: 09/25/2023] Open
Abstract
BACKGROUND The dense stroma of pancreatic ductal adenocarcinomas is a major barrier to drug delivery. To increase the local drug diffusion gradient, high doses of chemotherapeutic agent doxorubicin can be released from thermally-sensitive liposomes (ThermoDox®) using ultrasound-mediated hyperthermia at the tumour target. PanDox is designed as a Phase 1 single centre study to investigate enhancing drug delivery to adult patients with non-operable pancreatic ductal adenocarcinomas. The study compares a single cycle of either conventional doxorubicin alone or ThermoDox® with focused ultrasound-induced hyperthermia for targeted drug release. METHODS Adults with non-resectable pancreatic ductal adenocarcinoma are allocated to receive a single cycle of either doxorubicin alone (Arm A) or ThermoDox® with focused ultrasound-induced hyperthermia (Arm B), based on patient- and tumour-specific safety conditions. Participants in Arm B will undergo a general anaesthetic and pre-heating of the tumour by extra-corporal focused ultrasound (FUS). Rather than employing invasive thermometry, ultrasound parameters are derived from a patient-specific treatment planning model to reach the 41 °C target temperature for drug release. ThermoDox® is then concurrently infused with further ultrasound exposure. Tumour biopsies at the targeted site from all patients are analysed post-treatment using high performance liquid chromatography to quantify doxorubicin delivered to the tumour. The primary endpoint is defined as a statistically significant enhancement in concentration of total intra-tumoural doxorubicin, comparing samples from patients receiving liposomal drug with FUS to free drug alone. Participants are followed for 21 days post-treatment to assess secondary endpoints, including radiological assessment to measure changes in tumour activity by Positron Emission Tomography Response Criteria in Solid Tumours (PERCIST) criteria, adverse events and patient-reported symptoms. DISCUSSION This early phase study builds on previous work targeting tumours in the liver to investigate whether enhancement of chemotherapy delivery using ultrasound-mediated hyperthermia can be translated to the stroma-dense environment of pancreatic ductal adenocarcinoma. If successful, it could herald a new approach towards managing these difficult-to-treat tumours. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04852367 . Registered 21st April 2022. EudraCT number: 2019-003950-10 (Registered 2019) Iras Project ID: 272253 (Registered 2019) Ethics Number: 20/EE/0284.
Collapse
Affiliation(s)
- Laura Spiers
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Michael Gray
- NIHR Oxford Biomedical Research Centre, Oxford, UK
- Institute of Biomedical Engineering, University of Oxford, Marcella Wing, Botnar Research Centre, Old Rd, Headington, Oxford, OX3 7LD, UK
| | - Paul Lyon
- Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK
| | - Shivan Sivakumar
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK
| | - Noor Bekkali
- Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK
| | - Shaun Scott
- Nuffield Department of Anaesthetics, John Radcliffe Hospital, Oxford, OX3 7LE, UK
| | - Linda Collins
- Department of Oncology, Oncology Clinical Trials Office (OCTO), University of Oxford, Oxford, UK
| | - Robert Carlisle
- Institute of Biomedical Engineering, University of Oxford, Marcella Wing, Botnar Research Centre, Old Rd, Headington, Oxford, OX3 7LD, UK
| | - Feng Wu
- Nuffield Department of Surgery, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Mark Middleton
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Constantin Coussios
- Institute of Biomedical Engineering, University of Oxford, Marcella Wing, Botnar Research Centre, Old Rd, Headington, Oxford, OX3 7LD, UK.
| |
Collapse
|
32
|
Lahooti B, Akwii RG, Zahra FT, Sajib MS, Lamprou M, Alobaida A, Lionakis MS, Mattheolabakis G, Mikelis CM. Targeting endothelial permeability in the EPR effect. J Control Release 2023; 361:212-235. [PMID: 37517543 DOI: 10.1016/j.jconrel.2023.07.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/19/2023] [Accepted: 07/23/2023] [Indexed: 08/01/2023]
Abstract
The characteristics of the primary tumor blood vessels and the tumor microenvironment drive the enhanced permeability and retention (EPR) effect, which confers an advantage towards enhanced delivery of anti-cancer nanomedicine and has shown beneficial effects in preclinical models. Increased vascular permeability is a landmark feature of the tumor vessels and an important driver of the EPR. The main focus of this review is the endothelial regulation of vascular permeability. We discuss current challenges of targeting vascular permeability towards clinical translation and summarize the structural components and mechanisms of endothelial permeability, the principal mediators and signaling players, the targeted approaches that have been used and their outcomes to date. We also critically discuss the effects of the tumor-infiltrating immune cells, their interplay with the tumor vessels and the impact of immune responses on nanomedicine delivery, the impact of anti-angiogenic and tumor-stroma targeting approaches, and desirable nanoparticle design approaches for greater translational benefit.
Collapse
Affiliation(s)
- Behnaz Lahooti
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Racheal G Akwii
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Fatema Tuz Zahra
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Md Sanaullah Sajib
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Margarita Lamprou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece
| | - Ahmed Alobaida
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA.
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece.
| |
Collapse
|
33
|
Huang D, Wang J, Wen B, Zhao Y. Emerging diagnostic and therapeutic technologies based on ultrasound-triggered biomaterials. MATERIALS FUTURES 2023; 2:032001. [DOI: 10.1088/2752-5724/acdf05] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Abstract
Abstract
Ultrasound (US) is a kind of acoustic wave with frequency higher than 20 kHz. Learning from the echo detection ability of bats and dolphins, scientists applied US for clinical imaging by sending out US waves and detecting echoes with shifted intensities and frequencies from human tissue. US has long played a critical role in noninvasive, real-time, low-cost and portable diagnostic imaging. With the in-depth study of US in multidisciplinary fields, US and US-responsive materials have shown practical value in not only disease diagnosis, but also disease treatment. In this review, we introduce the recently proposed and representative US-responsive materials for biomedical applications, including diagnostic and therapeutic applications. We focused on US-mediated physicochemical therapies, such as sonodynamic therapy, high-intensity focused US ablation, sonothermal therapy, thrombolysis, etc, and US-controlled delivery of chemotherapeutics, gases, genes, proteins and bacteria. We conclude with the current challenges facing the clinical translation of smart US-responsive materials and prospects for the future development of US medicine.
Collapse
|
34
|
Khan F, Jones K, Lyon P. Immune checkpoint inhibition: a future guided by radiology. Br J Radiol 2023; 96:20220565. [PMID: 36752570 PMCID: PMC10321249 DOI: 10.1259/bjr.20220565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 01/04/2023] [Accepted: 01/29/2023] [Indexed: 02/09/2023] Open
Abstract
The limitation of the function of antitumour immune cells is a common hallmark of cancers that enables their survival. As such, the potential of immune checkpoint inhibition (ICI) acts as a paradigm shift in the treatment of a range of cancers but has not yet been fully capitalised. Combining minimally and non-invasive locoregional therapies offered by radiologists with ICI is now an active field of research with the aim of furthering therapeutic capabilities in medical oncology. In parallel to this impending advancement, the "imaging toolbox" available to radiologists is also growing, enabling more refined tumour characterisation as well as greater accuracy in evaluating responses to therapy. Options range from metabolite labelling to cellular localisation to immune checkpoint screening. It is foreseeable that these novel imaging techniques will be integrated into personalised treatment algorithms. This growth in the field must include updating the current standardised imaging criteria to ensure they are fit for purpose. Such criteria is crucial to both appropriately guide clinical decision-making regarding next steps of treatment, but also provide reliable prognosis. Quantitative approaches to these novel imaging techniques are also already being investigated to further optimise personalised therapeutic decision-making. The therapeutic potential of specific ICIs and locoregional therapies could be determined before administration thus limiting unnecessary side-effects whilst maintaining efficacy. Several radiological aspects of oncological care are advancing simultaneously. Therefore, it is essential that each development is assessed for clinical use and optimised to ensure the best treatment decisions are being offered to the patient. In this review, we discuss state of the art advances in novel functional imaging techniques in the field of immuno-oncology both pre-clinically and clinically.
Collapse
Affiliation(s)
- Faraaz Khan
- Foundation Doctor, Buckinghamshire Hospitals NHS Trust, Amersham, Buckinghamshire, United Kingdom
| | - Keaton Jones
- Academic Clinical Lecturer Nuffield Department of Surgical Sciences University of Oxford, Wellington Square, Oxford, United Kingdom
| | - Paul Lyon
- Consultant Radiologist, Department of Radiology, Oxford University Hospitals, Headington, Oxford, United Kingdom
| |
Collapse
|
35
|
Honari A, Sirsi SR. The Evolution and Recent Trends in Acoustic Targeting of Encapsulated Drugs to Solid Tumors: Strategies beyond Sonoporation. Pharmaceutics 2023; 15:1705. [PMID: 37376152 DOI: 10.3390/pharmaceutics15061705] [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: 04/13/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Despite recent advancements in ultrasound-mediated drug delivery and the remarkable success observed in pre-clinical studies, no delivery platform utilizing ultrasound contrast agents has yet received FDA approval. The sonoporation effect was a game-changing discovery with a promising future in clinical settings. Various clinical trials are underway to assess sonoporation's efficacy in treating solid tumors; however, there are disagreements on its applicability to the broader population due to long-term safety issues. In this review, we first discuss how acoustic targeting of drugs gained importance in cancer pharmaceutics. Then, we discuss ultrasound-targeting strategies that have been less explored yet hold a promising future. We aim to shed light on recent innovations in ultrasound-based drug delivery including newer designs of ultrasound-sensitive particles specifically tailored for pharmaceutical usage.
Collapse
Affiliation(s)
- Arvin Honari
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Shashank R Sirsi
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
36
|
Imran KM, Tintera B, Morrison HA, Tupik JD, Nagai-Singer MA, Ivester H, Council-Troche M, Edwards M, Coutermarsh-Ott S, Byron C, Clark-Deener S, Uh K, Lee K, Boulos P, Rowe C, Coviello C, Allen IC. Improved Therapeutic Delivery Targeting Clinically Relevant Orthotopic Human Pancreatic Tumors Engrafted in Immunocompromised Pigs Using Ultrasound-Induced Cavitation: A Pilot Study. Pharmaceutics 2023; 15:1585. [PMID: 37376034 PMCID: PMC10302458 DOI: 10.3390/pharmaceutics15061585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/03/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Pancreatic tumors can be resistant to drug penetration due to high interstitial fluid pressure, dense stroma, and disarrayed vasculature. Ultrasound-induced cavitation is an emerging technology that may overcome many of these limitations. Low-intensity ultrasound, coupled with co-administered cavitation nuclei consisting of gas-stabilizing sub-micron scale SonoTran Particles, is effective at increasing therapeutic antibody delivery to xenograft flank tumors in mouse models. Here, we sought to evaluate the effectiveness of this approach in situ using a large animal model that mimics human pancreatic cancer patients. Immunocompromised pigs were surgically engrafted with human Panc-1 pancreatic ductal adenocarcinoma (PDAC) tumors in targeted regions of the pancreas. These tumors were found to recapitulate many features of human PDAC tumors. Animals were intravenously injected with the common cancer therapeutics Cetuximab, gemcitabine, and paclitaxel, followed by infusion with SonoTran Particles. Select tumors in each animal were targeted with focused ultrasound to induce cavitation. Cavitation increased the intra-tumor concentrations of Cetuximab, gemcitabine, and paclitaxel by 477%, 148%, and 193%, respectively, compared to tumors that were not targeted with ultrasound in the same animals. Together, these data show that ultrasound-mediated cavitation, when delivered in combination with gas-entrapping particles, improves therapeutic delivery in pancreatic tumors under clinically relevant conditions.
Collapse
Affiliation(s)
- Khan Mohammad Imran
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA 24061, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Benjamin Tintera
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Holly A. Morrison
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Juselyn D. Tupik
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Margaret A. Nagai-Singer
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Hannah Ivester
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA 24061, USA
| | - McAlister Council-Troche
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Michael Edwards
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Sheryl Coutermarsh-Ott
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Christopher Byron
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Sherrie Clark-Deener
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Kyungjun Uh
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO 65211, USA
| | - Kiho Lee
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO 65211, USA
| | - Paul Boulos
- OxSonics Therapeutics, Oxford Science Park, Oxford OX4 4GA, UK
| | - Cliff Rowe
- OxSonics Therapeutics, Oxford Science Park, Oxford OX4 4GA, UK
| | | | - Irving C. Allen
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA 24061, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| |
Collapse
|
37
|
Huang D, Wang J, Song C, Zhao Y. Ultrasound-responsive matters for biomedical applications. Innovation (N Y) 2023; 4:100421. [PMID: 37192908 PMCID: PMC10182333 DOI: 10.1016/j.xinn.2023.100421] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/03/2023] [Indexed: 05/18/2023] Open
Abstract
Ultrasound (US) is a biofavorable mechanical wave that has shown practical significance in biomedical fields. Due to the cavitation effect, sonoluminescence, sonoporation, pyrolysis, and other biophysical and chemical effects, a wide range of matters have been elucidated to be responsive to the stimulus of US. This review addresses and discusses current developments in US-responsive matters, including US-breakable intermolecular conjugations, US-catalytic sonosensitizers, fluorocarbon compounds, microbubbles, and US-propelled micro- and nanorobots. Meanwhile, the interactions between US and advanced matters create various biochemical products and enhanced mechanical effects, leading to the exploration of potential biomedical applications, from US-facilitated biosensing and diagnostic imaging to US-induced therapeutic applications and clinical translations. Finally, the current challenges are summarized and future perspectives on US-responsive matters in biomedical applications and clinical translations are proposed.
Collapse
Affiliation(s)
- Danqing Huang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210002, China
| | - Jinglin Wang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210002, China
| | - Chuanhui Song
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210002, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210002, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| |
Collapse
|
38
|
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: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [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
|
39
|
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
|
40
|
Ashar H, Ranjan A. Immunomodulation and targeted drug delivery with high intensity focused ultrasound (HIFU): Principles and mechanisms. Pharmacol Ther 2023; 244:108393. [PMID: 36965581 DOI: 10.1016/j.pharmthera.2023.108393] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/04/2023] [Accepted: 03/20/2023] [Indexed: 03/27/2023]
Abstract
High intensity focused ultrasound (HIFU) is a non-invasive and non-ionizing sonic energy-based therapeutic technology for inducing thermal and non-thermal effects in tissues. Depending on the parameters, HIFU can ablate tissues by heating them to >55 °C to induce denaturation and coagulative necrosis, improve radio- and chemo-sensitizations and local drug delivery from nanoparticles at moderate hyperthermia (∼41-43 °C), and mechanically fragment cells using acoustic cavitation (also known as histotripsy). HIFU has already emerged as an attractive modality for treating human & veterinary cancers, infectious diseases, and neuromodulation. Herein, we comprehensively review the role of HIFU in enhancing drug delivery and immunomodulation in soft and calcified tissues. Specifically, the ability of HIFU to improve adjuvant treatments from various classes of therapeutic agents are described. These crucial insights highlight the opportunities and challenges of HIFU technology and its potential to support new clinical trials and translation to patients.
Collapse
Affiliation(s)
- Harshini Ashar
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, United States of America
| | - Ashish Ranjan
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, United States of America.
| |
Collapse
|
41
|
Zhu Y, Li Q, Wang C, Hao Y, Yang N, Chen M, Ji J, Feng L, Liu Z. Rational Design of Biomaterials to Potentiate Cancer Thermal Therapy. Chem Rev 2023. [PMID: 36912061 DOI: 10.1021/acs.chemrev.2c00822] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Cancer thermal therapy, also known as hyperthermia therapy, has long been exploited to eradicate mass lesions that are now defined as cancer. With the development of corresponding technologies and equipment, local hyperthermia therapies such as radiofrequency ablation, microwave ablation, and high-intensity focused ultrasound, have has been validated to effectively ablate tumors in modern clinical practice. However, they still face many shortcomings, including nonspecific damages to adjacent normal tissues and incomplete ablation particularly for large tumors, restricting their wide clinical usage. Attributed to their versatile physiochemical properties, biomaterials have been specially designed to potentiate local hyperthermia treatments according to their unique working principles. Meanwhile, biomaterial-based delivery systems are able to bridge hyperthermia therapies with other types of treatment strategies such as chemotherapy, radiotherapy and immunotherapy. Therefore, in this review, we discuss recent progress in the development of functional biomaterials to reinforce local hyperthermia by functioning as thermal sensitizers to endow more efficient tumor-localized thermal ablation and/or as delivery vehicles to synergize with other therapeutic modalities for combined cancer treatments. Thereafter, we provide a critical perspective on the further development of biomaterial-assisted local hyperthermia toward clinical applications.
Collapse
Affiliation(s)
- Yujie Zhu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Quguang Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Chunjie Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Yu Hao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Nailin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, Zhejiang, P.R. China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, Zhejiang, P.R. China
| | - Liangzhu Feng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| |
Collapse
|
42
|
Papaioannou L, Kolokithas-Ntoukas A, Karkaletsou L, Didaskalou S, Koffa MD, Avgoustakis K. NIR-responsive, lapatinib-loaded gold nanorods for combined photothermal and pharmacological treatment of HER2 positive breast cancer: In vitro evaluation and cell studies. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2023]
|
43
|
Huang D, Wang J, Che J, Wen B, Kong W. Ultrasound-responsive microparticles from droplet microfluidics. BIOMEDICAL TECHNOLOGY 2023; 1:1-9. [DOI: 10.1016/j.bmt.2022.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
|
44
|
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: 66] [Impact Index Per Article: 33.0] [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
|
45
|
Ultrasound-excited temozolomide sonosensitization induces necroptosis in glioblastoma. Cancer Lett 2023; 554:216033. [PMID: 36493901 DOI: 10.1016/j.canlet.2022.216033] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/22/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Temozolomide (TMZ) has been determined to be the chemotherapeutic drug with efficacy for glioblastoma (GBM). Thus, potentiating the therapeutic effect of TMZ can undoubtedly yield twice the result with half the effort. In this study, we found for the first time that TMZ can produce reactive oxygen species (ROS) under the influence of ultrasound (US). This property allows TMZ-US therapy to have better efficacy in the treatment of GBM. Given that the increasing use of US in central nervous system (CNS) diseases and the importance of TMZ for GBM therapy, our results will facilitate the development of TMZ-associated glioblastoma therapies. Moreover, we found that chemotherapeutic drugs might have the ability to generate ROS under the excitation of US. On a larger scale, our findings may be applicable to a wide range of known drugs.
Collapse
|
46
|
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
|
47
|
Moradi Kashkooli, Jakhmola A, Ferrier GA, Hornsby TK, Tavakkoli J(J, Kolios MC. Integrating Therapeutic Ultrasound With Nanosized Drug Delivery Systems in the Battle Against Cancer. Technol Cancer Res Treat 2023; 22:15330338231211472. [PMID: 37946517 PMCID: PMC10637173 DOI: 10.1177/15330338231211472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/27/2017] [Accepted: 10/16/2023] [Indexed: 11/12/2023] Open
Abstract
Controlled, localized, and timely activation of nanosized drug delivery systems (NSDDSs), using an external stimulus such as therapeutic ultrasound (TUS), can improve the efficacy of cancer treatments compared to either conventional chemotherapy methods or passive NSDDSs alone. Specifically, TUS induces thermal and mechanical effects that trigger drug release from NSDDSs and overcomes drug delivery barriers in tumor microenvironments to allow nanoparticle drug carriers to penetrate more deeply into tumor tissue while minimizing side effects. This review highlights recent advancements, contemplates future prospects, and addresses challenges in using TUS-mediated NSDDSs for cancer treatment, encompassing preclinical and clinical applications.
Collapse
Affiliation(s)
- Moradi Kashkooli
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
| | - Anshuman Jakhmola
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
| | - Graham A. Ferrier
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
| | - Tyler K. Hornsby
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
| | - Jahangir (Jahan) Tavakkoli
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Michael C. Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| |
Collapse
|
48
|
Gray M, Spiers L, Coussios C. Effects of human tissue acoustic properties, abdominal wall shape, and respiratory motion on ultrasound-mediated hyperthermia for targeted drug delivery to pancreatic tumors. Int J Hyperthermia 2022; 39:918-934. [PMID: 35853611 PMCID: PMC9612938 DOI: 10.1080/02656736.2022.2091799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background PanDox is a Phase-1 trial of chemotherapeutic drug delivery to pancreatic tumors using ultrasound-mediated hyperthermia to release doxorubicin from thermally sensitive liposomes. This report describes trial-related hyperthermia simulations featuring: (i) new ultrasonic properties of human pancreatic tissues, (ii) abdomen deflections imposed by a water balloon, and (iii) respiration-driven organ motion. Methods Pancreas heating simulations were carried out using three patient body models. Pancreas acoustic properties were varied between values found in the literature and those determined from our human tissue study. Acoustic beam distortion was assessed with and without balloon-induced abdomen deformation. Target heating was assessed for static, normal respiratory, and jet-ventilation-controlled pancreas motion. Results Human pancreatic tumor attenuation is 63% of the literature values, so that pancreas treatments require commensurately higher input intensity to achieve adequate hyperthermia. Abdominal wall deformation decreased the peak field pressure by as much as 3.5 dB and refracted the focal spot by as much as 4.5 mm. These effects were thermally counteracted by sidelobe power deposition, so the net impact on achieving mild hyperthermia was small. Respiratory motion during moving beam hyperthermia produced localized regions overheated by more than 8.0 °C above the 4.0 °C volumetric goal. The use of jet ventilation reduced this excess to 0.7 °C and yielded temperature field uniformity that was nearly identical to having no respiratory motion. Conclusion Realistic modeling of the ultrasonic propagation environment is critical to achieving adequate mild hyperthermia without the use of real time thermometry for targeted drug delivery in pancreatic cancer patients.
Collapse
Affiliation(s)
- Michael Gray
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Laura Spiers
- NIHR Oxford Biomedical Research Centre, Oxford, UK.,Department of Oncology, University of Oxford, Oxford, UK
| | - Constantin Coussios
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
49
|
Amin M, Lammers T, Ten Hagen TLM. Temperature-sensitive polymers to promote heat-triggered drug release from liposomes: Towards bypassing EPR. Adv Drug Deliv Rev 2022; 189:114503. [PMID: 35998827 DOI: 10.1016/j.addr.2022.114503] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/14/2022] [Accepted: 08/17/2022] [Indexed: 01/24/2023]
Abstract
Heat-triggered drug release from temperature-sensitive nanocarriers upon the application of mild hyperthermia is a promising approach to achieve site-specific delivery of drugs. The combination of mild hyperthermia (41-42 °C) and temperature-sensitive liposomes (TSL) that undergo lipid phase-transition and drug release has been studied extensively and has shown promising therapeutic outcome in a variety of animal tumor models as well as initial indications of success in humans. Sensitization of liposomes to mild hyperthermia by means of exploiting the thermal behavior of temperature-sensitive polymers (TSP) provides novel opportunities. Recently, TSP-modified liposomes (TSPL) have shown potential for enhancing tumor-directed drug delivery, either by triggered drug release or by triggered cell interactions in response to heat. In this review, we describe different classes of TSPL, and analyze and discuss the mechanisms and kinetics of content release from TSPL in response to local heating. In addition, the impact of lipid composition, polymer and copolymer characteristics, serum components and PEGylation on the mechanism of content release and TSPL performance is addressed. This is done from the perspective of rationally designing TSPL, with the overall goal of conceiving efficient strategies to increase the efficacy of TSPL plus hyperthermia to improve the outcome of targeted anticancer therapy.
Collapse
Affiliation(s)
- Mohamadreza Amin
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Center for Biohybrid Medical Systems, Aachen, Germany.
| | - Timo L M Ten Hagen
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
50
|
Chan H, Chang HY, Lin WL, Chen GS. Large-Volume Focused-Ultrasound Mild Hyperthermia for Improving Blood-Brain Tumor Barrier Permeability Application. Pharmaceutics 2022; 14:pharmaceutics14102012. [PMID: 36297445 PMCID: PMC9610093 DOI: 10.3390/pharmaceutics14102012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Mild hyperthermia can locally enhance permeability of the blood-tumor barrier in brain tumors, improving delivery of antitumor nanodrugs. However, a clinical transcranial focused ultrasound (FUS) system does not provide this modality yet. The study aimed at the development of the transcranial FUS technique dedicated for large-volume mild hyperthermia in the brain. Acoustic pressure, multiple-foci, temperature and thermal dose induced by FUS were simulated in the brain through the skull. A 1-MHz, 114-element, spherical helmet transducer was fabricated to verify large-volume hyperthermia in the phantom. The simulated results showed that two foci were simultaneously formed at (2, 0, 0) and (−2, 0, 0) and at (0, 2, 0) and (0, −2, 0), using the phases of focusing pattern 1 and the phases of focusing pattern 2, respectively. Switching two focusing patterns at 5 Hz produced a hyperthermic zone with an ellipsoid of 7 mm × 6 mm × 11 mm in the brain and the temperature was 41–45 °C in the ellipsoid as the maximum intensity was 150 W/cm2 and sonication time was 3 min. The phased array driven by switching two mode phases generated a 41 °C-contour region of 10 ± 1 mm × 8 ± 2 mm × 13 ± 2 mm in the phantom after 3-min sonication. Therefore, we have demonstrated our developed FUS technique for large-volume mild hyperthermia.
Collapse
Affiliation(s)
- Hsin Chan
- Institute of Biomedical Engineering, National Taiwan University, Taipei 100, Taiwan
| | - Hsin-Yun Chang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Win-Li Lin
- Institute of Biomedical Engineering, National Taiwan University, Taipei 100, Taiwan
| | - Gin-Shin Chen
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 35053, Taiwan
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Correspondence: ; Tel.: +886-37-206166 (ext. 37108)
| |
Collapse
|