1
|
Zhi S, Zheng J, Yan Y, Wang Y, Wu C, Bi S. Enzymatic interlocking aptamer-micelles for enhanced cellular internalization and nucleus-targeted cancer phototherapy. J Colloid Interface Sci 2025; 678:1132-1141. [PMID: 39341144 DOI: 10.1016/j.jcis.2024.09.182] [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: 06/09/2024] [Revised: 08/28/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024]
Abstract
Multifunctional micelles that permit both diagnosis and treatment present enormous advantage and potential for precision medicine. However, the inherent complexities and structural instability of these systems often cause unsatisfactory targeting and therapeutic performances. Herein, by ingenious design of a 2,5-bis(2-thienyl)pyrrole (SNS) modifier to covalently link with AS1411 aptamer and lipid segment, a simple strategy is proposed for one-step enzymatic preparation of interlocked aptamer-micelle (IApM) under bio-friendly conditions. The interlocked poly(SNS) skeleton in IApM can not only stabilize the micelle structure but also enhance near-infrared (NIR) absorption ability, thus further enhancing cellular internalization and photothermal therapy. In addition, the multivalent AS1411 aptamers tethered in the hydrophilic shell can simultaneously increase the specific binding affinity of DNA micelles and induce nucleus-targeted accumulation for DNA damage-triggered apoptosis. This DNA micelle achieves "best of both worlds" with enhanced biostability for cellular internalization and improved NIR photothermal conversion efficiency for nucleus-targeted therapy, which provides a promising formulation strategy for precision cancer treatment.
Collapse
Affiliation(s)
- Shuangcheng Zhi
- College of Chemistry and Chemical Engineering, Key Laboratory of Shandong Provincial Universities for Functional Molecules and Materials, Qingdao University, Qingdao 266071, China
| | - Jiao Zheng
- College of Chemistry and Chemical Engineering, Key Laboratory of Shandong Provincial Universities for Functional Molecules and Materials, Qingdao University, Qingdao 266071, China
| | - Yongcun Yan
- College of Chemistry and Chemical Engineering, Key Laboratory of Shandong Provincial Universities for Functional Molecules and Materials, Qingdao University, Qingdao 266071, China
| | - Yanfang Wang
- College of Chemistry and Chemical Engineering, Key Laboratory of Shandong Provincial Universities for Functional Molecules and Materials, Qingdao University, Qingdao 266071, China
| | - Chuancheng Wu
- College of Chemistry and Chemical Engineering, Key Laboratory of Shandong Provincial Universities for Functional Molecules and Materials, Qingdao University, Qingdao 266071, China
| | - Sai Bi
- College of Chemistry and Chemical Engineering, Key Laboratory of Shandong Provincial Universities for Functional Molecules and Materials, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
2
|
Pinho S, Coelho JMP, Gaspar MM, Reis CP. Advances in localized prostate cancer: A special focus on photothermal therapy. Eur J Pharmacol 2024; 983:176982. [PMID: 39260812 DOI: 10.1016/j.ejphar.2024.176982] [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: 04/18/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024]
Abstract
Prostate cancer (PCa) is a high prevalence disease, per 10000 habitants, that tends to increase with age. This pathology is difficult to detect at an early stage due to the absence of symptoms, hence the importance of monitoring signs for early detection. This disease can be detected by various methods, including plasmatic levels of prostate-specific antigen (PSA) and rectal touch, with biopsy being necessary to confirm the diagnosis. Patients affected by prostate cancer can have localized or advanced disease. There are conventional approaches that have been used as a reference in localized cancer, such as active surveillance, surgery, or radiotherapy. However, the adverse effects might vary and, sometimes, they can be permanent. An overview about the innovative therapeutic approaches to improve outcomes in terms of both tumor remission and side effects for localized PCa is presented. In case of emerging light-based treatment strategies, they aimed at ablating tumor tissue by inducing an external light are non-invasive, localized and, considerably, they are able to reduce lesions in peripheral tissues. One is photodynamic therapy (PDT) and it involves the photooxidation of molecules culminating in the formation of reactive oxygen species (ROS), inducing cell death. On the other hand, photothermal therapy (PTT) is based on inducing hyperthermia in cancer cells by irradiating them with beams of light at a specific wavelength. To improve the heat generated, gold nanoparticles (AuNPs) have those desirable characteristics that have drawn attention to PTT. Various studies point to AuNPs as efficient nanomaterials in PTT for the treatment of tumors, including prostate cancer. This review includes the most representative advances in this research field, dated from 1998 to 2023. It is noticed that several advances have been made and the way to find the effective treatment without impacting adverse side effects is shorter.
Collapse
Affiliation(s)
- Sara Pinho
- Research Institute for Medicines, IMed.ULisboa - Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - João M P Coelho
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines, IMed.ULisboa - Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal; Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - Catarina Pinto Reis
- Research Institute for Medicines, IMed.ULisboa - Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal; Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal.
| |
Collapse
|
3
|
Li Z, Lan J, Liu L, Wang Y, Chen L, Zeng R, Gu D, Hu R, Zhang T, Ding Y. Versatile Thermo-Sensitive liposomes with HSP inhibition and Anti-Inflammation for synergistic Chemo-Photothermal to inhibit breast cancer metastasis. Int J Pharm 2024; 664:124583. [PMID: 39153642 DOI: 10.1016/j.ijpharm.2024.124583] [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: 04/26/2024] [Revised: 07/13/2024] [Accepted: 08/10/2024] [Indexed: 08/19/2024]
Abstract
Photothermal therapy (PTT) is a prospective therapeutic method for breast cancer. However, excess inflammatory response induced by PTT may aggravate tumor metastasis. Meanwhile, the overexpressed heat shock proteins (HSPs) by cancer cells can protect them from hyperthermia during PTT. Therefore, to attenuate the PTT-induced inflammation and inhibit tumor metastasis, a folate receptor-targeted thermo-sensitive liposome (BI-FA-LP) co-loading Berberine (BBR) and Indocyanine green (ICG) was developed. BI-FA-LP utilized enhanced permeability and retention (EPR) effect and FA receptor-mediated endocytosis to selectively accumulate at tumor, reducing off-target toxicity during the treatment. After targeting to the tumor site, BBR and ICG were released from BI-FA-LP upon laser irradiation, and ICG showed good photothermal performance, while BBR inhibited HSP70 and HSP90 expression during PTT, exerting chemo-photothermal synergetic anti-tumor effect. Moreover, BBR could suppress the PTT induced inflammation, thus inhibiting tumor metastasis and ameliorating tissue injury. Thus, this versatile liposome provided a new strategy to enhance PTT and anti-inflammatory effects for breast cancer treatment.
Collapse
Affiliation(s)
- Zhe Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jinshuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yu Wang
- Shanghai Children's Medical Center, Shanghai 200127, China
| | - Lixia Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ruifeng Zeng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Donghao Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ruolu Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; National Innovation Platform for medical industry-education integration, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
4
|
Lu H, Li Z, Duan Z, Liao Y, Liu K, Zhang Y, Fan L, Xu T, Yang D, Wang S, Fu Y, Xiang H, Chen Y, Li G. Photothermal Catalytic Reduction and Bone Tissue Engineering Towards a Three-in-One Therapy Strategy for Osteosarcoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408016. [PMID: 39165073 DOI: 10.1002/adma.202408016] [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: 06/05/2024] [Revised: 07/15/2024] [Indexed: 08/22/2024]
Abstract
Osteosarcoma is one of the most dreadful bone neoplasms in young people, necessitating the development of innovative therapies that can effectively eliminate tumors while minimizing damage to limb function. An ideal therapeutic strategy should possess three essential capabilities: antitumor effects, tissue-protective properties, and the ability to enhance osteogenesis. In this study, self-assembled Ce-substituted molybdenum blue (CMB) nanowheel crystals are synthesized and loaded onto 3D-printed bioactive glass (CMB@BG) scaffolds to develop a unique three-in-one treatment approach for osteosarcoma. The CMB@BG scaffolds exhibit outstanding photothermally derived tumor ablation within the near-infrared-II window due to the surface plasmon resonance properties of the CMB nanowheel crystals. Furthermore, the photothermally synergistic catalytic effect of CMB promotes the rapid scavenging of reactive oxygen species caused by excessive heat, thereby suppressing inflammation and protecting surrounding tissues. The CMB@BG scaffolds possess pro-proliferation and pro-differentiation capabilities that efficiently accelerate bone regeneration within bone defects. Altogether, the CMB@BG scaffolds that combine highly efficient tumor ablation, tissue protection based on anti-inflammatory mechanisms, and enhanced osteogenic ability are likely to be a point-to-point solution for the comprehensive therapeutic needs of osteosarcoma.
Collapse
Affiliation(s)
- Hengli Lu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Zihua Li
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Zhengwei Duan
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Yuxin Liao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Kaiyuan Liu
- Department of Bone Tumor Surgery, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200025, P. R. China
| | - Yiwei Zhang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Lin Fan
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Tianyang Xu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Dong Yang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Sen Wang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Yuesong Fu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Huijing Xiang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang, 325088, P. R. China
- Shanghai Institute of Materdicine, Shanghai, 200051, P. R. China
| | - Guodong Li
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| |
Collapse
|
5
|
Leng G, Duan B, Liu J, Li S, Zhao W, Wang S, Hou G, Qu J. The advancements and prospective developments in anti-tumor targeted therapy. Neoplasia 2024; 56:101024. [PMID: 39047659 PMCID: PMC11318541 DOI: 10.1016/j.neo.2024.101024] [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: 02/02/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
Cancer poses a major threat to human health worldwide. The development of anti-tumor materials provides new modalities for cancer diagnosis and treatment. In this review, we comprehensively summarize the research progress and clinical applications of anti-tumor materials. First, we introduce the etiology and pathogenesis of cancer, and the significance and challenges of anti-tumor materials research. Then, we classify anti-tumor materials and discuss their mechanisms of action. After that, we elaborate the research advances and clinical applications of anti-tumor materials, including those targeting tumor cells and therapeutic instruments. Finally, we discuss the future perspectives and challenges in the field of anti-tumor materials. This review aims to provide an overview of the current status of anti-tumor materials research and application, and to offer insights into future directions in this rapidly evolving field, which holds promise for more precise, efficient and customized treatment of cancer.
Collapse
Affiliation(s)
- Guorui Leng
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai 264003, China
| | - Baorong Duan
- Research Center for Leather and Protein of College of Chemistry & Chemical Engineering, Yantai University, Yantai 264005, China
| | - Junjie Liu
- Department of Physics, Binzhou Medical University, Yantai 264003, China
| | - Song Li
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai 264003, China
| | - Wenwen Zhao
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai 264003, China
| | - Shanshan Wang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Guige Hou
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China.
| | - Jiale Qu
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China; Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS); Shandong Provincial Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, China.
| |
Collapse
|
6
|
Prakash A, Yadav S, Saxena PS, Srivastava A. Development of folate-conjugated polypyrrole nanoparticles incorporated with nitrogen-doped carbon quantum dots for targeted bioimaging and photothermal therapy. Talanta 2024; 278:126528. [PMID: 38996560 DOI: 10.1016/j.talanta.2024.126528] [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: 03/09/2024] [Revised: 06/18/2024] [Accepted: 07/07/2024] [Indexed: 07/14/2024]
Abstract
PPy nanoparticles are widely employed as PTT agents, because of their exceptional near-infrared absorption properties. Nonetheless, the efficacy of PTT with PPy nanoparticles is hindered by a challenge, specifically, a lack of precise targeting. In this study, a PTT imaging agent was developed by combining NCQDs having bright green fluorescent properties with PPy nanoparticles along with the masking of folic acid to overcome the challenge of targeting. The synthesized PPy:NCQDs:FA nanocomposite, characterized by extraordinary photothermal property, was utilized for imaging of folate receptor positive (FA+) MCF-7 cancer cells through the emission of green fluorescence by NCQDs incorporated within the nanocomposite. Additionally, these nanoparticles demonstrated a good level of cell viability, exceeding 82 %, even at a concentration of 600 μg mL-1. Even the in vivo toxicity inspection of the nanocomposite exemplified no observed acute toxicity at experimental dosages of 1 and 3 mg per kg body weight. By subjecting MCF-7 cells, inoculated with 100 μg mL-1 of nanocomposite, to NIR laser irradiation for 5 min, a significant decline in cell viability was witnessed, establishing the photothermal therapeutic potency of the nanocomposite. The death of cancer cells induced by nanocomposite was verified through MTT assay, imaging of cells by NCQDs alone, with nanocomposite, and by live/dead cell Calcein AM/PI staining assay. Quantification of induced apoptosis post-laser treatment is conducted through staining with Annexin V-FITC/PI. These findings establish potential use of PPy:NCQDs:FA nanocomposite as versatile theranostic agents, capable of targeted bioimaging and treatment for cancer cells exhibiting folate receptors.
Collapse
Affiliation(s)
- Aakriti Prakash
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Sujit Yadav
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Preeti S Saxena
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| | - Anchal Srivastava
- Department of Physics, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| |
Collapse
|
7
|
Fan J, Qin Y, Qiu W, Liang J, Xiao C, Xie Q, Tong C, Yuan L, Long Y, Liu B. Gamabufotalin loaded micro-nanocomposites for multimodal therapy of metastatic TNBC by efficiently inducing ICD. Biomaterials 2024; 314:122851. [PMID: 39366186 DOI: 10.1016/j.biomaterials.2024.122851] [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: 05/09/2024] [Revised: 08/30/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Gamabufotalin (CS-6), a main active compound derived from Chinese medicine Chansu, exhibits a robust inhibitory effect on programmed death-ligand 1 (PD-L1) in triple-negative breast cancer (TNBC) cells. Despite its potential for tumor therapy, the medical application of CS-6 is constrained by its hydrophobic nature, lack of targeting capability, and weak immunogenic cell death (ICD) effect. To address these limitations and improve the therapeutic efficiency of this drug against metastatic TNBC, we designed a new kind of CS-6@CPB-S.lux that integrates carboxy-Prussian blue nanoparticles (CPB NPs), CS-6, and attenuated Salmonella typhimurium (S.lux) for TNBC therapy. In vitro and in vivo results have confirmed that CS-6@CPB NPs were efficiently delivered to neoplastic tissue by the tumor hypoxic chemotaxis property of S.lux, wherein the nanomedicine induced significant tumor cell necroptosis and apoptosis via photothermal therapy (PTT) of CPB NPs and chemotherapy of CS-6, which elicited ICD and inhibited PD-L1 expression, resulting in dendritic cells (DCs) maturation and effector T cells activation to comprehensively eliminate tumors. Additionally, the CS-6@CPB-S.lux + Laser treatment significantly transformed the immunosuppressive tumor microenvironment (TME), enhancing antitumor immunity through promoting the polarization of tumor-associated macrophages into antitumorigenic M1 and reducing Tregs recruitment. Consequently, this comprehensive therapy not only inhibited primary and abscopal tumor progression but also prevented TNBC metastasis, which significantly prolonged survival time in animal models. In summary, these findings indicated an alternative approach for metastatic TNBC therapy.
Collapse
Affiliation(s)
- Jialong Fan
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Yan Qin
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China; TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Wensheng Qiu
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Jiahao Liang
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Chang Xiao
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Qian Xie
- Department of Pharmacy, Maternal and Child Health of Hunan Province, Changsha, 410008, China
| | - Chunyi Tong
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Liqin Yuan
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Ying Long
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China.
| | - Bin Liu
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China; NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
8
|
Gomes SM, Gaspar MM, Coelho JMP, Reis CP. Targeting superficial cancers with gold nanoparticles: a review of current research. Ther Deliv 2024; 15:781-799. [PMID: 39314189 PMCID: PMC11457633 DOI: 10.1080/20415990.2024.2395249] [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: 04/04/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
Superficial cancers typically refer to cancers confined to the surface layers of tissue. Low-targeting therapies or side effects prompt exploration of novel therapeutic approaches. Gold nanoparticles (AuNPs), due to their unique optical properties, serve as effective photosensitizers, enabling tumor ablation through photothermal therapy (PTT). PTT induced by AuNPs can be achieved through light sources externally applied to the skin. Near-infrared radiation is the main light candidate due to its deep tissue penetration capability. This review explores recent advancements in AuNP-based PTT for superficial cancers, specifically breast, head and neck, thyroid, bladder and prostate cancers. Additionally, challenges and future directions in utilizing AuNPs for cancer treatment are discussed, emphasizing the importance of balancing efficacy with safety in clinical applications.
Collapse
Affiliation(s)
- Susana M Gomes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - João MP Coelho
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - Catarina Pinto Reis
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| |
Collapse
|
9
|
Pedrosa TDL, de Oliveira GMF, Pereira ACMV, Crispim MJBDS, da Silva LA, da Silva MS, de Souza IA, Melo AMMDA, Gomes ASL, de Araujo RE. Tailoring Plasmonic Nanoheaters Size for Enhanced Theranostic Agent Performance. Bioengineering (Basel) 2024; 11:934. [PMID: 39329676 PMCID: PMC11428745 DOI: 10.3390/bioengineering11090934] [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: 08/01/2024] [Revised: 09/04/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
The introduction of optimized nanoheaters, which function as theranostic agents integrating both diagnostic and therapeutic processes, holds significant promise in the medical field. Therefore, developing strategies for selecting and utilizing optimized plasmonic nanoheaters is crucial for the effective use of nanostructured biomedical agents. This work elucidates the use of the Joule number (Jo) as a figure of merit to identify high-performance plasmonic theranostic agents. A framework for optimizing metallic nanoparticles for heat generation was established, uncovering the size dependence of plasmonic nanoparticles optical heating. Gold nanospheres (AuNSs) with a diameter of 50 nm and gold nanorods (AuNRs) with dimensions of 41×10 nm were identified as effective nanoheaters for visible (530 nm) and infrared (808 nm) excitation. Notably, AuNRs achieve higher Jo values than AuNSs, even when accounting for the possible orientations of the nanorods. Theoretical results estimate that 41×10 nm gold nanorods have an average Joule number of 80, which is significantly higher compared to larger rods. The photothermal performance of optimal and suboptimal nanostructures was evaluated using photoacoustic imaging and photothermal therapy procedures. The photoacoustic images indicate that, despite having larger absorption cross-sections, the large nanoparticle volume of bigger particles leads to less efficient conversion of light into heat, which suggests that the use of optimized nanoparticles promotes higher contrast, benefiting photoacoustic-based procedures in diagnostic applications. The photothermal therapy procedure was performed on S180-bearing mice inoculated with 41×10 nm and 90×25 nm PEGylated AuNRs. Five minutes of laser irradiation of tumor tissue with 41×10 nm produced an approximately 9.5% greater temperature rise than using 90×25 AuNRs in the therapy trials. Optimizing metallic nanoparticles for heat generation may reduce the concentration of the nanoheaters used or decrease the light fluence for bioscience applications, paving the way for the development of more economical theranostic agents.
Collapse
Affiliation(s)
- Túlio de L Pedrosa
- Laboratory of Biomedical Optics and Imaging, Federal University of Pernambuco, Recife 50740-540, Brazil
| | - Gabrielli M F de Oliveira
- Laboratory of Biomedical Optics and Imaging, Federal University of Pernambuco, Recife 50740-540, Brazil
| | - Arthur C M V Pereira
- Department of Physics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Mariana J B da S Crispim
- Laboratory of Biomedical Optics and Imaging, Federal University of Pernambuco, Recife 50740-540, Brazil
| | - Luzia A da Silva
- Graduate Program in Biological Sciences, Federal University of Pernambuco, Recife 50670-420, Brazil
| | - Marcilene S da Silva
- Laboratory of Pharmacology and Experimental Cancerology, Federal University of Pernambuco, Recife 50740-521, Brazil
| | - Ivone A de Souza
- Laboratory of Pharmacology and Experimental Cancerology, Federal University of Pernambuco, Recife 50740-521, Brazil
| | - Ana M M de A Melo
- Department of Biophysics and Radiobiology, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Anderson S L Gomes
- Department of Physics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Renato E de Araujo
- Laboratory of Biomedical Optics and Imaging, Federal University of Pernambuco, Recife 50740-540, Brazil
| |
Collapse
|
10
|
Li L, Lin Y, Liu K, Huang R, Wen W, Huang Y, Liu M, Zhou C, Ding S, Luo B. Multiple-Effect Combined Hydrogels: "Temporal Regulation" Treatment of Osteosarcoma-Associated Bone Defects with Switchable Hyperthermia and Bioactive Agents. Adv Healthc Mater 2024:e2402505. [PMID: 39233538 DOI: 10.1002/adhm.202402505] [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: 07/08/2024] [Revised: 08/25/2024] [Indexed: 09/06/2024]
Abstract
Achieving the clinically staged treatment of osteosarcoma-associated bone defects encounters the multiple challenges of promptly removing postoperative residual tumor cells and bacterial infection, followed by bone reconstruction. Herein, a core/shell hydrogel with multiple-effect combination is designed to first exert antitumor and antibacterial activities and then promote osteogenesis. Specifically, doxorubicin (DOX) is loaded by magnesium-iron-based layered double hydroxide (LDH) to prepare LDOX, which is introduced into a thermo-sensitive hydrogel to serve as an outer shell of the core/shell hydrogel, meanwhile, LDH-contained liquid crystal hydrogel, abbreviated as LCgel-L, is served as an inner core. At the early stage of treatment, the dissociation of the outer shell triggered by moderate hyperthermia led to the thermo-sensitive release of LDOX, which can be targeted for the release of DOX within tumor cells, thereby promptly removing postoperative residual tumor cells based on the synergistic effect of photothermal therapy (PTT) and DOX, and postoperative bacterial infection can also be effectively prevented by PTT simultaneously. More importantly, the dissociation of the outer shell prompted the full exposure of the inner core, which will exert osteogenic activity based on the synergy of liquid crystal hydrogel as well as LDH-induced mild hyperthermia and ion effects, thereby enabling "temporal regulation" treatment of osteosarcoma-associated bone defects. This study provides a valuable insight for the development of osteosarcoma-associated bone repair materials.
Collapse
Affiliation(s)
- Lin Li
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
| | - Yating Lin
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
| | - Kun Liu
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
| | - Runshan Huang
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
| | - Wei Wen
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, P. R. China
| | - Yadong Huang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Guangzhou, 510632, P. R. China
| | - Mingxian Liu
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, P. R. China
| | - Changren Zhou
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, P. R. China
| | - Shan Ding
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, P. R. China
| | - Binghong Luo
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, P. R. China
| |
Collapse
|
11
|
Zhu YQ, Chen Z, Chen ZY, Zhou ZW, Bai Q, Wu MX, Wang XH. Discrete Macrocyclic Polymer Hosts-Induced Cascade Luminescence Enhancement and Application in Bioimaging. Chemistry 2024:e202402808. [PMID: 39207820 DOI: 10.1002/chem.202402808] [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: 07/25/2024] [Revised: 08/24/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
The integration of polymers, supramolecular macrocycles and aggregation-induced emission (AIE) molecules provides numerous possibilities for constructing various functional supramolecular systems. Herein, we constructed supramolecular assembled systems based on discrete macrocyclic polymer hosts via the cooperation of hydra-headed macrocycles containing two or three pillar[5]arene units (defined as P2, P3), the block polymer F127 and AIE molecules (alkyl-cyano modified tetraphenylethene, alkyl-triazole-cyano modified 9,10-distyrylanthracene, defined as TPE-(CN)4 and DSA-(TACN)2). Compared with the binary assembly between hydra-headed hosts or F127 and AIE molecules, cascaded supramolecular assembly-induced emission enhancement (SAIEE) in aqueous solution was achieved in discrete macrocyclic polymer-based supramolecular assembled systems. Considering the cascaded SAIEE performance, we have successfully applied discrete macrocyclic polymer-based supramolecular assembled systems to bioimaging and constructed an artificial light-harvesting system (LHs) to explore more potential applications. The supramolecular assembly form of discrete macrocyclic polymers hosts and AIE molecules proposed in this work provides new inspiration for the construction and application of high-performance supramolecular luminescent systems.
Collapse
Affiliation(s)
- Yu-Qi Zhu
- Institute for Sustainable Energy and Resources, College of Chemistry and Chemical Engineering, Qingdao University, 308 Ningxia Road, Qingdao, 266071, P. R. China
| | - Zhaojun Chen
- Institute for Sustainable Energy and Resources, College of Chemistry and Chemical Engineering, Qingdao University, 308 Ningxia Road, Qingdao, 266071, P. R. China
| | - Zhong-Yuan Chen
- Institute for Sustainable Energy and Resources, College of Chemistry and Chemical Engineering, Qingdao University, 308 Ningxia Road, Qingdao, 266071, P. R. China
| | - Zhi-Wei Zhou
- Institute for Sustainable Energy and Resources, College of Chemistry and Chemical Engineering, Qingdao University, 308 Ningxia Road, Qingdao, 266071, P. R. China
| | - Qian Bai
- Center for Medical Experiment, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, P. R. China
| | - Ming-Xue Wu
- Institute for Sustainable Energy and Resources, College of Chemistry and Chemical Engineering, Qingdao University, 308 Ningxia Road, Qingdao, 266071, P. R. China
| | - Xing-Huo Wang
- Institute for Sustainable Energy and Resources, College of Chemistry and Chemical Engineering, Qingdao University, 308 Ningxia Road, Qingdao, 266071, P. R. China
| |
Collapse
|
12
|
Farivar N, Khazamipour N, Roberts ME, Nelepcu I, Marzban M, Moeen A, Oo HZ, Nakouzi NA, Dolleris C, Black PC, Daugaard M. Pulsed Photothermal Therapy of Solid Tumors as a Precondition for Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309495. [PMID: 38511548 DOI: 10.1002/smll.202309495] [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: 10/19/2023] [Revised: 03/07/2024] [Indexed: 03/22/2024]
Abstract
Photothermal therapy (PTT) refers to the use of plasmonic nanoparticles to convert electromagnetic radiation in the near infrared region to heat and kill tumor cells. Continuous wave lasers have been used clinically to induce PTT, but the treatment is associated with heat-induced tissue damage that limits usability. Here, the engineering and validation of a novel long-pulsed laser device able to induce selective and localized mild hyperthermia in tumors while reducing the heat affected zone and unwanted damage to surrounding tissue are reported. Long-pulsed PTT induces acute necrotic cell death in heat affected areas and the release of tumor associated antigens. This antigen release triggers maturation and stimulation of CD80/CD86 in dendritic cells in vivo that primes a cytotoxic T cell response. Accordingly, long-pulsed PTT enhances the therapeutic effects of immune checkpoint inhibition and increases survival of mice with bladder cancer. Combined, the data promote long-pulsed PTT as a safe and effective strategy for enhancing therapeutic responses to immune checkpoint inhibitors while minimizing unwanted tissue damage.
Collapse
Affiliation(s)
- Negin Farivar
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Nastaran Khazamipour
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Morgan E Roberts
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Irina Nelepcu
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Mona Marzban
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Alireza Moeen
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Htoo Zarni Oo
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Nader Al Nakouzi
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Casper Dolleris
- Dolleris Scientific Corp., 2327 Collingwood Street, Vancouver, BC, V6R 3L2, Canada
| | - Peter C Black
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Mads Daugaard
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| |
Collapse
|
13
|
Wang S, McCoy CP, Li P, Li Y, Zhao Y, Andrews GP, Wylie MP, Ge Y. Carbon Dots in Photodynamic/Photothermal Antimicrobial Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1250. [PMID: 39120355 PMCID: PMC11314369 DOI: 10.3390/nano14151250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024]
Abstract
Antimicrobial resistance (AMR) presents an escalating global challenge as conventional antibiotic treatments become less effective. In response, photodynamic therapy (PDT) and photothermal therapy (PTT) have emerged as promising alternatives. While rooted in ancient practices, these methods have evolved with modern innovations, particularly through the integration of lasers, refining their efficacy. PDT harnesses photosensitizers to generate reactive oxygen species (ROS), which are detrimental to microbial cells, whereas PTT relies on heat to induce cellular damage. The key to their effectiveness lies in the utilization of photosensitizers, especially when integrated into nano- or micron-scale supports, which amplify ROS production and enhance antimicrobial activity. Over the last decade, carbon dots (CDs) have emerged as a highly promising nanomaterial, attracting increasing attention owing to their distinctive properties and versatile applications, including PDT and PTT. They can not only function as photosensitizers, but also synergistically combine with other photosensitizers to enhance overall efficacy. This review explores the recent advancements in CDs, underscoring their significance and potential in reshaping advanced antimicrobial therapeutics.
Collapse
Affiliation(s)
| | - Colin P. McCoy
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (S.W.)
| | | | | | | | | | | | - Yi Ge
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (S.W.)
| |
Collapse
|
14
|
Wang L, Song K, Jiang C, Liu S, Huang S, Yang H, Li X, Zhao F. Metal-Coordinated Polydopamine Structures for Tumor Imaging and Therapy. Adv Healthc Mater 2024:e2401451. [PMID: 39021319 DOI: 10.1002/adhm.202401451] [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: 04/20/2024] [Revised: 05/27/2024] [Indexed: 07/20/2024]
Abstract
Meticulously engineered nanomaterials achieve significant advances in the diagnosis and therapy of solid tumors by improving tumor delivery efficiency; and thereby, enhancing imaging and therapeutic efficacy. Currently, polydopamine (PDA) attracts widespread attention because of its biocompatibility, simplicity of preparation, abundant surface groups, and high photothermal conversion efficiency, which can be applied in drug delivery, photothermal therapy, theranostics, and other nanomedicine fields. Inspired by PDA structures that are rich in catechol and amino functional groups that can coordinate with various metal ions, which have charming qualities and characteristics, metal-coordinated PDA structures are exploited for tumor theranostics, but are not thoroughly summarized. Herein, this review summarizes the recent progress in the fabrication of metal-coordinated PDA structures and their availabilities in tumor imaging and therapy, with further in-depth discussion of the challenges and future perspectives of metal-coordinated PDA structures, with the aim that this systematic review can promote interdisciplinary intersections and provide inspiration for the further growth and clinical translation of PDA materials.
Collapse
Affiliation(s)
- Lihua Wang
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Kaiyue Song
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Cong Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Shanping Liu
- Library of Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Shaorong Huang
- Institute of Geriatrics, Jiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
| | - Huang Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310003, China
| | - Xianglong Li
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Feng Zhao
- Jiangxi Province Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| |
Collapse
|
15
|
Wu W, Wang Y, Yang H, Chen H, Wang C, Liang J, Song Y, Xu S, Sun Y, Wang L. Antibacterial and Biofilm Removal Strategies Based on Micro/Nanomotors in the Biomedical Field. ChemMedChem 2024:e202400349. [PMID: 38965060 DOI: 10.1002/cmdc.202400349] [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: 05/08/2024] [Revised: 06/30/2024] [Accepted: 07/04/2024] [Indexed: 07/06/2024]
Abstract
Bacterial infection, which can trigger varieties of diseases and tens of thousands of deaths each year, poses serious threats to human health. Particularly, the new dilemma caused by biofilms is gradually becoming a severe and tough problem in the biomedical field. Thus, the strategies to address these problems are considered an urgent task at present. Micro/nanomotors (MNMs), also named micro/nanoscale robots, are mostly driven by chemical energy or external field, exhibiting strong diffusion and self-propulsion in the liquid media, which has the potential for antibacterial applications. In particular, when MNMs are assembled in swarms, they become robust and efficient for biofilm removal. However, there is a lack of comprehensive review discussing the progress in this aspect. Bearing it in mind and based on our own research experience in this regard, the studies on MNMs driven by different mechanisms orchestrated for antibacterial activity and biofilm removal are timely and concisely summarized and discussed in this work, aiming to show the advantages of MNMs brought to this field. In addition, an outlook was proposed, hoping to provide the fundamental guidance for future development in this area.
Collapse
Affiliation(s)
- Wenlu Wu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Yuxin Wang
- Emergency Department, Harbin First Hospital, Harbin, 150010, China
| | - Haiyue Yang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Haixu Chen
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Cong Wang
- Department of Microwave Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Junge Liang
- Department of Electronic Engineering, Jiangnan University, Wuxi, 214122, China
| | - Yiran Song
- Department of Electronic Engineering, Jiangnan University, Wuxi, 214122, China
| | - Shanshan Xu
- Emergency Department, Harbin First Hospital, Harbin, 150010, China
| | - Yuan Sun
- Center of Pharmaceutical Engineering and Technology, Harbin University of Commerce, Harbin, 150076, China
| | - Lei Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| |
Collapse
|
16
|
Kim G, Luo Y, Shin M, Bouffard J, Bae J, Kim Y. Making the Brightest Ones Dim: Maximizing the Photothermal Conversion Efficiency of BODIPY-Based Photothermal Agents. Adv Healthc Mater 2024; 13:e2400885. [PMID: 38573765 DOI: 10.1002/adhm.202400885] [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: 03/10/2024] [Revised: 04/01/2024] [Indexed: 04/06/2024]
Abstract
The successful implementation of photothermal therapy (PTT) in cancer treatment hinges on the development of highly effective photothermal agents (PTAs). Boron dipyrromethene (BODIPY) dyes, being well known for their high brightness and quantum efficiencies, are the antithesis of PTAs. Nonetheless, a systematic exploration of the photophysics and photothermal characteristics of a series of π-extended BODIPY dyes with high absorptivity in the near-infrared (NIR) region has achieved superior photothermal conversion efficiencies (>90%), in both monomeric state and nanoparticles after encapsulation in a biocompatible polyethyleneglycol 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy-(polyethylene glycol)-2000]. Optimal PTA candidates combine strong NIR absorption provided by extended donor-acceptor conjugation and an optimization of the electronic and steric effects of meso-substituents to maximize photothermal conversion performance. The PTT-optimized meso-CF3-BODIPY, TCF3PEn exhibits exceptional efficacy in inducing cancer cell apoptosis and in vivo tumor ablation using low-power NIR laser irradiation (0.3 W cm-2, 808 nm) as well as excellent biological safety, underscoring its potential for advancing light-induced cancer therapies.
Collapse
Affiliation(s)
- Gibeom Kim
- Department of Chemistry and Research Institute of Basic Sciences, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, South Korea
| | - Yongyang Luo
- Department of Life Science, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Myunghwan Shin
- Department of Chemistry and Research Institute of Basic Sciences, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, South Korea
| | - Jean Bouffard
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, South Korea
| | - Jeehyeon Bae
- School of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Youngmi Kim
- Department of Chemistry and Research Institute of Basic Sciences, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, South Korea
| |
Collapse
|
17
|
Ramezani P, De Smedt SC, Sauvage F. Supramolecular dye nanoassemblies for advanced diagnostics and therapies. Bioeng Transl Med 2024; 9:e10652. [PMID: 39036081 PMCID: PMC11256156 DOI: 10.1002/btm2.10652] [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: 09/12/2023] [Revised: 01/09/2024] [Accepted: 01/19/2024] [Indexed: 07/23/2024] Open
Abstract
Dyes have conventionally been used in medicine for staining cells, tissues, and organelles. Since these compounds are also known as photosensitizers (PSs) which exhibit photoresponsivity upon photon illumination, there is a high desire towards formulating these molecules into nanoparticles (NPs) to achieve improved delivery efficiency and enhanced stability for novel imaging and therapeutic applications. Furthermore, it has been shown that some of the photophysical properties of these molecules can be altered upon NP formation thereby playing a major role in the outcome of their application. In this review, we primarily focus on introducing dye categories, their formulation strategies and how these strategies affect their photophysical properties in the context of photothermal and non-photothermal applications. More specifically, the most recent progress showing the potential of dye supramolecular assemblies in modalities such as photoacoustic and fluorescence imaging, photothermal and photodynamic therapies as well as their employment in photoablation as a novel modality will be outlined. Aside from their photophysical activity, we delve shortly into the emerging application of dyes as drug stabilizing agents where these molecules are used together with aggregator molecules to form stable nanoparticles.
Collapse
Affiliation(s)
- Pouria Ramezani
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences Ghent University Ghent Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences Ghent University Ghent Belgium
| | - Félix Sauvage
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences Ghent University Ghent Belgium
| |
Collapse
|
18
|
Wang H, Bo W, Feng X, Zhang J, Li G, Chen Y. Strategies and Recent Advances on Improving Efficient Antitumor of Lenvatinib Based on Nanoparticle Delivery System. Int J Nanomedicine 2024; 19:5581-5603. [PMID: 38882543 PMCID: PMC11177867 DOI: 10.2147/ijn.s460844] [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: 01/23/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
Lenvatinib (LVN) is a potentially effective multiple-targeted receptor tyrosine kinase inhibitor approved for treating hepatocellular carcinoma, metastatic renal cell carcinoma and thyroid cancer. Nonetheless, poor pharmacokinetic properties including poor water solubility and rapid metabolic, complex tumor microenvironment, and drug resistance have impeded its satisfactory therapeutic efficacy. This article comprehensively reviews the uses of nanotechnology in LVN to improve antitumor effects. With the characteristic of high modifiability and loading capacity of the nano-drug delivery system, an active targeting approach, controllable drug release, and biomimetic strategies have been devised to deliver LVN to target tumors in sequence, compensating for the lack of passive targeting. The existing applications and advances of LVN in improving therapeutic efficacy include improving longer-term efficiency, achieving higher efficiency, combination therapy, tracking and diagnosing application and reducing toxicity. Therefore, using multiple strategies combined with photothermal, photodynamic, and immunoregulatory therapies potentially overcomes multi-drug resistance, regulates unfavorable tumor microenvironment, and yields higher synergistic antitumor effects. In brief, the nano-LVN delivery system has brought light to the war against cancer while at the same time improving the antitumor effect. More intelligent and multifunctional nanoparticles should be investigated and further converted into clinical applications in the future.
Collapse
Affiliation(s)
- Haiqing Wang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Wentao Bo
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Xielin Feng
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Jinliang Zhang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Ge Li
- Department of Emergency, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Yan Chen
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| |
Collapse
|
19
|
Nosrati-Siahmazgi V, Abbaszadeh S, Musaie K, Eskandari MR, Rezaei S, Xiao B, Ghorbani-Bidkorpeh F, Shahbazi MA. NIR-Responsive injectable hydrogel cross-linked by homobifunctional PEG for photo-hyperthermia of melanoma, antibacterial wound healing, and preventing post-operative adhesion. Mater Today Bio 2024; 26:101062. [PMID: 38706729 PMCID: PMC11066557 DOI: 10.1016/j.mtbio.2024.101062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/29/2024] [Accepted: 04/14/2024] [Indexed: 05/07/2024] Open
Abstract
Current therapeutic approaches for skin cancer face significant challenges, including wound infection, delayed skin regeneration, and tumor recurrence. To overcome these challenges, an injectable adhesive near-infrared (NIR)-responsive hydrogel with time-dependent enhancement in viscosity is developed for combined melanoma therapy and antibacterial wound healing acceleration. The multifunctional hydrogel is prepared through the chemical crosslinking between poly(methyl vinyl ether-alt-maleic acid) and gelatin, followed by the incorporation of CuO nanosheets and allantoin. The synergistic inherent antibacterial potential of CuO nanosheets, the regenerative and smoothing effect of allantoin, the extracellular matrix-mimicking effect of gelatin, and the desirable swelling behavior of the hydrogel results in fast wound recovery after photothermal ablation of the tumor. Additionally, the hydrogel can serve as an alternative to sutures owing to its tissue adhesiveness ability, which can further render it the merits for accelerated repair of abdominal lesions while acting as a biocompatible barrier to prevent peritoneal adhesion.
Collapse
Affiliation(s)
- Vahideh Nosrati-Siahmazgi
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184, Zanjan, Iran
| | - Samin Abbaszadeh
- Department of Pharmacology, School of Medicine, Zanjan University of Medical Sciences, 45139-56111, Zanjan, Iran
| | - Kiyan Musaie
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713, AV Groningen, the Netherlands
| | - Mohammad Reza Eskandari
- Department of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Science, 45139-56184, Zanjan, Iran
| | - Saman Rezaei
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713, AV Groningen, the Netherlands
| | - Bo Xiao
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Fatemeh Ghorbani-Bidkorpeh
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Ali Shahbazi
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184, Zanjan, Iran
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713, AV Groningen, the Netherlands
| |
Collapse
|
20
|
Song W, He Y, Feng Y, Wang Y, Li X, Wu Y, Zhang S, Zhong L, Yan F, Sun L. Image-Guided Photothermal and Immune Therapy of Tumors via Melanin-Producing Genetically Engineered Bacteria. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305764. [PMID: 38368252 DOI: 10.1002/smll.202305764] [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: 07/10/2023] [Revised: 01/10/2024] [Indexed: 02/19/2024]
Abstract
Photothermal therapy (PTT) is a new treatment modality for tumors. However, the efficient delivery of photothermal agents into tumors remains difficult, especially in hypoxic tumor regions. In this study, an approach to deliver melanin, a natural photothermal agent, into tumors using genetically engineered bacteria for image-guided photothermal and immune therapy is developed. An Escherichia coli MG1655 is transformed with a recombinant plasmid harboring a tyrosinase gene to produce melanin nanoparticles. Melanin-producing genetically engineered bacteria (MG1655-M) are systemically administered to 4T1 tumor-bearing mice. The tumor-targeting properties of MG1655-M in the hypoxic environment integrate the properties of hypoxia targeting, photoacoustic imaging, and photothermal therapeutic agents in an "all-in-one" manner. This eliminates the need for post-modification to achieve image-guided hypoxia-targeted cancer photothermal therapy. Tumor growth is significantly suppressed by irradiating the tumor with an 808 nm laser. Furthermore, strong antitumor immunity is triggered by PTT, thereby producing long-term immune memory effects that effectively inhibit tumor metastasis and recurrence. This work proposes a new photothermal and immune therapy guided by an "all-in-one" melanin-producing genetically engineered bacteria, which can offer broad potential applications in cancer treatment.
Collapse
Affiliation(s)
- Weijian Song
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310030, P. R. China
- Bengbu Medical University, Bengbu, Anhui, 233030, P. R. China
| | - Yaling He
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Yanan Feng
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, P. R. China
| | - Yuanyuan Wang
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Xiaoying Li
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310030, P. R. China
- Bengbu Medical University, Bengbu, Anhui, 233030, P. R. China
| | - Yingnan Wu
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310030, P. R. China
- Bengbu Medical University, Bengbu, Anhui, 233030, P. R. China
| | - Shanxin Zhang
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310030, P. R. China
- Bengbu Medical University, Bengbu, Anhui, 233030, P. R. China
| | - Lin Zhong
- School of Public Health, Nanchang University, Nanchang, Jiangxi, 330019, P. R. China
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310030, P. R. China
- Bengbu Medical University, Bengbu, Anhui, 233030, P. R. China
| |
Collapse
|
21
|
Dong Z, Xue K, Verma A, Shi J, Wei Z, Xia X, Wang K, Zhang X. Photothermal therapy: a novel potential treatment for prostate cancer. Biomater Sci 2024; 12:2480-2503. [PMID: 38592730 DOI: 10.1039/d4bm00057a] [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: 04/10/2024]
Abstract
Prostate cancer (PCa) is a leading cause of cancer-related death in men, and most PCa patients treated with androgen deprivation therapy will progress to metastatic castration-resistant prostate cancer (mCRPC) due to the lack of efficient treatment. Recently, lots of research indicated that photothermal therapy (PTT) was a promising alternative that provided an accurate and efficient prostate cancer therapy. A photothermic agent (PTA) is a basic component of PPT and is divided into organic and inorganic PTAs. Besides, the combination of PTT and other therapies, such as photodynamic therapy (PDT), immunotherapy (IT), chemotherapy (CT), etc., provides an more efficient strategy for PCa therapy. Here, we introduce basic information about PTT and summarize the PTT treatment strategies for prostate cancer. Based on recent works, we think the combination of PPT and other therapies provides a novel possibility for PCa, especially CRPC clinical treatment.
Collapse
Affiliation(s)
- Zirui Dong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Kaming Xue
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Anushikha Verma
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian Shi
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Zhihao Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xiaotian Xia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan 430022, Hubei, China.
| | - Keshan Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
22
|
Khatun S, Pebam M, Sankaranarayanan SA, Pogu SV, Bantal VS, Rengan AK. Glutathione - IR 797 coupled Casein Nano-Trojan for augmenting the therapeutic efficacy of camptothecin in highly invasive triple negative breast cancer. BIOMATERIALS ADVANCES 2024; 159:213802. [PMID: 38401401 DOI: 10.1016/j.bioadv.2024.213802] [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: 08/25/2023] [Revised: 02/10/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
The rapid metastasis & heterogenic constitution of triple negative breast cancer (TNBC) limits drug entry to the tumor, reducing treatment effectiveness. To address this, we have synthesized Casein nanoparticles (Cn NPs) with attached glutathione (GSH), a natural ligand for cancer cell overexpressed γ-glutamyl transpeptidase (GGT). Cn NPs encapsulated with Camptothecin and NIR dye IR 797 (CCN NPs) for combinatorial therapy of TNBC. The GSH-CCN nanoparticles (CCNG NPs) act as a Nano-Trojan to deceive the cancer cells by delivering therapeutic payloads directly to specific target cells. In this study, Casein Nano-Trojan is equipped with GSH as a targeting ligand for GGT. The binding of CCNG NPs with cell surface receptors switched the anionic charge to catanionic, prompting the target cell to engulf the nanoparticles. The Casein Nano-Trojan releases its therapeutic payload inside the target cell, potentially inhibiting proliferation & inducing a high percentage of cell death (85 ± 7 %). Disintegration of mitochondrial membrane potential, inhibition of both migration & re-growth were observed. Immunofluorescence, acridine orange/ethidium bromide stain, and nuclear fragmentation assay further confirmed the substantial DNA damage induced by the high expression of γH2AX and p53. Significant therapeutic efficacy was observed in the 3D spheroids of 4T1 cells and in vivo breast cancer mice model (BALB/c). These findings demonstrate that CCNG NPs could be an effective treatment approach for highly metastatic triple negative breast cancer.
Collapse
Affiliation(s)
- Sajmina Khatun
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, Telangana 502285, India
| | - Monika Pebam
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, Telangana 502285, India
| | | | - Sunil Venkanna Pogu
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, Telangana 502285, India
| | | | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, Telangana 502285, India.
| |
Collapse
|
23
|
Wu Q, Li Z, Zhou X, Wei Z, Ramadan S, Xu Y, Xu L, Li D. Photothermal Ferrotherapy - Induced Immunogenic Cell Death via Iron-Based Ternary Chalcogenide Nanoparticles Against Triple-Negative Breast Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306766. [PMID: 38095479 DOI: 10.1002/smll.202306766] [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: 08/08/2023] [Revised: 11/14/2023] [Indexed: 02/01/2024]
Abstract
Triple-negative breast cancer (TNBC) is highly malignant and prone to recurrence and metastasis. Patients with TNBC have limited therapeutic options, often resulting in poor prognosis. Some new treatments for TNBC have been considered in the past decade, such as immunotherapy, photothermal therapy (PTT), and ferroptosis therapy, that allow the rapid and minimally invasive ablation of cancer. However, a multifunctional nanodrug system with more potent efficacy for TNBC is still needed. The use of iron-based ternary chalcogenide nanoparticles (NPs), namely AgFeS2, is reported, which synergistically combines photothermal therapy, ferrotherapy, and immunotherapy in one system for the treatment of TNBC. AgFeS2 possesses excellent photothermal conversion performance for tumor near-infrared (NIR) phototherapy. Upon photoirradiation, these NPs generate heat, accelerate the release of iron ions, and effectively catalyze the Fenton reaction, resulting in cell apoptosis and ferroptosis. Additionally, AgFeS2 promotes the release of tumor-specific antigens and triggers an immune response via immunogenic cell death (ICD), thereby providing unique synergistic mechanisms for cancer therapy. The present study demonstrates the great potential of iron-based ternary chalcogenide as a new therapeutic platform for a combination of photothermal therapy, ferrotherapy, and immunotherapy for the suppression of TNBC.
Collapse
Affiliation(s)
- Qiang Wu
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhuoyuan Li
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xin Zhou
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhewei Wei
- Department of Gastrointestinopancreatic Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Sami Ramadan
- Department of Materials, Imperial College London, London, SW7 2AZ, UK
| | - Yunsheng Xu
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, 518107, China
| | - Lizhou Xu
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Danyang Li
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, 518107, China
| |
Collapse
|
24
|
Belyaev IB, Zelepukin IV, Kotelnikova PA, Tikhonowski GV, Popov AA, Kapitannikova AY, Barman J, Kopylov AN, Bratashov DN, Prikhozhdenko ES, Kabashin AV, Deyev SM, Zvyagin AV. Laser-Synthesized Germanium Nanoparticles as Biodegradable Material for Near-Infrared Photoacoustic Imaging and Cancer Phototherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307060. [PMID: 38516744 PMCID: PMC11132077 DOI: 10.1002/advs.202307060] [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: 09/25/2023] [Revised: 02/20/2024] [Indexed: 03/23/2024]
Abstract
Biodegradable nanomaterials can significantly improve the safety profile of nanomedicine. Germanium nanoparticles (Ge NPs) with a safe biodegradation pathway are developed as efficient photothermal converters for biomedical applications. Ge NPs synthesized by femtosecond-laser ablation in liquids rapidly dissolve in physiological-like environment through the oxidation mechanism. The biodegradation of Ge nanoparticles is preserved in tumor cells in vitro and in normal tissues in mice with a half-life as short as 3.5 days. Biocompatibility of Ge NPs is confirmed in vivo by hematological, biochemical, and histological analyses. Strong optical absorption of Ge in the near-infrared spectral range enables photothermal treatment of engrafted tumors in vivo, following intravenous injection of Ge NPs. The photothermal therapy results in a 3.9-fold reduction of the EMT6/P adenocarcinoma tumor growth with significant prolongation of the mice survival. Excellent mass-extinction of Ge NPs (7.9 L g-1 cm-1 at 808 nm) enables photoacoustic imaging of bones and tumors, following intravenous and intratumoral administrations of the nanomaterial. As such, strongly absorbing near-infrared-light biodegradable Ge nanomaterial holds promise for advanced theranostics.
Collapse
Affiliation(s)
- Iaroslav B. Belyaev
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of SciencesMoscow117997Russia
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute)Moscow115409Russia
| | - Ivan V. Zelepukin
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of SciencesMoscow117997Russia
- Department of Medicinal ChemistryUppsala UniversityUppsala751 23Sweden
| | - Polina A. Kotelnikova
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of SciencesMoscow117997Russia
| | - Gleb V. Tikhonowski
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute)Moscow115409Russia
| | - Anton A. Popov
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute)Moscow115409Russia
| | | | - Jugal Barman
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of SciencesMoscow117997Russia
| | - Alexey N. Kopylov
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute)Moscow115409Russia
| | | | | | - Andrei V. Kabashin
- CNRSLP3Campus de Luminy – Case 917Aix Marseille UniversityMarseilleCedex13288France
| | - Sergey M. Deyev
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of SciencesMoscow117997Russia
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute)Moscow115409Russia
- Institute of Molecular TheranosticsSechenov UniversityMoscow119435Russia
| | - Andrei V. Zvyagin
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of SciencesMoscow117997Russia
- Institute of Molecular TheranosticsSechenov UniversityMoscow119435Russia
- MQ Photonics CentreMacquarie UniversitySydney2109Australia
| |
Collapse
|
25
|
Ma H, Lu C, Jin Z, Liu R, Miao Z, Zha Z, Tao Z. Rhodium-Rhenium Alloy Nanozymes for Non-inflammatory Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:21653-21664. [PMID: 38644787 DOI: 10.1021/acsami.4c02550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Analogous to thermal ablation techniques in clinical settings, cell necrosis induced during tumor photothermal therapy (PTT) can provoke an inflammatory response that is detrimental to the treatment of tumors. In this study, we employed a straightforward one-step liquid-phase reduction process to synthesize uniform RhRe nanozymes with an average hydrodynamic size of 41.7 nm for non-inflammatory photothermal therapy. The obtained RhRe nanozymes showed efficient near-infrared (NIR) light absorption for effective PTT, coupled with a remarkable capability to scavenge reactive oxygen species (ROS) for anti-inflammatory treatment. After laser irradiation, the 4T1 tumors were effectively ablated without obvious tumor recurrence within 14 days, along with no obvious increase in pro-inflammatory cytokine levels. Notably, these RhRe nanozymes demonstrated high biocompatibility with normal cells and tissues, both in vitro and in vivo, as evidenced by the lack of significant toxicity in female BALB/c mice treated with 10 mg/kg of RhRe nanozymes over a 14 day period. This research highlights RhRe alloy nanoparticles as bioactive nanozymes for non-inflammatory PTT in tumor therapy.
Collapse
Affiliation(s)
- Hongna Ma
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Chenxin Lu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Zhaoying Jin
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Rui Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Zhaohua Miao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Zhenchao Tao
- Department of Radiation Oncology, The First Affiliated Hospital of USTC West District, Anhui Provincial Cancer Hospital, Hefei, Anhui 230031, People's Republic of China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, People's Republic of China
| |
Collapse
|
26
|
Turkmen Koc SN, Rezaei Benam S, Aral IP, Shahbazi R, Ulubayram K. Gold nanoparticles-mediated photothermal and photodynamic therapies for cancer. Int J Pharm 2024; 655:124057. [PMID: 38552752 DOI: 10.1016/j.ijpharm.2024.124057] [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: 11/17/2023] [Revised: 03/18/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
Cancer remains one of the major causes of death globally, with one out of every six deaths attributed to the disease. The impact of cancer is felt on psychological, physical, and financial levels, affecting individuals, communities, and healthcare institutions. Conventional cancer treatments have many challenges and inadequacies. Nanomedicine, however, presents a promising solution by not only overcoming these problems but also offering the advantage of combined therapy for treatment-resistant cancers. Nanoparticles specifically engineered for use in nanomedicine can be efficiently targeted to cancer cells through a combination of active and passive techniques, leading to superior tumor-specific accumulation, enhanced drug availability, and reduced systemic toxicity. Among various nanoparticle formulations designed for cancer treatment, gold nanoparticles have gained prominence in the field of nanomedicine due to their photothermal, photodynamic, and immunologic effects without the need for photosensitizers or immunotherapeutic agents. To date, there is no comprehensive literature review that focuses on the photothermal, photodynamic, and immunologic effects of gold nanoparticles. In this review, significant attention has been devoted to examining the parameters pertaining to the structure of gold nanoparticles and laser characteristics, which play a crucial role in influencing the efficacy of photothermal therapy (PTT) and photodynamic therapy (PDT). Moreover, this article provides insights into the success of PTT and PDT mediated by gold nanoparticles in primary cancer treatment, as well as the immunological effects of PTT and PDT on metastasis and recurrence, providing a promising strategy for cancer therapy. In summary, gold nanoparticles, with their unique properties, have the potential for clinical application in various cancer therapies, including the treatment of primary cancer, recurrence and metastasis.
Collapse
Affiliation(s)
- Seyma Nur Turkmen Koc
- Department of Nanotechnology and Nanomedicine, Hacettepe University, Ankara, Türkiye
| | - Sanam Rezaei Benam
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Ipek Pınar Aral
- Department of Radiation Oncology, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara Bilkent City Hospital, Ankara, Türkiye
| | - Reza Shahbazi
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, USA; Tumor Microenvironment & Metastasis, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, USA; Brown Center for Immunotherapy, Indiana University School of Medicine, Indianapolis, USA.
| | - Kezban Ulubayram
- Department of Nanotechnology and Nanomedicine, Hacettepe University, Ankara, Türkiye; Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, Türkiye; Department of Bioengineering, Hacettepe University, Ankara, Türkiye.
| |
Collapse
|
27
|
Pinho S, Ferreira-Gonçalves T, Lopes J, Amaral MN, Viana AS, Coelho JMP, Gaspar MM, Reis CP. A Step Forward for the Treatment of Localized Prostate Cancer Using Gold Nanoparticles Combined with Laser Irradiation. Int J Mol Sci 2024; 25:4488. [PMID: 38674073 PMCID: PMC11050317 DOI: 10.3390/ijms25084488] [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: 03/14/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Prostate cancer (PCA) is the second most common cancer diagnosis in men and the fifth leading cause of death worldwide. The conventional treatments available are beneficial to only a few patients and, in those, some present adverse side effects that eventually affect the quality of life of most patients. Thus, there is an urgent need for effective, less invasive and targeted specific treatments for PCA. Photothermal therapy (PTT) is a minimally invasive therapy that provides a localized effect for tumour cell ablation by activating photothermal agents (PTA) that mediate the conversion of the light beam's energy into heat at the site. As tumours are unable to easily dissipate heat, they become more susceptible to temperature increases. In the PTT field, gold nanoparticles (AuNPs) have been attracting interest as PTA. The aim of this study was to formulate AuNPs capable of remaining retained in the tumour and subsequently generating heat at the tumour site. AuNPs were synthesized and characterized in terms of size, polydispersity index (PdI), zeta potential (ZP), morphology and the surface plasmon resonance (SPR). The safety of AuNPs and their efficacy were assessed using in vitro models. A preliminary in vivo safety assessment of AuNPs with a mean size lower than 200 nm was confirmed. The morphology was spherical-like and the SPR band showed good absorbance at the laser wavelength. Without laser, AuNPs proved to be safe both in vitro (>70% viability) and in vivo. In addition, with laser irradiation, they proved to be relatively effective in PCA cells. Overall, the formulation appears to be promising for use in PTT.
Collapse
Affiliation(s)
- Sara Pinho
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal; (S.P.); (T.F.-G.); (J.L.); (M.N.A.)
| | - Tânia Ferreira-Gonçalves
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal; (S.P.); (T.F.-G.); (J.L.); (M.N.A.)
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal;
| | - Joana Lopes
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal; (S.P.); (T.F.-G.); (J.L.); (M.N.A.)
| | - Mariana Neves Amaral
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal; (S.P.); (T.F.-G.); (J.L.); (M.N.A.)
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal;
| | - Ana S. Viana
- Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal;
| | - João M. P. Coelho
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal;
| | - Maria Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal; (S.P.); (T.F.-G.); (J.L.); (M.N.A.)
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal;
| | - Catarina Pinto Reis
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal; (S.P.); (T.F.-G.); (J.L.); (M.N.A.)
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal;
| |
Collapse
|
28
|
Wan S, Wang K, Huang P, Guo X, Liu W, Li Y, Zhang J, Li Z, Song J, Yang W, Zhang X, Ding X, Leong DT, Wang L. Mechanoelectronic stimulation of autologous extracellular vesicle biosynthesis implant for gut microbiota modulation. Nat Commun 2024; 15:3343. [PMID: 38637580 PMCID: PMC11026491 DOI: 10.1038/s41467-024-47710-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] [Received: 11/20/2023] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
Pathogenic gut microbiota is responsible for a few debilitating gastrointestinal diseases. While the host immune cells do produce extracellular vesicles to counteract some deleterious effects of the microbiota, the extracellular vesicles are of insufficient doses and at unreliable exposure times. Here we use mechanical stimulation of hydrogel-embedded macrophage in a bioelectronic controller that on demand boost production of up to 20 times of therapeutic extracellular vesicles to ameliorate the microbes' deleterious effects in vivo. Our miniaturized wireless bioelectronic system termed inducible mechanical activation for in-situ and sustainable generating extracellular vesicles (iMASSAGE), leverages on wireless electronics and responsive hydrogel to impose mechanical forces on macrophages to produce extracellular vesicles that rectify gut microbiome dysbiosis and ameliorate colitis. This in vivo controllable extracellular vesicles-produced system holds promise as platform to treat various other diseases.
Collapse
Affiliation(s)
- Shuangshuang Wan
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 210023, Nanjing, China
| | - Kepeng Wang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 210023, Nanjing, China
| | - Peihong Huang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 210023, Nanjing, China
| | - Xian Guo
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 210023, Nanjing, China
| | - Wurui Liu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 210023, Nanjing, China
| | - Yaocheng Li
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 210023, Nanjing, China
| | - Jingjing Zhang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 210023, Nanjing, China
| | - Zhiyang Li
- Department of Clinical Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University, 210008, Nanjing, China
| | - Jiacheng Song
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, 210023, Nanjing, China
| | - Wenjing Yang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 210023, Nanjing, China
| | - Xianzheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, 430072, Wuhan, China
| | - Xianguang Ding
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 210023, Nanjing, China.
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, 117585, Singapore.
| | - Lianhui Wang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 210023, Nanjing, China.
| |
Collapse
|
29
|
Xie M, Gong T, Wang Y, Li Z, Lu M, Luo Y, Min L, Tu C, Zhang X, Zeng Q, Zhou Y. Advancements in Photothermal Therapy Using Near-Infrared Light for Bone Tumors. Int J Mol Sci 2024; 25:4139. [PMID: 38673726 PMCID: PMC11050412 DOI: 10.3390/ijms25084139] [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: 02/27/2024] [Revised: 03/31/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Bone tumors, particularly osteosarcoma, are prevalent among children and adolescents. This ailment has emerged as the second most frequent cause of cancer-related mortality in adolescents. Conventional treatment methods comprise extensive surgical resection, radiotherapy, and chemotherapy. Consequently, the management of bone tumors and bone regeneration poses significant clinical challenges. Photothermal tumor therapy has attracted considerable attention owing to its minimal invasiveness and high selectivity. However, key challenges have limited its widespread clinical use. Enhancing the tumor specificity of photosensitizers through targeting or localized activation holds potential for better outcomes with fewer adverse effects. Combinations with chemotherapies or immunotherapies also present avenues for improvement. In this review, we provide an overview of the most recent strategies aimed at overcoming the limitations of photothermal therapy (PTT), along with current research directions in the context of bone tumors, including (1) target strategies, (2) photothermal therapy combined with multiple therapies (immunotherapies, chemotherapies, and chemodynamic therapies, magnetic, and photodynamic therapies), and (3) bifunctional scaffolds for photothermal therapy and bone regeneration. We delve into the pros and cons of these combination methods and explore current research focal points. Lastly, we address the challenges and prospects of photothermal combination therapy.
Collapse
Affiliation(s)
- Mengzhang Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (M.X.); (T.G.); (Y.W.); (Z.L.); (M.L.); (Y.L.); (L.M.); (C.T.)
| | - Taojun Gong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (M.X.); (T.G.); (Y.W.); (Z.L.); (M.L.); (Y.L.); (L.M.); (C.T.)
| | - Yitian Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (M.X.); (T.G.); (Y.W.); (Z.L.); (M.L.); (Y.L.); (L.M.); (C.T.)
| | - Zhuangzhuang Li
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (M.X.); (T.G.); (Y.W.); (Z.L.); (M.L.); (Y.L.); (L.M.); (C.T.)
| | - Minxun Lu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (M.X.); (T.G.); (Y.W.); (Z.L.); (M.L.); (Y.L.); (L.M.); (C.T.)
| | - Yi Luo
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (M.X.); (T.G.); (Y.W.); (Z.L.); (M.L.); (Y.L.); (L.M.); (C.T.)
| | - Li Min
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (M.X.); (T.G.); (Y.W.); (Z.L.); (M.L.); (Y.L.); (L.M.); (C.T.)
| | - Chongqi Tu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (M.X.); (T.G.); (Y.W.); (Z.L.); (M.L.); (Y.L.); (L.M.); (C.T.)
| | - Xingdong Zhang
- National Engineering Biomaterials, Sichuan University Research Center for Chengdu, Chengdu 610064, China;
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterials, Institute of Regulatory Science for Medical Devices, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Qin Zeng
- National Engineering Biomaterials, Sichuan University Research Center for Chengdu, Chengdu 610064, China;
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterials, Institute of Regulatory Science for Medical Devices, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yong Zhou
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China; (M.X.); (T.G.); (Y.W.); (Z.L.); (M.L.); (Y.L.); (L.M.); (C.T.)
| |
Collapse
|
30
|
Shin MJ, Park JY, Park JY, Lim SH, Lim H, Choi JK, Park CK, Kang YJ, Khang D. Inflammation-Targeting Mesenchymal Stem Cells Combined with Photothermal Treatment Attenuate Severe Joint Inflammation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304333. [PMID: 38096399 DOI: 10.1002/adma.202304333] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 12/07/2023] [Indexed: 12/23/2023]
Abstract
Current clinical therapeutic efficacy for the treatment of osteo- and rheumatoid-arthritis is obviously limited. Although mesenchymal stem cells (MSCs) are considered as a source of promising regenerative therapy, un-modified or genetically engineered MSCs injected in vivo restrict their clinical utility because of the low drug efficacy and unpredicted side effect, respectively. Herein, a strategy to enhance the migration efficacy of MSCs to inflamed joints via an inflammation-mediated education process is demonstrated. To reinforce the limited anti-inflammatory activity of MSCs, gold nanostar loaded with triamcinolone is conjugated to MSC. Furthermore, near-infrared laser-assisted photothermal therapy (PTT) induced by gold nanostar significantly elevates the anti-inflammatory efficacy of the developed drugs, even in advanced stage arthritis model. An immunological regulation mechanism study of PTT is first suggested in this study; the expression of the interleukin 22 receptor, implicated in the pathogenesis of arthritis, is downregulated in T lymphocytes by PTT, and Th17 differentiation from naïve CD4 T cell is inhibited. Collectively, inflammation-targeting MSCs conjugated with triamcinolone-loaded gold nanostar (Edu-MSCs-AuS-TA) promote the repolarization of macrophages and decrease neutrophil recruitment in joints. In addition, Edu-MSCs-AuS-TA significantly alleviate arthritis-associated pain, improve general locomotor activity, and more importantly, induce cartilage regeneration even for severe stages of arthritis model.
Collapse
Affiliation(s)
- Min Jun Shin
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
| | - Jun-Young Park
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Jun Young Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
| | - Su Hyun Lim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
| | - Hyoungsub Lim
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Jin Kyeong Choi
- Department of Immunology, School of Medicine, Jeonbuk National University, Jeonju, 54907, South Korea
| | - Chul-Kyu Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
- Department of Physiology, School of Medicine, Gachon University, Incheon, 21999, South Korea
| | - Youn Joo Kang
- Department of Rehabilitation Medicine, Eulji Hospital, School of Medicine, Eulji University, Seoul, 01830, South Korea
| | - Dongwoo Khang
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
- Department of Physiology, School of Medicine, Gachon University, Incheon, 21999, South Korea
- Ectosome Inc., Incheon, 21999, South Korea
| |
Collapse
|
31
|
Kim J, Nah Y, Kim S, Kim WJ. Transformation of nanoparticles via the transition of functional DNAs responsive to pH and vascular endothelial growth factor for photothermal anti-tumor therapy. Biomater Sci 2024; 12:1031-1041. [PMID: 38214329 DOI: 10.1039/d3bm01968c] [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: 01/13/2024]
Abstract
This study presents a novel approach for the development of DNA-functionalized gold nanoparticles (AuNPs) capable of responding to disease-specific factors and microenvironmental changes, resulting in an effective anti-tumor effect via photothermal therapy. The AuNPs are decorated with two types of DNAs, an i-motif duplex and a VEGF split aptamer, enabling recognition of changes in pH and VEGF, respectively. The formation of VEGF aptamers on the AuNPs induces their aggregation, further enhanced by VEGF ligands. The resulting changes in the optical properties of the AuNPs are detected by monitoring the absorbance. Upon irradiation with a near-infrared laser, the aggregated AuNPs generate heat due to their thermoplasmonic characteristic, leading to an anti-tumor effect. This study demonstrates the enhanced anti-tumor effect of DNA-functionalized AuNPs via photothermal therapy in both in vitro and in vivo tumor models. These findings suggest the potential utilization of such functional AuNPs for precise disease diagnosis and treatment by detecting disease-related factors in the microenvironment.
Collapse
Affiliation(s)
- Jinseong Kim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea.
| | - Yunyoung Nah
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea.
| | - Seongmin Kim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea.
| | - Won Jong Kim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea.
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- OmniaMed Co., Ltd, Pohang, Republic of Korea
| |
Collapse
|
32
|
Zeng H, Yan G, Zheng R, Wang X. Cancer Cell Membrane-Biomimetic Nanoparticles Based on Gelatin and Mitoxantrone for Synergetic Chemo-Photothermal Therapy of Metastatic Breast Cancer. ACS Biomater Sci Eng 2024; 10:875-889. [PMID: 38284758 DOI: 10.1021/acsbiomaterials.3c01325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
The purpose of this paper is to develop a cancer cell membrane biomimetic nanodrug delivery system (NDDS) to achieve an enhanced chemo-photothermal synergistic antitumor effect. The biomimetic NDDSs are composed of mitoxantrone (MIT)-loaded gelatin nanoparticles and IR820-encapsulated 4T1 cancer cell membrane-derived vesicles. The biomimetic NDDS displayed excellent stability and photothermal conversion efficiency. Compared to naked nanoparticles, the cell membrane-coated nanoparticles improved 4T1 cell uptake through homologous targeting and effectively reduced internalization of macrophages. In vivo photothermal imaging results further showed that the NDDS could be enriched at the tumor site for 48 h and could raise the temperature of the tumor area to 60 °C within 5 min under 808 nm laser irradiation. Finally, NDDS successfully inhibited primary tumor growth (over 89% inhibition) and significantly inhibited lung metastasis. This study may provide a new strategy for personalized chemotherapy-photothermal combination therapy of metastatic breast cancer using tumor cell membranes from cancer patients as drug carriers.
Collapse
Affiliation(s)
- Huihui Zeng
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Bengbu Medical College, Bengbu 233004, Anhui Province, PR China
- Department of Medical Oncology, the First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, Anhui Province, PR China
| | - Guoqing Yan
- School of Life Science, Anhui University, Hefei 230601, Anhui, PR China
| | - Rongsheng Zheng
- Department of Medical Oncology, the First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, Anhui Province, PR China
| | - Xin Wang
- School of Life Science, Anhui University, Hefei 230601, Anhui, PR China
| |
Collapse
|
33
|
Wang W, Cheng Z, Xing H, Zhou S, Ye Q, Xiong G, Wang G, Ma D. Red cell membrane-coating Prussian blue for combined photothermal and NO gas therapy for nasopharyngeal carcinoma. J Mater Chem B 2024; 12:1579-1591. [PMID: 38259153 DOI: 10.1039/d3tb02444j] [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: 01/24/2024]
Abstract
Nitric oxide (NO) gas molecules have demonstrated remarkable anti-tumor effects and minimal susceptibility to drug resistance, establishing as a promising modality for effective tumor treatment. However, how to realize its stable and efficient delivery in vivo is still a challenge. In this study, we have developed a heat-responsive biomimetic nano erythrocyte (M/B@R) by loading a NO donor (BNN6) onto mesoporous Prussian blue (M-PB) and subsequently enveloping them with red blood cell membranes. The preserved integrity of the red blood cell membrane (RBCm) structure could ensure its excellent biosafety, prolong its circulation time within the bloodstream and then enhance the accumulation of BNN6 at tumor sites. When M/B@R is stimulated by near-infrared light (NIR-II, 808 nm) irradiation, the nanoparticle could generate significant heat for photothermal therapy (PTT) by the characteristic NIR absorption of M-PB and then NO could also be efficiently released. The generated NO further facilitates the formation of ONOO-, a highly toxic species to tumors, while also alleviating tumor hypoxia. Remarkably, M/B@R, with NIR as the excitation source, induces combined lethality through hyperthermia, DNA damage, and tumor hypoxia relief. This novel combination strategy provides a new avenue for PTT/NO-induced cancer therapy.
Collapse
Affiliation(s)
- Wenbo Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Zhaoyi Cheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Hui Xing
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Shihao Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Qiaozhang Ye
- Dalang Hospital of Dongguan, Dongguan 523000, China.
| | - Gaofei Xiong
- Dalang Hospital of Dongguan, Dongguan 523000, China.
| | - Guanhai Wang
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Dong Ma
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
34
|
Ko MJ, Yoo W, Min S, Zhang YS, Joo J, Kang H, Kim DH. Photonic control of image-guided ferroptosis cancer nanomedicine. Coord Chem Rev 2024; 500:215532. [PMID: 38645709 PMCID: PMC11027759 DOI: 10.1016/j.ccr.2023.215532] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Photonic nanomaterials, characterized by their remarkable photonic tunability, empower a diverse range of applications, including cutting-edge advances in cancer nanomedicine. Recently, ferroptosis has emerged as a promising alternative strategy for effectively killing cancer cells with minimizing therapeutic resistance. Novel design of photonic nanomaterials that can integrate photoresponsive-ferroptosis inducers, -diagnostic imaging, and -synergistic components provide significant benefits to effectively trigger local ferroptosis. This review provides a comprehensive overview of recent advancements in photonic nanomaterials for image-guided ferroptosis cancer nanomedicine, offering insights into their strengths, constraints, and their potential as a future paradigm in cancer treatment.
Collapse
Affiliation(s)
- Min Jun Ko
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Woojung Yoo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital Harvard Medical School, Cambridge, MA 02139, USA
| | - Jinmyoung Joo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
- College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, USA
| |
Collapse
|
35
|
Bashiru M, Macchi S, Forson M, Khan A, Ishtiaq A, Oyebade A, Jalihal A, Ali N, Griffin RJ, Oyelere AK, Hooshmand N, Siraj N. Doxorubicin-Based Ionic Nanomedicines for Combined Chemo-Phototherapy of Cancer. ACS APPLIED NANO MATERIALS 2024; 7:2176-2189. [PMID: 38410412 PMCID: PMC10896075 DOI: 10.1021/acsanm.3c05464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Synergistic combination therapy approach offers lots of options for delivery of materials with anticancer properties, which is a very promising strategy to treat a variety of malignant lesions with enhanced therapeutic efficacy. The current study involves a detailed investigation of combination ionic nanomedicines where a chemotherapeutic drug is coupled with a photothermal agent to attain dual mechanisms (chemotherapy (chemo) and photothermal therapy (PTT)) to improve the drug's efficacy. An FDA-approved Doxorubicin hydrochloride (DOX·HCl) is electrostatically attached with a near-infrared cyanine dye (ICG, IR783, and IR820), which serves as a PTT drug using ionic liquid chemistry to develop three ionic material (IM)-based chemo-PTT drugs. Carrier-free ionic nanomedicines (INMs) are derived from ionic materials (IMs). The photophysical properties of the developed combination IMs and their INMs were studied in depth. The phototherapeutic efficiency of the combination drugs was evaluated by measuring the photothermal conversion efficiency and singlet-oxygen quantum yield. The improved photophysical properties of the combination nanomedicines in comparison to their parent compounds significantly enhanced INMs' photothermal efficiency. Cellular uptake, dark and light toxicity studies, and cell death mechanisms of the chemo-PTT nanoparticles were also studied in vitro. The combination INMs exhibited enhanced cytotoxicity compared to their respective parent compounds. Moreover, the apoptosis cell death mechanism was almost doubled for combination nanomedicine than the free DOX, which is attributed to enhanced cellular uptake. Examination of the combination index and improved in vitro cytotoxicity results revealed a great synergy between chemo and PTT drugs in the developed combination nanomedicines.
Collapse
Affiliation(s)
- Mujeebat Bashiru
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Samantha Macchi
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Mavis Forson
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Amna Khan
- Department of Chemistry, University of Arkansas at Fayetteville, Fayetteville, Arkansas 72701, United States
| | - Arisha Ishtiaq
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Adeniyi Oyebade
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Amanda Jalihal
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Nawab Ali
- Department of Biology, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Robert J Griffin
- Department of Radiation Oncology, Arkansas Nanomedicine Center, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Adegboyega K Oyelere
- School of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Nasrin Hooshmand
- Laser Dynamics Laboratory, School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Noureen Siraj
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| |
Collapse
|
36
|
Yan Z, Liu Z, Zhang H, Guan X, Xu H, Zhang J, Zhao Q, Wang S. Current trends in gas-synergized phototherapy for improved antitumor theranostics. Acta Biomater 2024; 174:1-25. [PMID: 38092250 DOI: 10.1016/j.actbio.2023.12.012] [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: 09/04/2023] [Revised: 11/14/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
Phototherapy, such as photothermal therapy (PTT) and photodynamic therapy (PDT), has been considered an elegant solution to eradicate tumors due to its minimal invasiveness and low systemic toxicity. Nevertheless, it is still challenging for phototherapy to achieve ideal outcomes and clinical translation due to its inherent drawbacks. Owing to the unique biological functions, diverse gases have attracted growing attention in combining with phototherapy to achieve super-additive therapeutic effects. Specifically, gases such as nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S) have been proven to kill tumor cells by inducing mitochondrial damage in synergy with phototherapy. Additionally, several gases not only enhance the thermal damage in PTT and the reactive oxygen species (ROS) production in PDT but also improve the tumor accumulation of photoactive agents. The inflammatory responses triggered by hyperthermia in PTT are also suppressed by the combination of gases. Herein, we comprehensively review the latest studies on gas-synergized phototherapy for cancer therapy, including (1) synergistic mechanisms of combining gases with phototherapy; (2) design of nanoplatforms for gas-synergized phototherapy; (3) multimodal therapy based on gas-synergized phototherapy; (4) imaging-guided gas-synergized phototherapy. Finally, the current challenges and future opportunities of gas-synergized phototherapy for tumor treatment are discussed. STATEMENT OF SIGNIFICANCE: 1. The novelty and significance of the work with respect to the existing literature. (1) Strategies to design nanoplatforms for gas-synergized anti-tumor phototherapy have been summarized for the first time. Meanwhile, the integration of various imaging technologies and therapy modalities which endow these nanoplatforms with advanced theranostic capabilities has been summarized. (2) The mechanisms by which gases synergize with phototherapy to eradicate tumors are innovatively and comprehensively summarized. 2. The scientific impact and interest. This review elaborates current trends in gas-synergized anti-tumor phototherapy, with special emphases on synergistic anti-tumor mechanisms and rational design of therapeutic nanoplatforms to achieve this synergistic therapy. It aims to provide valuable guidance for researchers in this field.
Collapse
Affiliation(s)
- Ziwei Yan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Zhu Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Haotian Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Xinyao Guan
- Experimental Teaching Center, Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Hongwei Xu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Jinghai Zhang
- Department of Biomedical Engineering, School of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|
37
|
Premji TP, Dash BS, Das S, Chen JP. Functionalized Nanomaterials for Inhibiting ATP-Dependent Heat Shock Proteins in Cancer Photothermal/Photodynamic Therapy and Combination Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:112. [PMID: 38202567 PMCID: PMC10780407 DOI: 10.3390/nano14010112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024]
Abstract
Phototherapies induced by photoactive nanomaterials have inspired and accentuated the importance of nanomedicine in cancer therapy in recent years. During these light-activated cancer therapies, a nanoagent can produce heat and cytotoxic reactive oxygen species by absorption of light energy for photothermal therapy (PTT) and photodynamic therapy (PDT). However, PTT is limited by the self-protective nature of cells, with upregulated production of heat shock proteins (HSP) under mild hyperthermia, which also influences PDT. To reduce HSP production in cancer cells and to enhance PTT/PDT, small HSP inhibitors that can competitively bind at the ATP-binding site of an HSP could be employed. Alternatively, reducing intracellular glucose concentration can also decrease ATP production from the metabolic pathways and downregulate HSP production from glucose deprivation. Other than reversing the thermal resistance of cancer cells for mild-temperature PTT, an HSP inhibitor can also be integrated into functionalized nanomaterials to alleviate tumor hypoxia and enhance the efficacy of PDT. Furthermore, the co-delivery of a small-molecule drug for direct HSP inhibition and a chemotherapeutic drug can integrate enhanced PTT/PDT with chemotherapy (CT). On the other hand, delivering a glucose-deprivation agent like glucose oxidase (GOx) can indirectly inhibit HSP and boost the efficacy of PTT/PDT while combining these therapies with cancer starvation therapy (ST). In this review, we intend to discuss different nanomaterial-based approaches that can inhibit HSP production via ATP regulation and their uses in PTT/PDT and cancer combination therapy such as CT and ST.
Collapse
Affiliation(s)
- Thejas P. Premji
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (T.P.P.); (B.S.D.); (S.D.)
| | - Banendu Sunder Dash
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (T.P.P.); (B.S.D.); (S.D.)
| | - Suprava Das
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (T.P.P.); (B.S.D.); (S.D.)
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (T.P.P.); (B.S.D.); (S.D.)
- Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33305, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
38
|
Komedchikova EN, Kolesnikova OA, Syuy AV, Volkov VS, Deyev SM, Nikitin MP, Shipunova VO. Targosomes: Anti-HER2 PLGA nanocarriers for bioimaging, chemotherapy and local photothermal treatment of tumors and remote metastases. J Control Release 2024; 365:317-330. [PMID: 37996056 DOI: 10.1016/j.jconrel.2023.11.036] [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: 07/10/2023] [Revised: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 11/25/2023]
Abstract
Developing combined cancer therapy strategies is of utmost importance as it can enhance treatment efficacy, overcome drug resistance, and ultimately improve patient outcomes by targeting multiple pathways and mechanisms involved in cancer growth and progression. Specifically, the potential of developing a combination chemo&photothermal therapy using targeted polymer nanoparticles as nanocarriers offers a promising approach for synergistic cancer treatment by combining the benefits of both therapies, such as targeted drug delivery and localized hyperthermia. Here, we report the first targeted anti-HER2 PLGA nanocarriers, called targosomes, that simultaneously possess photothermal, chemotherapeutic and diagnostic properties using only molecular payloads. Biocompatible poly(lactic-co-glycolic acid), PLGA, nanoparticles were loaded with photosensitizer phthalocyanine, diagnostic dye Nile Blue, and chemotherapeutic drug irinotecan, which was chosen as a result of screening a panel of theragnostic nanoparticles. The targeted delivery to cell surface oncomarker HER2 was ensured by nanoparticle modification with the anti-HER2 monoclonal antibody, trastuzumab, using the one-pot synthesis method without chemical conjugation. The irradiation tests revealed prominent photothermal properties of nanoparticles, namely heating by 35 °C in 10 min. Nanoparticles exhibited a 7-fold increase in binding and nearly an 18-fold increase in cytotoxicity for HER2-overexpressing cells compared to cells lacking HER2 expression. This enhancement of cytotoxicity was further amplified by >20-fold under NIR light irradiation. In vivo studies proved the efficacy of nanoparticles for bioimaging of primary tumor and metastasis sites and demonstrated 93% tumor growth inhibition, making these nanoparticles excellent candidates for translation into theragnostic applications.
Collapse
Affiliation(s)
- E N Komedchikova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - O A Kolesnikova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - A V Syuy
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - V S Volkov
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - S M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - M P Nikitin
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - V O Shipunova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia.
| |
Collapse
|
39
|
Ramon J, Engelen Y, De Keersmaecker H, Goemaere I, Punj D, Mejía Morales J, Bonte C, Berx G, Hoste E, Stremersch S, Lentacker I, De Smedt SC, Raemdonck K, Braeckmans K. Laser-induced vapor nanobubbles for B16-F10 melanoma cell killing and intracellular delivery of chemotherapeutics. J Control Release 2024; 365:1019-1036. [PMID: 38065413 DOI: 10.1016/j.jconrel.2023.12.006] [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: 10/07/2022] [Revised: 11/23/2023] [Accepted: 12/02/2023] [Indexed: 12/25/2023]
Abstract
The most lethal form of skin cancer is cutaneous melanoma, a tumor that develops in the melanocytes, which are found in the epidermis. The treatment strategy of melanoma is dependent on the stage of the disease and often requires combined local and systemic treatment. Over the years, systemic treatment of melanoma has been revolutionized and shifted toward immunotherapeutic approaches. Phototherapies like photothermal therapy (PTT) have gained considerable attention in the field, mainly because of their straightforward applicability in melanoma skin cancer, combined with the fact that these strategies are able to induce immunogenic cell death (ICD), linked with a specific antitumor immune response. However, PTT comes with the risk of uncontrolled heating of the surrounding healthy tissue due to heat dissipation. Here, we used pulsed laser irradiation of endogenous melanin-containing melanosomes to induce cell killing of B16-F10 murine melanoma cells in a non-thermal manner. Pulsed laser irradiation of the B16-F10 cells resulted in the formation of water vapor nanobubbles (VNBs) around endogenous melanin-containing melanosomes, causing mechanical cell damage. We demonstrated that laser-induced VNBs are able to kill B16-F10 cells with high spatial resolution. When looking more deeply into the cell death mechanism, we found that a large part of the B16-F10 cells succumbed rapidly after pulsed laser irradiation, reaching maximum cell death already after 4 h. Practically all necrotic cells demonstrated exposure of phosphatidylserine on the plasma membrane and caspase-3/7 activity, indicative of regulated cell death. Furthermore, calreticulin, adenosine triphosphate (ATP) and high-mobility group box 1 (HMGB1), three key damage-associated molecular patterns (DAMPs) in ICD, were found to be exposed from B16-F10 cells upon pulsed laser irradiation to an extent that exceeded or was comparable to the bona fide ICD-inducer, doxorubicin. Finally, we could demonstrate that VNB formation from melanosomes induced plasma membrane permeabilization. This allowed for enhanced intracellular delivery of bleomycin, an ICD-inducing chemotherapeutic, which further boosted cell death with the potential to improve the systemic antitumor immune response.
Collapse
Affiliation(s)
- Jana Ramon
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium.
| | - Yanou Engelen
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Ghent University, 9000 Ghent, Belgium.
| | - Herlinde De Keersmaecker
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Light Microscopy Core Facility, Ghent University, 9000 Ghent, Belgium.
| | - Ilia Goemaere
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium.
| | - Deep Punj
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium.
| | - Julián Mejía Morales
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium.
| | - Cédric Bonte
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium.
| | - Geert Berx
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; VIB Center for Inflammation Research, 9052 Ghent, Belgium; Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| | - Esther Hoste
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| | - Stephan Stremersch
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium
| | - Ine Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Ghent University, 9000 Ghent, Belgium.
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Ghent University, 9000 Ghent, Belgium.
| | - Koen Raemdonck
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Ghent University, 9000 Ghent, Belgium.
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium.
| |
Collapse
|
40
|
Li M, Guo Q, Zhong C, Zhang Z. Multifunctional cell membranes-based nano-carriers for targeted therapies: a review of recent trends and future perspective. Drug Deliv 2023; 30:2288797. [PMID: 38069500 PMCID: PMC10987056 DOI: 10.1080/10717544.2023.2288797] [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: 07/31/2023] [Accepted: 11/05/2023] [Indexed: 12/18/2023] Open
Abstract
Nanotechnology has ignited a transformative revolution in disease detection, prevention, management, and treatment. Central to this paradigm shift is the innovative realm of cell membrane-based nanocarriers, a burgeoning class of biomimetic nanoparticles (NPs) that redefine the boundaries of biomedical applications. These remarkable nanocarriers, designed through a top-down approach, harness the intrinsic properties of cell-derived materials as their fundamental building blocks. Through shrouding themselves in natural cell membranes, these nanocarriers extend their circulation longevity and empower themselves to intricately navigate and modulate the multifaceted microenvironments associated with various diseases. This comprehensive review provides a panoramic view of recent breakthroughs in biomimetic nanomaterials, emphasizing their diverse applications in cancer treatment, cardiovascular therapy, viral infections, COVID-19 management, and autoimmune diseases. In this exposition, we deliver a concise yet illuminating overview of the distinctive properties underpinning biomimetic nanomaterials, elucidating their pivotal role in biomedical innovation. We subsequently delve into the exceptional advantages these nanomaterials offer, shedding light on the unique attributes that position them at the forefront of cutting-edge research. Moreover, we briefly explore the intricate synthesis processes employed in creating these biomimetic nanocarriers, shedding light on the methodologies that drive their development.
Collapse
Affiliation(s)
- Mo Li
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun, China
| | - Qiushi Guo
- Pharmacy Department, First Hospital of Jilin University—the Eastern Division, Changchun, China
| | - Chongli Zhong
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun, China
| | - Ziyan Zhang
- Department of Orthopedics, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
41
|
Zając A, Maciejczyk A, Sumorek-Wiadro J, Filipek K, Deryło K, Langner E, Pawelec J, Wasiak M, Ścibiorski M, Rzeski W, Tchórzewski M, Reichert M, Jakubowicz-Gil J. The Role of Bcl-2 and Beclin-1 Complex in "Switching" between Apoptosis and Autophagy in Human Glioma Cells upon LY294002 and Sorafenib Treatment. Cells 2023; 12:2670. [PMID: 38067099 PMCID: PMC10705223 DOI: 10.3390/cells12232670] [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: 10/25/2023] [Revised: 11/13/2023] [Accepted: 11/19/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Gliomas are the most malignant tumors of the central nervous system. One of the factors in their high drug resistance is avoiding programmed death (PCD) induction. This is related to the overexpression of intracellular survival pathways: PI3K-Akt/PKB-mTOR and Ras-Raf-MEK-ERK. Apoptosis and autophagy are co-existing processes due to the interactions between Bcl-2 and beclin-1 proteins. Their complex may be a molecular "toggle-switch" between PCD types. The aim of this research was to investigate the role of Bcl-2:beclin-1 complex in glioma cell elimination through the combined action of LY294002 and sorafenib. METHODS Drug cytotoxicity was estimated with an MTT test. The type of cell death was evaluated using variant microscopy techniques (fluorochrome staining, immunocytochemistry, and transmission electron microscopy), as well as the Bcl-2:beclin-1 complex formation and protein localization. Molecular analysis of PCD indicators was conducted through immunoblotting, immunoprecipitation, and ELISA testing. SiRNA was used to block Bcl-2 and beclin-1 expression. RESULTS The results showed the inhibitors used in simultaneous application resulted in Bcl-2:beclin-1 complex formation and apoptosis becoming dominant. This was accompanied by changes in the location of the tested proteins. CONCLUSIONS "Switching" between apoptosis and autophagy using PI3K and Raf inhibitors with Bcl-2:beclin-1 complex formation opens new therapeutic perspectives against gliomas.
Collapse
Affiliation(s)
- Adrian Zając
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (A.M.); (J.S.-W.); (M.Ś.); (W.R.); (J.J.-G.)
| | - Aleksandra Maciejczyk
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (A.M.); (J.S.-W.); (M.Ś.); (W.R.); (J.J.-G.)
| | - Joanna Sumorek-Wiadro
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (A.M.); (J.S.-W.); (M.Ś.); (W.R.); (J.J.-G.)
| | - Kamil Filipek
- Department of Molecular Biology, Institute of Biological Sciences, Maria Curie-Skłodowska University, 20-033 Lublin, Poland; (K.F.); (K.D.); (M.T.)
| | - Kamil Deryło
- Department of Molecular Biology, Institute of Biological Sciences, Maria Curie-Skłodowska University, 20-033 Lublin, Poland; (K.F.); (K.D.); (M.T.)
| | - Ewa Langner
- Department of Medical Biology, Institute of Rural Health, Jaczewskiego 2, 20-950 Lublin, Poland;
| | - Jarosław Pawelec
- Institute Microscopy Laboratory, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland;
| | - Magdalena Wasiak
- Department of Pathological Anatomy, National Veterinary Research Institute, 57 Partyzantów Avenue, 24-100 Puławy, Poland; (M.W.); (M.R.)
| | - Mateusz Ścibiorski
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (A.M.); (J.S.-W.); (M.Ś.); (W.R.); (J.J.-G.)
| | - Wojciech Rzeski
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (A.M.); (J.S.-W.); (M.Ś.); (W.R.); (J.J.-G.)
- Department of Medical Biology, Institute of Rural Health, Jaczewskiego 2, 20-950 Lublin, Poland;
| | - Marek Tchórzewski
- Department of Molecular Biology, Institute of Biological Sciences, Maria Curie-Skłodowska University, 20-033 Lublin, Poland; (K.F.); (K.D.); (M.T.)
| | - Michał Reichert
- Department of Pathological Anatomy, National Veterinary Research Institute, 57 Partyzantów Avenue, 24-100 Puławy, Poland; (M.W.); (M.R.)
| | - Joanna Jakubowicz-Gil
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (A.M.); (J.S.-W.); (M.Ś.); (W.R.); (J.J.-G.)
| |
Collapse
|
42
|
Shao C, Gong X, Zhang D, Jiang XD, Du J, Wang G. Aza-BODIPY with two efficacious fragments for NIR light-driven photothermal therapy by triggering cancer cell apoptosis. J Mater Chem B 2023; 11:10625-10631. [PMID: 37920935 DOI: 10.1039/d3tb02132g] [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: 11/04/2023]
Abstract
The reasonable structure of aza-BODIPY renders it as an efficient photothermal reagent for photothermal therapy. Herein, we describe the design and synthesis of aza-BODIPY NMeBu with the free rotating tert-butyl group and the dimethylamino-substituted segment to promote the photothermal conversion via the excited state non-radiative transition. NMeBu was found to be the π-π stacking form in the unit cell based on X-ray analysis. NMeBu-NPs by self-assembly possessed a near-infrared absorption (λabs = 772 nm), and once activated by near-infrared light, the photothermal efficiency in aqueous solution can reach 49.3%. NMeBu-NPs can penetrate the cell and trigger cell death via the apoptosis pathway under low concentration and low light power irradiation, thereby avoiding dark toxicity. Aza-BODIPY created using this procedure has excellent photothermal efficiency and could serve as a potential candidate for the treatment of cancer cells and tumors.
Collapse
Affiliation(s)
- Chunyu Shao
- Liaoning & Shenyang Key Laboratory of Functional Dye and Pigment, Shenyang University of Chemical Technology, Shenyang, China.
| | - Xiuyan Gong
- Department of Cell Biology, China Medical University, Shenyang, 110122, China.
| | - Dongxiang Zhang
- Liaoning & Shenyang Key Laboratory of Functional Dye and Pigment, Shenyang University of Chemical Technology, Shenyang, China.
| | - Xin-Dong Jiang
- Liaoning & Shenyang Key Laboratory of Functional Dye and Pigment, Shenyang University of Chemical Technology, Shenyang, China.
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, China.
| | - Guiling Wang
- Department of Cell Biology, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
43
|
Ma W, Sun R, Tang L, Li Z, Lin L, Mai Z, Chen G, Yu Z. Bioactivable STING Nanoagonists to Synergize NIR-II Mild Photothermal Therapy Primed Robust and Long-Term Anticancer Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303149. [PMID: 37691545 DOI: 10.1002/adma.202303149] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/07/2023] [Indexed: 09/12/2023]
Abstract
Pharmacological activation of the stimulator of interferon genes (STING) pathway has become a promising strategy for cancer immunotherapy. However, the insufficient tumorous accumulation, rapid clearance, and short duration of drug efficacy in the tumor microenvironment of small structural STING agonists greatly compromise the therapeutic efficacy. Herein, a tumorous extracellular matrix (ECM) is presented anchoring STING agonist-based photoimmunothernostic nanomedicine (SAPTN) that can be activated by mild-temperature photothermal therapy (mild PTT) induced neutrophilic inflammation. The SAPTN owns second window near-infrared (NIR-II) photonics properties fitting for NIR-II fluorescence and photoacoustic imaging-guided cancer therapy. The aggregates SAPTN targeting to the ECM provide slow and continuous release of potent STING agonists diABZIs. The mild PTT and long-lasting STING agonists released in the ECM synergistically prime systematic, robust, and long-term anticancer immunity. In a tumor model, this approach leads to complete tumor eradication in about 100% of mice with orthotopic breast tumors, and the mice regained tumor-free survival of at least 2 months. In addition, the immune-mediated abscopal effect shows inhibition of the distant solid tumor growth by intratumoral administration of SAPTN with laser irradiation. Overall, this approach represents a generalized photoactivable nanomedicine to prime anticancer immunity for improved cancer theranostics.
Collapse
Affiliation(s)
- Wen Ma
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Rui Sun
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Longguang Tang
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Zibo Li
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Ling Lin
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Ziyi Mai
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Gui Chen
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Zhiqiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
44
|
Xiao G, Zhao Y, Wang X, Zeng C, Luo F, Jing J. Photothermally sensitive gold nanocage augments the antitumor efficiency of immune checkpoint blockade in immune "cold" tumors. Front Immunol 2023; 14:1279221. [PMID: 37942337 PMCID: PMC10628457 DOI: 10.3389/fimmu.2023.1279221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/12/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction Immune checkpoint blockade (ICB) has revolutionized the therapy landscape of malignancy melanoma. However, the clinical benefits from this regimen remain limited, especially in tumors lacking infiltrated T cells (known as "cold" tumors). Nanoparticle-mediated photothermal therapy (PTT) has demonstrated improved outcomes in the ablation of solid tumors by inducing immunogenic cell death (ICD) and reshaping the tumor immune microenvironment. Therefore, the combination of PTT and ICB is a promising regimen for patients with "cold" tumors. Methods A second near-infrared (NIR-II) light-activated gold nanocomposite AuNC@SiO2@HA with AuNC as a kernel, silica as shell, and hyaluronic acid (HA) polymer as a targeting molecule, was synthesized for PTT. The fabricated AuNC@SiO2@HA nanocomposites underwent various in vitro studies to characterize their physicochemical properties, light absorption spectra, photothermal conversion ability, cellular uptake ability, and bioactivities. The synergistic effect of AuNC@SiO2@HA-mediated PTT and anti-PD-1 immunotherapy was evaluated using a mouse model of immune "cold" melanoma. The tumor-infiltrating T cells were assessed by immunofluorescence staining and flow cytometry. Furthermore, the mechanism of AuNC@SiO2@HA-induced T-cell infiltration was investigated through immunochemistry staining of the ICD-related markers, including HSP70, CRT, and HMGB1. Finally, the safety of AuNC@SiO2@HA nanocomposites was evaluated in vivo. Results The AuNC@SiO2@HA nanocomposite with absorption covering 1064 nm was successfully synthesized. The nano-system can be effectively delivered into tumor cells, transform the optical energy into thermal energy upon laser irradiation, and induce tumor cell apoptosis in vitro. In an in vivo mouse melanoma model, AuNC@SiO2@HA nanocomposites significantly induced ICD and T-cell infiltration. The combination of AuNC@SiO2@HA and anti-PD-1 antibody synergistically inhibited tumor growth via stimulating robust T lymphocyte immune responses. Discussion The combination of AuNC@SiO2@HA-mediated PTT and anti-PD-1 immunotherapy proposed a neoteric strategy for oncotherapy, which efficiently convert the immune "cold" tumors into "hot" ones.
Collapse
Affiliation(s)
- Guixiu Xiao
- State Key Laboratory of Biotherapy, West China Hospital, Institute for Breast Health Medicine, Sichuan University, Chengdu, Sichuan, China
- Department of Medical Oncology, Cancer Center, Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yujie Zhao
- State Key Laboratory of Biotherapy, West China Hospital, Institute for Breast Health Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Xueyan Wang
- State Key Laboratory of Biotherapy, West China Hospital, Institute for Breast Health Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Chuan Zeng
- Radiology Department, Sichuan Jianzhu Hospital, Chengdu, Sichuan, China
| | - Feng Luo
- Department of Medical Oncology, Cancer Center, Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jing Jing
- State Key Laboratory of Biotherapy, West China Hospital, Institute for Breast Health Medicine, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
45
|
Villuendas H, Vilches C, Quidant R. Standardization of In Vitro Studies for Plasmonic Photothermal therapy. ACS NANOSCIENCE AU 2023; 3:347-352. [PMID: 37868227 PMCID: PMC10588432 DOI: 10.1021/acsnanoscienceau.3c00011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 10/24/2023]
Abstract
Lack of standardization is a systematic problem that impacts nanomedicine by challenging data comparison from different studies. Translation from preclinical to clinical stages indeed requires reproducible data that can be easily accessed and compared. In this work, we propose a series of experimental standards for in vitro plasmonic photothermal therapy (PPTT). This best practice guide covers the five main aspects of PPTT studies in vitro: nanomaterials, biological samples, pre-, during, and postirradiation characterization. We are confident that such standardization of experimental protocols and reported data will benefit the development of PPTT as a transversal therapy.
Collapse
Affiliation(s)
- Helena Villuendas
- Nanophotonic
Systems Laboratory, Department of Mechanical and Process Engineering, ETH Zürich, 8092 Zürich, Switzerland
| | - Clara Vilches
- ICFO
− Institut de Ciències Fotòniques, the Barcelona Institute of Science and Technology, 08860 Castelldefels, Barcelona, Spain
| | - Romain Quidant
- Nanophotonic
Systems Laboratory, Department of Mechanical and Process Engineering, ETH Zürich, 8092 Zürich, Switzerland
| |
Collapse
|
46
|
Fernandes DA. Liposomes for Cancer Theranostics. Pharmaceutics 2023; 15:2448. [PMID: 37896208 PMCID: PMC10610083 DOI: 10.3390/pharmaceutics15102448] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/16/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Cancer is one of the most well-studied diseases and there have been significant advancements over the last few decades in understanding its molecular and cellular mechanisms. Although the current treatments (e.g., chemotherapy, radiotherapy, gene therapy and immunotherapy) have provided complete cancer remission for many patients, cancer still remains one of the most common causes of death in the world. The main reasons for the poor response rates for different cancers include the lack of drug specificity, drug resistance and toxic side effects (i.e., in healthy tissues). For addressing the limitations of conventional cancer treatments, nanotechnology has shown to be an important field for constructing different nanoparticles for destroying cancer cells. Due to their size (i.e., less than 1 μm), nanoparticles can deliver significant amounts of cancer drugs to tumors and are able to carry moieties (e.g., folate, peptides) for targeting specific types of cancer cells (i.e., through receptor-mediated endocytosis). Liposomes, composed of phospholipids and an interior aqueous core, can be used as specialized delivery vehicles as they can load different types of cancer therapy agents (e.g., drugs, photosensitizers, genetic material). In addition, the ability to load imaging agents (e.g., fluorophores, radioisotopes, MRI contrast media) enable these nanoparticles to be used for monitoring the progress of treatment. This review examines a wide variety of different liposomes for cancer theranostics, with the different available treatments (e.g., photothermal, photodynamic) and imaging modalities discussed for different cancers.
Collapse
|
47
|
Hong H, Kim M, Lee W, Jeon M, Lee C, Kim H, Im HJ, Piao Y. Injectable biocompatible nanocomposites of Prussian blue nanoparticles and bacterial cellulose as a safe and effective photothermal cancer therapy. J Nanobiotechnology 2023; 21:365. [PMID: 37798714 PMCID: PMC10552393 DOI: 10.1186/s12951-023-02108-6] [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: 06/02/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023] Open
Abstract
Photothermal therapy (PTT) is a novel cancer treatment using a photoabsorber to cause hyperthermia to kill tumors by laser irradiation. Prussian blue nanoparticles (PB NPs) are considered as next-generation photothermal agents due to the facile synthesis and excellent absorption of near-infrared light. Although PB NPs demonstrate remarkable PTT capabilities, their clinical application is limited due to their systemic toxicity. Bacterial cellulose (BC) has been applied to various bio-applications based on its unique properties and biocompatibility. Herein, we design composites with PB NPs and BC as an injectable, highly biocompatible PTT agent (IBC-PB composites). Injectable bacterial cellulose (IBC) is produced through the trituration of BC, with PB NPs synthesized on the IBC surface to prepare IBC-PB composites. IBC-PB composites show in vitro and in vivo photothermal therapeutic effects similar to those of PB NPs but with significantly greater biocompatibility. Specifically, in vitro therapeutic index of IBC-PB composites is 26.5-fold higher than that of PB NPs. Furthermore, unlike PB NPs, IBC-PB composites exhibit no overt toxicity in mice as assessed by blood biochemical analysis and histological images. Hence, it is worth pursuing further research and development of IBC-PB composites as they hold promise as safe and efficacious PTT agents for clinical application.
Collapse
Affiliation(s)
- Hwichan Hong
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - MinKyu Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Wooseung Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Miyeon Jeon
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Chaedong Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hoonsub Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyung-Jun Im
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea.
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea.
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea.
- Advanced Institutes of Convergence Technology, Seoul National University, Suwon-si, Gyeonggi-do, Republic of Korea.
- Research Institute for Convergence Science, Seoul National University, Seoul, Republic of Korea.
| | - Yuanzhe Piao
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea.
- Advanced Institutes of Convergence Technology, Seoul National University, Suwon-si, Gyeonggi-do, Republic of Korea.
- Research Institute for Convergence Science, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
48
|
Rethi L, Rethi L, Liu CH, Hyun TV, Chen CH, Chuang EY. Fortification of Iron Oxide as Sustainable Nanoparticles: An Amalgamation with Magnetic/Photo Responsive Cancer Therapies. Int J Nanomedicine 2023; 18:5607-5623. [PMID: 37814664 PMCID: PMC10560484 DOI: 10.2147/ijn.s404394] [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: 01/11/2023] [Accepted: 05/10/2023] [Indexed: 10/11/2023] Open
Abstract
Due to their non-toxic function in biological systems, Iron oxide NPs (IO-NPs) are very attractive in biomedical applications. The magnetic properties of IO-NPs enable a variety of biomedical applications. We evaluated the usage of IO-NPs for anticancer effects. This paper lists the applications of IO-NPs in general and the clinical targeting of IO-NPs. The application of IONPs along with photothermal therapy (PTT), photodynamic therapy (PDT), and magnetic hyperthermia therapy (MHT) is highlighted in this review's explanation for cancer treatment strategies. The review's study shows that IO-NPs play a beneficial role in biological activity because of their biocompatibility, biodegradability, simplicity of production, and hybrid NPs forms with IO-NPs. In this review, we have briefly discussed cancer therapy and hyperthermia and NPs used in PTT, PDT, and MHT. IO-NPs have a particular effect on cancer therapy when combined with PTT, PDT, and MHT were the key topics of the review and were covered in depth. The IO-NPs formulations may be uniquely specialized in cancer treatments with PTT, PDT, and MHT, according to this review investigation.
Collapse
Affiliation(s)
- Lekha Rethi
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - Lekshmi Rethi
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chia-Hung Liu
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Tin Van Hyun
- International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Interventional Cardiology, Thong Nhat Hospital, Ho Chi Minh City, 700000, Vietnam
| | - Chih-Hwa Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Orthopedics, Taipei Medical University – Shuang Ho Hospital, New Taipei City, Taiwan
| | - Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
49
|
Huang S, Xing F, Dai Y, Zhang Z, Zhou G, Yang S, Liu YC, Yuan Z, Luo KQ, Ying T, Chu D, Liu TM, Deng CX, Zhao Q. Navigating chimeric antigen receptor-engineered natural killer cells as drug carriers via three-dimensional mapping of the tumor microenvironment. J Control Release 2023; 362:524-535. [PMID: 37673307 DOI: 10.1016/j.jconrel.2023.09.007] [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: 04/13/2023] [Revised: 08/31/2023] [Accepted: 09/03/2023] [Indexed: 09/08/2023]
Abstract
Chimeric antigen receptor (CAR)-modified natural killer (NK) cells are recognized as promising immunotherapeutic agents for cancer treatment. However, the efficacy and trafficking of CAR-NK cells in solid tumors are hindered by the complex barriers present in the tumor microenvironment (TME). We have developed a novel strategy that utilizes living CAR-NK cells as carriers to deliver anticancer drugs specifically to the tumor site. We also introduce a time-lapse method for evaluating the efficacy and tumor specificity of CAR-NK cells using a two-photon microscope in live mouse models and three-dimensional (3D) tissue slide cultures. Our results demonstrate that CAR-NK cells exhibit enhanced antitumor immunity when combined with photosensitive chemicals in both in vitro and in vivo tumor models. Additionally, we have successfully visualized the trafficking, infiltration, and accumulation of drug-loaded CAR-NK cells in deeply situated TME using non-invasive intravital two-photon microscopy. Our findings highlight that tumor infiltration of CAR-NK cells can be intravitally monitored through the two-photon microscope approach. In conclusion, our study demonstrates the successful integration of CAR-NK cells as drug carriers and paves the way for combined cellular and small-molecule therapies in cancer treatment. Furthermore, our 3D platform offers a valuable tool for assessing the behavior of CAR cells within solid tumors, facilitating the development and optimization of immunotherapeutic strategies with clinical imaging approaches.
Collapse
Affiliation(s)
- Shigao Huang
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Radiation Oncology of Xijing Hospital, Fourth Military Medical University, Xi' an, China
| | - Fuqiang Xing
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China; Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Yeneng Dai
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Zhiming Zhang
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Guangyu Zhou
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Shuo Yang
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Yu-Cheng Liu
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Zhen Yuan
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Kathy Qian Luo
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Dafeng Chu
- Geneleap Biotechnology LLC, Woburn, MA, USA.
| | - Tzu-Ming Liu
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China.
| | - Chu-Xia Deng
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China.
| | - Qi Zhao
- Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China.
| |
Collapse
|
50
|
Guo S, Gu D, Yang Y, Tian J, Chen X. Near-infrared photodynamic and photothermal co-therapy based on organic small molecular dyes. J Nanobiotechnology 2023; 21:348. [PMID: 37759287 PMCID: PMC10523653 DOI: 10.1186/s12951-023-02111-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Near-infrared (NIR) organic small molecule dyes (OSMDs) are effective photothermal agents for photothermal therapy (PTT) due to their advantages of low cost and toxicity, good biodegradation, and strong NIR absorption over a wide wavelength range. Nevertheless, OSMDs have limited applicability in PTT due to their low photothermal conversion efficiency and inadequate destruction of tumor regions that are nonirradiated by NIR light. However, they can also act as photosensitizers (PSs) to produce reactive oxygen species (ROS), which can be further eradicated by using ROS-related therapies to address the above limitations of PTT. In this review, the synergistic mechanism, composition, and properties of photodynamic therapy (PDT)-PTT nanoplatforms were comprehensively discussed. In addition, some specific strategies for further improving the combined PTT and PDT based on OSMDs for cancer to completely eradicate cancer cells were outlined. These strategies include performing image-guided co-therapy, enhancing tumor infiltration, increasing H2O2 or O2 in the tumor microenvironment, and loading anticancer drugs onto nanoplatforms to enable combined therapy with phototherapy and chemotherapy. Meanwhile, the intriguing prospects and challenges of this treatment modality were also summarized with a focus on the future trends of its clinical application.
Collapse
Affiliation(s)
- Shuang Guo
- School of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, 116034, China
| | - Dongyu Gu
- College of Marine Science and Environment, Dalian Ocean University, Dalian, 116023, China
| | - Yi Yang
- School of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, 116034, China.
| | - Jing Tian
- School of Biological Engineering, Dalian Polytechnic University, Dalian, 116034, China.
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine, Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore.
| |
Collapse
|