1
|
Tao W, Lai Y, Zhou X, Yang G, Wu P, Yuan L. A narrative review: Ultrasound-Assisted drug delivery: Improving treatments via multiple mechanisms. ULTRASONICS 2025; 151:107611. [PMID: 40068411 DOI: 10.1016/j.ultras.2025.107611] [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: 05/07/2024] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 04/02/2025]
Abstract
Safe and efficient drug delivery is as important as drug development. Biological barriers, such as cell membranes, present significant challenges in drug delivery, especially for newly developed protein-, nucleic acid-, and cell-based drugs. Ultrasound-mediated drug delivery systems offer a promising strategy to overcome these challenges. Ultrasound, a mechanical wave with energy, produces thermal effects, cavitation, acoustic radiation, and other biophysical effects. Used alone or in combination with microbubbles or sonosensitizers, it breaks biological barriers, enhances targeted drug delivery, reduces adverse reactions, controls drug release, switches on/off drug functions, and ultimately improves therapeutic efficiency. Various ultrasound-mediated drug delivery methods, including transdermal drug delivery, nebulization, targeted microbubble destruction, and sonodynamic therapy, are being actively explored for the treatment of various diseases. This review article introduces the principles, advantages, and applications of ultrasound-mediated drug delivery methods for improved therapeutic outcomes and discusses future prospects in this field.
Collapse
Affiliation(s)
- Wenxin Tao
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Yubo Lai
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Xueying Zhou
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Guodong Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University Xi'an, Shaanxi 710032, China
| | - Pengying Wu
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Lijun Yuan
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China.
| |
Collapse
|
2
|
Liu D, Ling Y, Dong L, Zhang J, Li X, Chen X, Huang H, Deng J, Guo Y. Ultrasound-triggered drug-loaded nanobubbles for enhanced T cell recruitment in cancer chemoimmunotherapy. Biomaterials 2025; 317:123086. [PMID: 39805187 DOI: 10.1016/j.biomaterials.2025.123086] [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: 08/16/2024] [Revised: 12/09/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025]
Abstract
Chemotherapy combined with immunotherapy is a highly promising approach for treating tumors. However, chemotherapeutic drugs often fail to accumulate effectively at the tumor site after systemic administration and they lack sufficient immunogenicity to activate adaptive immunity, making an effective T-cell immune response within the tumor microenvironment difficult to achieve. Here, this work developed drug-loaded nanobubbles (DTX-R837@NBs) that encapsulate the chemotherapy drug docetaxel and the immune adjuvant R837 via a thin-film hydration method. Ultrasound-targeted nanobubble destruction promoted drug accumulation within tumor tissues and damaged tumor cells through the cavitation effect, inducing immunogenic cell death and releasing damage-associated molecular patterns to augment dendritic cell maturation. Notably, DTX-R837@NBs exhibited excellent contrast-enhanced ultrasound imaging capabilities, enabling the seamless integration of diagnosis and treatment. In combination with immune checkpoint blockade targeting programmed cell death protein 1 (PD-1), the generated immunological responses attacked residual tumor cells and ameliorated the immunosuppressive tumor microenvironment, inhibiting distant tumor growth and metastasis. Moreover, this strategy exhibited robust immune memory effects, effectively protecting the host and preventing tumor recurrence upon rechallenge. Overall, ultrasound-mediated DTX-R837@NBs combined with anti-PD-1 immune checkpoint blockade therapy exhibits robust antitumor efficiency, represent a promising strategy for overcoming immunotherapy resistance in cold tumors, and warrant further investigation for clinical translation.
Collapse
Affiliation(s)
- Deng Liu
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China; Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Yi Ling
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Li Dong
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Jun Zhang
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xin Li
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xuemei Chen
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Haiyun Huang
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| | - Jun Deng
- Institute of Burn Research, Southwest Hospital, State Key Lab of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, China.
| | - Yanli Guo
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
3
|
Chen LE, Nittayacharn P, Exner AA. Progress and potential of nanobubbles for ultrasound-mediated drug delivery. Expert Opin Drug Deliv 2025:1-24. [PMID: 40353846 DOI: 10.1080/17425247.2025.2505044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/18/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Despite much progress, nanomedicine-based drug therapies in oncology remain limited by systemic toxicity and insufficient particle accumulation in the tumor. To address these barriers, formulations responsive to external physical stimuli have emerged. One most promising system is the ultrasound stimulation of drug-loaded, gas-core particles (bubbles). Ultrasound induces bubble cavitation for cell and tissue permeabilization, triggers on-demand drug release, and provides opportunities for real-time imaging of delivery. AREAS COVERED Here, we focus on shell-stabilized, gas-core nanoparticles (also termed nanobubbles or ultrafine bubbles) and their role in ultrasound-mediated therapeutic delivery to tumors. This review frames the advantages of nanobubbles within the ongoing deficits in nanomedicine, describes mechanisms of ultrasound-mediated therapy, and details formulation techniques for nanobubble delivery systems. It then highlights the past decade of research in nanobubble-facilitated drug delivery for cancer therapy and anticipates new directions in the field. EXPERT OPINION Nanobubble ultrasound contrast agents offer a spatiotemporally triggerable therapeutic coupled with a safe, accessible imaging modality. Nanobubbles can be loaded with diverse therapeutic cargoes to treat disease and overcome numerous barriers limiting delivery to solid tumors. Close attention to formulation, characterization methods, acoustic testing parameters, and the biological mechanisms of nanobubble delivery will facilitate preclinical research toward clinical adoption.
Collapse
Affiliation(s)
- Laura E Chen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Pinunta Nittayacharn
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
4
|
Cai X, Liu Y, Luo G, Yu Z, Jiang C, Xu C. Ultrasound-assisted immunotherapy for malignant tumour. Front Immunol 2025; 16:1547594. [PMID: 40433381 PMCID: PMC12106521 DOI: 10.3389/fimmu.2025.1547594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Malignant tumour represents a significant global public health concern. The advent of immunotherapy has brought about a revolutionary shift in the landscape of tumour treatment, offering a ray of hope to patients across the globe. Immunotherapy strategies have demonstrated considerable promise in clinical trials. However, the immunosuppressive environment within the tumour microenvironment has constituted a significant obstacle to the advancement of immunotherapies. It is therefore imperative to develop more efficacious and personalised approaches. The utilisation of non-invasive ultrasound-assisted immunotherapy represents a promising strategy. Ultrasound has the capacity to induce an immune response and stimulate other drugs to achieve a specific response, thereby reducing the toxic side effects of treatment and enhancing the outcome of immunotherapy. This paper presents a systematic introduction to the various mechanisms related to ultrasound and reviews the recent advancements of ultrasound-assisted tumour immunotherapy, including ultrasonic ablation, combined application with contrast agents, and sonodynamic therapy.
Collapse
Affiliation(s)
- Xiaowen Cai
- School of Applied Biology, Shenzhen City Polytechnic, Shenzhen, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yujie Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Guosheng Luo
- School of Applied Biology, Shenzhen City Polytechnic, Shenzhen, China
| | - Zhanwang Yu
- School of Applied Biology, Shenzhen City Polytechnic, Shenzhen, China
| | - Cheng Jiang
- School of Applied Biology, Shenzhen City Polytechnic, Shenzhen, China
| | - Chuanshan Xu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
5
|
Engelen Y, Demuynck R, Ramon J, Breckpot K, De Smedt S, Lajoinie GPR, Braeckmans K, Krysko DV, Lentacker I. Immunogenic cell death as interplay between physical anticancer modalities and immunotherapy. J Control Release 2025:113721. [PMID: 40368187 DOI: 10.1016/j.jconrel.2025.113721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/16/2025]
Abstract
Current cancer treatment strategies in practice nowadays often face limitations in effectiveness due to factors such as resistance, recurrence, or suboptimal outcomes. Traditional approaches like chemotherapy often come with severe systemic side effects due to their non-specific action, prompting the development of more targeted therapies. Among these, physical ablation techniques such as radiotherapy (RT) and focused ultrasound (FUS) have gained attention for their ability to precisely target malignant tissues, reduce physical and mental stress for the patients, and minimize recovery time. These therapies also aim to stimulate the immune system through a process referred to as immunogenic cell death (ICD), enhancing the body's ability to fight cancer, explaining abscopal effects. RT has been the most established of the abovementioned techniques for decades, and will not be included in the review. While initially focused on complete tumor ablation, these techniques are now shifting towards milder, more controlled applications that induce ICD without extensive tissue damage. This review explores how physical ablation therapies can harness ICD to boost anticancer immunity, emphasizing their potential to complement immunotherapies and improve outcomes for cancer patients.
Collapse
Affiliation(s)
- Y Engelen
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium.
| | - R Demuynck
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - J Ramon
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium
| | - K Breckpot
- Laboratory for Molecular and Cellular Therapy, Translational Oncology Research Center, Department of Biomedical Sciences, Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - S De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| | - G P R Lajoinie
- Physics of Fluids Group, Technical Medical (TechMed) Center, and Max Planck Center for Complex Fluid Dynamics, University of Twente, Enschede, the Netherlands
| | - K Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium
| | - D V Krysko
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - I Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
6
|
Tian S, Liu Y, Tan Y, Cui X, Liu R, Liu C, Zhao Y, Xu K, Zhou J. Necroptosis-inducing nanobubbles for effective oxygen delivery and enhanced sonodynamic immunotherapy of breast cancer via UTND. Eur J Pharm Biopharm 2025; 210:114675. [PMID: 39993510 DOI: 10.1016/j.ejpb.2025.114675] [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: 09/07/2024] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
PURPOSE Breast cancer (BC) is a global threat to female health. Sonodynamic therapy (SDT) has been shown to induce apoptosis in tumor cells and trigger immunogenic cell death, leading to the activation of antitumor immunity. However, the immunogenicity of this process may be compromised by oxidative stress and proteolysis. Necroptosis caused by ultrasound-targeted nanobubble destruction (UTND) could boost immunity. Therefore, we tested if necroptosis-inducible nanobubbles (NB) could enhance sonodynamic immunotherapy for BC. We also assessed whether O2-filled NB could address tumor hypoxia and enhance SDT efficacy. METHODS A novel multifunctional nano-system, comprising NB for UTND encapsulating chlorin e6 (Ce6) for SDT and O2 for hypoxia alleviation was established. Ce6-O2NB cytocompatibility and intracellular uptake was studied in vitro, as well as whether Ce6-O2NB could generated reactive oxygen species when exposed to ultrasound irradiation in order to induce apoptosis in tumor cells. In vivo pharmacokinetics, therapeutic efficacy, and immune activation after Ce6-O2NB treatment were studied in 4T1 tumor-bearing mice. RESULTS Ce6-O2NB had a well-designed core-shell structure and desirable biocompatibility and safe therapeutic effects. Ce6-O2NB was able to load both ce6 and oxygen to increase ce6 and oxygen accumulation in tumors. After triggering by ultrasound, Ce6-O2NB generated reactive oxygen species (ROS) and acted as sonosensitizers for SDT, promoting tumor cell death through apoptotic and/or necrotic mechanisms. Furthermore, antitumor immunity was activated by stimulation of spleen lymphocyte proliferation and cytotoxicity, and increasing cytotoxic T lymphocyte numbers. Combination of oxygen with SDT ultimately strengthened its antitumor effects. In addition, Ce6-O2NB alleviated tumor hypoxia, induced increased ROS generation, and improved immune responses and therapeutic efficacy of SDT. Ce6-O2NB also facilitated fluorescence and contrast-enhanced ultrasound imaging. CONCLUSIONS Ce6-O2NB can mitigate tumor hypoxia, enhance SDT, and activate antitumor immunity by inducing simultaneous immunogenic apoptosis and necroptosis, ultimately activating antitumor immunity and inhibiting breast tumor growth in mice.
Collapse
Affiliation(s)
- Shun Tian
- Department of Ultrasound Imaging, The Second People's Hospital of China Three Gorges University, Yichang 443000, China
| | - Yun Liu
- Department of Ultrasound Imaging, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443008, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 334002, China
| | - Yandi Tan
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinwu Cui
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rong Liu
- Department of Ultrasound Imaging, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443008, China
| | - Chaoqi Liu
- Medical College, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 334002, China
| | - Yun Zhao
- Medical College, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 334002, China
| | - Kui Xu
- Department of Ultrasound Imaging, The Second People's Hospital of China Three Gorges University, Yichang 443000, China
| | - Jun Zhou
- Department of Ultrasound Imaging, The Second People's Hospital of China Three Gorges University, Yichang 443000, China.
| |
Collapse
|
7
|
Dewaele L, Fernandes RA. Bispecific T-cell engagers for the recruitment of T cells in solid tumors: a literature review. IMMUNOTHERAPY ADVANCES 2025; 5:ltae005. [PMID: 40083373 PMCID: PMC11904783 DOI: 10.1093/immadv/ltae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 01/25/2025] [Indexed: 03/16/2025] Open
Abstract
In the past decade, T-cell-based immunotherapies have grown to become some of the most promising treatments for cancer. Following the success of immune checkpoint inhibitors, other T-cell-based therapies emerged including CAR-T cells and bispecific T-cell engagers (BiTEs). BiTEs have the unique ability to crosslink T cells and tumor cells independently of major histocompatibility complex (MHC) restriction. They do not rely on TCR specificity or the CD4+/CD8+ costimulatory molecules, overcoming tumor MHC downregulation and low-affinity TCR binding. However, like many other immunotherapies, BiTEs have shown limited success beyond the treatment of hematological malignancies. BiTEs for the treatment of solid tumors still face challenges. Studies in gastrointestinal tumors have revealed Fc toxicity, short half-lives, and immunotoxicity, leading to Fc-silenced half-life extended BiTEs with humanized single-chain variable fragments. Studies in prostate tumors, lung tumors, and malignant gliomas have identified promising targets in PSMA, DLL3, and EGFRvIII, respectively, but also highlighted the problems of on-target off-tumor and BiTE-specific toxicities and inaccessible or immunosuppressive tumor microenvironments. Ongoing research to overcome these limitations remains an interesting field to follow, as BiTEs have the potential to be a powerful tool, especially when used in combination with other immunotherapies.
Collapse
Affiliation(s)
- Laura Dewaele
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ricardo A Fernandes
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
8
|
Liu D, Wang L, Guo Y. Advances in and prospects of immunotherapy for prostate cancer. Cancer Lett 2024; 601:217155. [PMID: 39127338 DOI: 10.1016/j.canlet.2024.217155] [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: 05/30/2024] [Revised: 07/07/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Immunotherapy has shown promising therapeutic effects in hematological malignancies and certain solid tumors and has emerged as a critical and highly potential treatment modality for cancer. However, prostate cancer falls under the category of immune-resistant cold tumors, for which immunotherapy exhibits limited efficacy in patients with solid tumors. Thus, it is important to gain a deeper understanding of the tumor microenvironment in prostate cancer to facilitate immune system activation and overcome immune suppression to advance immunotherapy for prostate cancer. In this review, we discuss the immunosuppressive microenvironment of prostate cancer, which is characterized by the presence of few tumor-infiltrating lymphocytes, abundant immunosuppressive cells, low immunogenicity, and a noninflammatory phenotype, which significantly influences the efficacy of immunotherapy for prostate cancer. Immunotherapy is mainly achieved by activating the host immune system and overcoming immunosuppression. In this regard, we summarize the therapeutic advances in immune checkpoint blockade, immunogenic cell death, reversal of the immunosuppressive tumor microenvironment, tumor vaccines, immune adjuvants, chimeric antigen receptor T-cell therapy, and overcoming penetration barriers in prostate cancer, with the aim of providing novel research insights and approaches to enhance the effectiveness of immunotherapy for prostate cancer.
Collapse
Affiliation(s)
- Deng Liu
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China; Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Luofu Wang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Yanli Guo
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
9
|
Sharma D, Petchiny TN, Czarnota GJ. A Promising Therapeutic Strategy of Combining Acoustically Stimulated Nanobubbles and Existing Cancer Treatments. Cancers (Basel) 2024; 16:3181. [PMID: 39335153 PMCID: PMC11431001 DOI: 10.3390/cancers16183181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
In recent years, ultrasound-stimulated microbubbles (USMBs) have gained great attention because of their wide theranostic applications. However, due to their micro-size, reaching the targeted site remains a challenge. At present, ultrasound-stimulated nanobubbles (USNBs) have attracted particular interest, and their small size allows them to extravasate easily in the blood vessels penetrating deeper into the tumor vasculature. Incorporating USNBs with existing cancer therapies such as chemotherapy, immunotherapy, and/or radiation therapy in several preclinical models has been demonstrated to have a profound effect on solid tumors. In this review, we provide an understanding of the composition and formation of nanobubbles (NBs), followed by the recent progress of the therapeutic combinatory effect of USNBs and other cancer therapies in cancer treatment.
Collapse
Affiliation(s)
- Deepa Sharma
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada;
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Departments of Medical Biophysics, and Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Tera N. Petchiny
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada;
| | - Gregory J. Czarnota
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada;
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Departments of Medical Biophysics, and Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
| |
Collapse
|
10
|
Yang N, Li J, Yu S, Xia G, Li D, Yuan L, Wang Q, Ding L, Fan Z, Li J. Application of Nanomaterial-Based Sonodynamic Therapy in Tumor Therapy. Pharmaceutics 2024; 16:603. [PMID: 38794265 PMCID: PMC11125068 DOI: 10.3390/pharmaceutics16050603] [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: 03/28/2024] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Sonodynamic therapy (SDT) has attracted significant attention in recent years as it is an innovative approach to tumor treatment. It involves the utilization of sound waves or ultrasound (US) to activate acoustic sensitizers, enabling targeted drug release for precise tumor treatment. This review aims to provide a comprehensive overview of SDT, encompassing its underlying principles and therapeutic mechanisms, the applications of nanomaterials, and potential synergies with combination therapies. The review begins by introducing the fundamental principle of SDT and delving into the intricate mechanisms through which it facilitates tumor treatment. A detailed analysis is presented, outlining how SDT effectively destroys tumor cells by modulating drug release mechanisms. Subsequently, this review explores the diverse range of nanomaterials utilized in SDT applications and highlights their specific contributions to enhancing treatment outcomes. Furthermore, the potential to combine SDT with other therapeutic modalities such as photothermal therapy (PTT) and chemotherapy is discussed. These combined approaches aim to synergistically improve therapeutic efficacy while mitigating side effects. In conclusion, SDT emerges as a promising frontier in tumor treatment that offers personalized and effective treatment options with the potential to revolutionize patient care. As research progresses, SDT is poised to play a pivotal role in shaping the future landscape of oncology by providing patients with a broader spectrum of efficacious and tailored treatment options.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhongxiong Fan
- School of Pharmaceutical Sciences, Institute of Materia Medica, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Jinyao Li
- School of Pharmaceutical Sciences, Institute of Materia Medica, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| |
Collapse
|
11
|
Dogheim GM, El Feel NE, Abd El-Maksod EA, Amer SS, El-Gizawy SA, Abd Elhamid AS, Elzoghby AO. Nanomedicines as enhancers of tumor immunogenicity to augment cancer immunotherapy. Drug Discov Today 2024; 29:103905. [PMID: 38295898 DOI: 10.1016/j.drudis.2024.103905] [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: 11/11/2023] [Revised: 01/05/2024] [Accepted: 01/24/2024] [Indexed: 02/07/2024]
Abstract
The potential of cancer immunotherapy is hampered by the poor immunogenicity of cancer cells. Strategies to enhance tumor immunogenicity are imperative to enhance T cell-mediated anti-tumor immunity. Although conventional therapeutics can increase tumor antigen expression or stimulate the release of danger signals to promote immunogenic cell death (ICD), they face challenges relating to efficacy and tumor-specific delivery. Nanomedicines can efficiently deliver tumor antigens, immune adjuvants, epigenetic modulators, or ICD inducers through targeted drug delivery with minimal off-target effects. Collectively, nanomedicines can overcome biological barriers to immunotherapy through targeted antigen delivery, induction of ICD, or epigenetic remodeling, resulting in increased tumor immunogenicity.
Collapse
Affiliation(s)
- Gaidaa M Dogheim
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Nourhan E El Feel
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21568, Egypt
| | - Esraa A Abd El-Maksod
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Sandra Sh Amer
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sanaa A El-Gizawy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Ahmed S Abd Elhamid
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt; Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), 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 02115, USA.
| |
Collapse
|
12
|
Wu Y, Li J, Shu L, Tian Z, Wu S, Wu Z. Ultrasound combined with microbubble mediated immunotherapy for tumor microenvironment. Front Pharmacol 2024; 15:1304502. [PMID: 38487163 PMCID: PMC10937735 DOI: 10.3389/fphar.2024.1304502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/11/2024] [Indexed: 03/17/2024] Open
Abstract
The tumor microenvironment (TME) plays an important role in dynamically regulating the progress of cancer and influencing the therapeutic results. Targeting the tumor microenvironment is a promising cancer treatment method in recent years. The importance of tumor immune microenvironment regulation by ultrasound combined with microbubbles is now widely recognized. Ultrasound and microbubbles work together to induce antigen release of tumor cell through mechanical or thermal effects, promoting antigen presentation and T cells' recognition and killing of tumor cells, and improve tumor immunosuppression microenvironment, which will be a breakthrough in improving traditional treatment problems such as immune checkpoint blocking (ICB) and himeric antigen receptor (CAR)-T cell therapy. In order to improve the therapeutic effect and immune regulation of TME targeted tumor therapy, it is necessary to develop and optimize the application system of microbubble ultrasound for organs or diseases. Therefore, the combination of ultrasound and microbubbles in the field of TME will continue to focus on developing more effective strategies to regulate the immunosuppression mechanisms, so as to activate anti-tumor immunity and/or improve the efficacy of immune-targeted drugs, At present, the potential value of ultrasound combined with microbubbles in TME targeted therapy tumor microenvironment targeted therapy has great potential, which has been confirmed in the experimental research and application of breast cancer, colon cancer, pancreatic cancer and prostate cancer, which provides a new alternative idea for clinical tumor treatment. This article reviews the research progress of ultrasound combined with microbubbles in the treatment of tumors and their application in the tumor microenvironment.
Collapse
Affiliation(s)
| | | | | | | | | | - Zuohui Wu
- Department of Ultrasound, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
13
|
Liang J, Qiao X, Qiu L, Xu H, Xiang H, Ding H, Chen Y. Engineering Versatile Nanomedicines for Ultrasonic Tumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305392. [PMID: 38041509 PMCID: PMC10797440 DOI: 10.1002/advs.202305392] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/15/2023] [Indexed: 12/03/2023]
Abstract
Due to the specific advantages of ultrasound (US) in therapeutic disease treatments, the unique therapeutic US technology has emerged. In addition to featuring a low-invasive targeted cancer-cell killing effect, the therapeutic US technology has been demonstrated to modulate the tumor immune landscape, amplify the therapeutic effect of other antitumor therapies, and induce immunosensitization of tumors to immunotherapy, shedding new light on the cancer treatment. Tremendous advances in nanotechnology are also expected to bring unprecedented benefits to enhancing the antitumor efficiency and immunological effects of therapeutic US, as well as therapeutic US-derived bimodal and multimodal synergistic therapies. This comprehensive review summarizes the immunological effects induced by different therapeutic US technologies, including ultrasound-mediated micro-/nanobubble destruction (UTMD/UTND), sonodynamic therapy (SDT), and focused ultrasound (FUS), as well as the main underlying mechanisms involved. It is also discussed that the recent research progress of engineering intelligent nanoplatform in improving the antitumor efficiency of therapeutic US technologies. Finally, focusing on clinical translation, the key issues and challenges currently faced are summarized, and the prospects for promoting the clinical translation of these emerging nanomaterials and ultrasonic immunotherapy in the future are proposed.
Collapse
Affiliation(s)
- Jing Liang
- Department of UltrasoundHuashan HospitalFudan UniversityShanghai200040China
| | - Xiaohui Qiao
- Department of UltrasoundHuashan HospitalFudan UniversityShanghai200040China
| | - Luping Qiu
- Department of UltrasoundHuashan HospitalFudan UniversityShanghai200040China
| | - Huning Xu
- Department of UltrasoundHuashan HospitalFudan UniversityShanghai200040China
| | - Huijing Xiang
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai2000444China
| | - Hong Ding
- Department of UltrasoundHuashan HospitalFudan UniversityShanghai200040China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai2000444China
| |
Collapse
|
14
|
Pelka S, Guha C. Enhancing Immunogenicity in Metastatic Melanoma: Adjuvant Therapies to Promote the Anti-Tumor Immune Response. Biomedicines 2023; 11:2245. [PMID: 37626741 PMCID: PMC10452223 DOI: 10.3390/biomedicines11082245] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/26/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Advanced melanoma is an aggressive form of skin cancer characterized by low survival rates. Less than 50% of advanced melanoma patients respond to current therapies, and of those patients that do respond, many present with tumor recurrence due to resistance. The immunosuppressive tumor-immune microenvironment (TIME) remains a major obstacle in melanoma therapy. Adjuvant treatment modalities that enhance anti-tumor immune cell function are associated with improved patient response. One potential mechanism to stimulate the anti-tumor immune response is by inducing immunogenic cell death (ICD) in tumors. ICD leads to the release of damage-associated molecular patterns within the TIME, subsequently promoting antigen presentation and anti-tumor immunity. This review summarizes relevant concepts and mechanisms underlying ICD and introduces the potential of non-ablative low-intensity focused ultrasound (LOFU) as an immune-priming therapy that can be combined with ICD-inducing focal ablative therapies to promote an anti-melanoma immune response.
Collapse
Affiliation(s)
- Sandra Pelka
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Chandan Guha
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Urology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Institute of Onco-Physics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
15
|
Maardalen M, Carlisle R, Coussios C. Cavitation-Mediated Immunomodulation and Its Use with Checkpoint Inhibitors. Pharmaceutics 2023; 15:2110. [PMID: 37631324 PMCID: PMC10458634 DOI: 10.3390/pharmaceutics15082110] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
The promotion of anti-tumour immune responses can be an effective route to the complete remission of primary and metastatic tumours in a small proportion of patients. Hence, researchers are currently investigating various methods to further characterise and enhance such responses to achieve a beneficial impact across a wider range of patients. Due to its non-invasive, non-ionising, and targetable nature, the application of ultrasound-mediated cavitation has proven to be a popular method to enhance the delivery and activity of immune checkpoint inhibitors. However, to optimise this approach, it is important to understand the biological and physical mechanisms by which cavitation may promote anti-tumour immune responses. Here, the published literature relating to the role that cavitation may play in modulating anti-tumour immunity is therefore assessed.
Collapse
Affiliation(s)
- Matilde Maardalen
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK
| | | | | |
Collapse
|
16
|
Sharma D, Xuan Leong K, Palhares D, Czarnota GJ. Radiation combined with ultrasound and microbubbles: A potential novel strategy for cancer treatment. Z Med Phys 2023; 33:407-426. [PMID: 37586962 PMCID: PMC10517408 DOI: 10.1016/j.zemedi.2023.04.007] [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: 01/11/2023] [Revised: 03/31/2023] [Accepted: 04/11/2023] [Indexed: 08/18/2023]
Abstract
Cancer is one of the leading causes of death worldwide. Several emerging technologies are helping to battle cancer. Cancer therapies have been effective at killing cancer cells, but a large portion of patients still die to this disease every year. As such, more aggressive treatments of primary cancers are employed and have been shown to be capable of saving a greater number of lives. Recent research advances the field of cancer therapy by employing the use of physical methods to alter tumor biology. It uses microbubbles to enhance radiation effect by damaging tumor vasculature followed by tumor cell death. The technique can specifically target tumor volumes by conforming ultrasound fields capable of microbubbles stimulation and localizing it to avoid vascular damage in surrounding tissues. Thus, this new application of ultrasound-stimulated microbubbles (USMB) can be utilized as a novel approach to cancer therapy by inducing vascular disruption resulting in tumor cell death. Using USMB alongside radiation has showed to augment the anti-vascular effect of radiation, resulting in enhanced tumor response. Recent work with nanobubbles has shown vascular permeation into intracellular space, extending the use of this new treatment method to potentially further improve the therapeutic effect of the ultrasound-based therapy. The significant enhancement of localized tumor cell kill means that radiation-based treatments can be made more potent with lower doses of radiation. This technique can manifest a greater impact on radiation oncology practice by increasing treatment effectiveness significantly while reducing normal tissue toxicity. This review article summarizes the past and recent advances in USMB enhancement of radiation treatments. The review mainly focuses on preclinical findings but also highlights some clinical findings that use USMB as a therapeutic modality in cancer therapy.
Collapse
Affiliation(s)
- Deepa Sharma
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Departments of Radiation Oncology, and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Kai Xuan Leong
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Daniel Palhares
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Departments of Radiation Oncology, and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Gregory J Czarnota
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Departments of Radiation Oncology, and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
17
|
Guo S, Yao Y, Tang Y, Xin Z, Wu D, Ni C, Huang J, Wei Q, Zhang T. Radiation-induced tumor immune microenvironments and potential targets for combination therapy. Signal Transduct Target Ther 2023; 8:205. [PMID: 37208386 DOI: 10.1038/s41392-023-01462-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/21/2023] [Accepted: 04/27/2023] [Indexed: 05/21/2023] Open
Abstract
As one of the four major means of cancer treatment including surgery, radiotherapy (RT), chemotherapy, immunotherapy, RT can be applied to various cancers as both a radical cancer treatment and an adjuvant treatment before or after surgery. Although RT is an important modality for cancer treatment, the consequential changes caused by RT in the tumor microenvironment (TME) have not yet been fully elucidated. RT-induced damage to cancer cells leads to different outcomes, such as survival, senescence, or death. During RT, alterations in signaling pathways result in changes in the local immune microenvironment. However, some immune cells are immunosuppressive or transform into immunosuppressive phenotypes under specific conditions, leading to the development of radioresistance. Patients who are radioresistant respond poorly to RT and may experience cancer progression. Given that the emergence of radioresistance is inevitable, new radiosensitization treatments are urgently needed. In this review, we discuss the changes in irradiated cancer cells and immune cells in the TME under different RT regimens and describe existing and potential molecules that could be targeted to improve the therapeutic effects of RT. Overall, this review highlights the possibilities of synergistic therapy by building on existing research.
Collapse
Affiliation(s)
- Siyu Guo
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Yihan Yao
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Tang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Zengfeng Xin
- Department of Orthopedic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Dang Wu
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Chao Ni
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jian Huang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Qichun Wei
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Ting Zhang
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
18
|
Ultrasound-targeted microbubble destruction remodels tumour microenvironment to improve immunotherapeutic effect. Br J Cancer 2023; 128:715-725. [PMID: 36463323 PMCID: PMC9977958 DOI: 10.1038/s41416-022-02076-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
Cancer immunotherapy (CIT) has gained increasing attention and made promising progress in recent years, especially immune checkpoint inhibitors such as antibodies blocking programmed cell death 1/programmed cell death ligand 1 (PD-1/PD-L1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4). However, its therapeutic efficacy is only 10-30% in solid tumours and treatment sensitivity needs to be improved. The complex tissue environment in which cancers originate is known as the tumour microenvironment (TME) and the complicated and dynamic TME is correlated with the efficacy of immunotherapy. Ultrasound-targeted microbubble destruction (UTMD) is an emerging technology that integrates diagnosis and therapy, which has garnered much traction due to non-invasive, targeted drug delivery and gene transfection characteristics. UTMD has also been studied to remodel TME and improve the efficacy of CIT. In this review, we analyse the effects of UTMD on various components of TME, including CD8+ T cells, tumour-infiltrating myeloid cells, regulatory T cells, natural killer cells and tumour vasculature. Moreover, UTMD enhances the permeability of the blood-brain barrier to facilitate drug delivery, thus improving CIT efficacy in vivo animal experiments. Based on this, we highlight the potential of immunotherapy against various cancer species and the clinical application prospects of UTMD.
Collapse
|
19
|
Wong DCP, Ding JL. The mechanobiology of NK cells- 'Forcing NK to Sense' target cells. Biochim Biophys Acta Rev Cancer 2023; 1878:188860. [PMID: 36791921 DOI: 10.1016/j.bbcan.2023.188860] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 02/16/2023]
Abstract
Natural killer (NK) cells are innate immune lymphocytes that recognize and kill cancer and infected cells, which makes them unique 'off-the-shelf' candidates for a new generation of immunotherapies. Biomechanical forces in homeostasis and pathophysiology accrue additional immune regulation for NK immune responses. Indeed, cellular and tissue biomechanics impact NK receptor clustering, cytoskeleton remodeling, NK transmigration through endothelial cells, nuclear mechanics, and even NK-dendritic cell interaction, offering a plethora of unexplored yet important dynamic regulation for NK immunotherapy. Such events are made more complex by the heterogeneity of human NK cells. A significant question remains on whether and how biochemical and biomechanical cues collaborate for NK cell mechanotransduction, a process whereby mechanical force is sensed, transduced, and translated to downstream mechanical and biochemical signalling. Herein, we review recent advances in understanding how NK cells perceive and mechanotransduce biophysical cues. We focus on how the cellular cytoskeleton crosstalk regulates NK cell function while bearing in mind the heterogeneity of NK cells, the direct and indirect mechanical cues for NK anti-tumor activity, and finally, engineering advances that are of translational relevance to NK cell biology at the systems level.
Collapse
Affiliation(s)
- Darren Chen Pei Wong
- Department of Biological Sciences, National University of Singapore, 117543, Singapore.
| | - Jeak Ling Ding
- Department of Biological Sciences, National University of Singapore, 117543, Singapore; Integrative Sciences and Engineering Programme, National University of Singapore, 119077, Singapore.
| |
Collapse
|
20
|
Wang T, Peng W, Du M, Chen Z. Immunogenic sonodynamic therapy for inducing immunogenic cell death and activating antitumor immunity. Front Oncol 2023; 13:1167105. [PMID: 37168380 PMCID: PMC10166230 DOI: 10.3389/fonc.2023.1167105] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023] Open
Abstract
Immunotherapy is widely regarded as a promising treatment for cancer. However, the immune effector phase suppression of tumor microenvironment (TME) and the generation of immune-related adverse events limit its application. Research indicates that sonodynamic therapy (SDT) can effectively activate antitumor immunity while killing tumor cells. SDT produces cytotoxic substances of tumors, and then cell apoptosis and immunogenic death occur by selectively activating the sonosensitizer under ultrasound. In recent years, various SDT alone as well as SDT in combination with other therapies have been developed to induce immunogenic cell death (ICD) and enhance immunotherapy. This paper overviews the research progress of SDT and nanotechnology in recent years, including the strategies involving SDT alone, SDT-based synergistic induction of antitumor immunity, and immunotherapy based on SDT for multimodal immunotherapy. Finally, the prospects and challenges of these SDT-based therapies in cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Ting Wang
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
| | - Wangrui Peng
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Meng Du
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- *Correspondence: Meng Du, ; Zhiyi Chen,
| | - Zhiyi Chen
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- *Correspondence: Meng Du, ; Zhiyi Chen,
| |
Collapse
|
21
|
Chen KW, Hsu PH, Huang HL, Liu HL, Lin YT, Hsu CY, Lin JH, Lin YH. Targeting nanoparticle-conjugated microbubbles combined with ultrasound-mediated microbubble destruction for enhanced tumor therapy. Pharmacol Res 2022; 186:106532. [DOI: 10.1016/j.phrs.2022.106532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/20/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
|
22
|
Han Y, Sun J, Wei H, Hao J, Liu W, Wang X. Ultrasound-Targeted Microbubble Destruction: Modulation in the Tumor Microenvironment and Application in Tumor Immunotherapy. Front Immunol 2022; 13:937344. [PMID: 35844515 PMCID: PMC9283646 DOI: 10.3389/fimmu.2022.937344] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/27/2022] [Indexed: 12/14/2022] Open
Abstract
Tumor immunotherapy has shown strong therapeutic potential for stimulating or reconstructing the immune system to control and kill tumor cells. It is a promising and effective anti-cancer treatment besides surgery, radiotherapy and chemotherapy. Presently, some immunotherapy methods have been approved for clinical application, and numerous others have demonstrated promising in vitro results and have entered clinical trial stages. Although immunotherapy has exhibited encouraging results in various cancer types, however, a large proportion of patients are limited from these benefits due to specific characteristics of the tumor microenvironment such as hypoxia, tumor vascular malformation and immune escape, and current limitations of immunotherapy such as off-target toxicity, insufficient drug penetration and accumulation and immune cell dysfunction. Ultrasound-target microbubble destruction (UTMD) treatment can help reduce immunotherapy-related adverse events. Using the ultrasonic cavitation effect of microstreaming, microjets and free radicals, UTMD can cause a series of changes in vascular endothelial cells, such as enhancing endothelial cells' permeability, increasing intracellular calcium levels, regulating gene expression, and stimulating nitric oxide synthase activities. These effects have been shown to promote drug penetration, enhance blood perfusion, increase drug delivery and induce tumor cell death. UTMD, in combination with immunotherapy, has been used to treat melanoma, non-small cell lung cancer, bladder cancer, and ovarian cancer. In this review, we summarized the effects of UTMD on tumor angiogenesis and immune microenvironment, and discussed the application and progress of UTMD in tumor immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaolei Wang
- In-Patient Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|