1
|
Irannejadrankouhi S, Mivehchi H, Eskandari-Yaghbastlo A, Nejati ST, Emrahoglu S, Nazarian M, Zahedi F, Madani SM, Nabi-Afjadi M. Innovative nanoparticle strategies for treating oral cancers. Med Oncol 2025; 42:182. [PMID: 40285805 DOI: 10.1007/s12032-025-02728-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
Conventional therapies for oral squamous cell carcinoma (OSCC), a serious worldwide health problem, are frequently constrained by inadequate targeting and serious side effects. Drug delivery systems (DDS) based on nanoparticles provide a possible substitute by improving drug stability, target accuracy, and lowering toxicity. By addressing issues like irregular vasculature and thick tumor matrices, these methods allow for more effective medication administration. For instance, the delivery of cisplatin via liposomes, as opposed to free drug formulations, results in a 40% improvement in tumor suppression. Likewise, compared to traditional techniques, poly (lactic-co-glycolic acid) (PLGA) nanoparticles can produce up to 2.3 times more intertumoral drug accumulation. These platforms have effectively administered natural substances like curcumin and chemotherapeutics like paclitaxel, enhancing therapeutic results while reducing adverse effects. Despite their promise, several types of nanoparticles have drawbacks. For example, PLGA nanoparticles have scaling issues because of their complicated production, whereas liposomes are quickly removed from circulation. In preclinical investigations, functionalized nanoparticles-like EGFR-targeted gold nanoparticles-improve selectivity and effectiveness by obtaining up to 90% receptor binding. By preferentially accumulating in tumors via the increased permeability and retention (EPR) effect, nanoparticles also improve immunotherapy and radiation. Mechanistically, they increase the death of cancer cells by causing DNA damage, interfering with cell division, and producing reactive oxygen species (ROS). There are still issues with toxicity (such as the buildup of metallic nanoparticles in the liver) and large-scale manufacturing. Nevertheless, developments in multifunctional platforms and stimuli-responsive nanoparticles show promise for getting over these obstacles. These developments open the door to more individualized and successful OSCC therapies.
Collapse
Affiliation(s)
| | - Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | | | | | - Sahand Emrahoglu
- School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Mohammad Nazarian
- Faculty of Dentistry, Belarusion State Medical University, Minsk, Belarus
| | - Farhad Zahedi
- Institute of Molecular Biophysics, Florida State University, 91 Chieftan Way, Tallahassee, FL, 32306, USA
| | - Seyed Mahdi Madani
- Faculty of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, University of Tarbiat Modares, Tehran, Iran.
| |
Collapse
|
2
|
Dai H, Han A, Wang X, Zhu P, Wang D, Wang Y. NIR-triggering cobalt single-atom enzyme switches off-to-on for boosting the interactive dynamic effects of multimodal phototherapy. Nat Commun 2025; 16:2058. [PMID: 40021633 PMCID: PMC11871330 DOI: 10.1038/s41467-025-57188-9] [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: 11/05/2022] [Accepted: 02/14/2025] [Indexed: 03/03/2025] Open
Abstract
Noninvasive phototherapy with functional preservation is considered to be a promising cancer therapeutic method. However, the clinical application of tumor phototherapy is severely restrained by the lack of appropriate multimodal phototherapy agents exhibiting an ideal tissue penetration depth to maximize the antitumor efficiency as well as to maintain important tissue functions. Herein, an innovative near-infrared ray (NIR)-triggered photodynamic-photocatalytic-photothermal therapy (PDT-PCT-PTT) agent based on an atomically dispersed cobalt single-atom enzyme (Co-SAE) anchored on hollow N-doped carbon sphere (HNCS) has been strategically developed. Reactive oxygen species (ROS) are highly activated and amplified through both the photogenerated electrons and the photothermal conversion induced by NIR irradiation, as systematically demonstrated by the experimental and density functional theory (DFT) calculation results. Mild hyperthermia is eventually achieved through apoptosis and ferroptosis caused by ROS, significantly boosting the interaction of ROS dynamic effects and thermodynamic effects in the tumor microenvironment (TME). More importantly, Co-SAEs/HNCS not only causes multimodal damage through limited TME products but also preserves important organ functions by the induction of mild local hyperthermia. This work expands the biomedical application field of SAEs and presents an innovative all-in-one, multimodal concept for the noninvasive treatment of head and neck cancer.
Collapse
Affiliation(s)
- Hao Dai
- Department of General Dentistry II/Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials & Central Laboratory Peking University School and Hospital, Beijing, China
| | - Ali Han
- Department of Chemistry, Tsinghua University, Beijing, China.
- Shenyang National Laboratory for Materials Science, Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, Shenyang, China.
| | - Xijun Wang
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA
| | - Peng Zhu
- Department of Chemistry, Tsinghua University, Beijing, China
| | - Dingsheng Wang
- Department of Chemistry, Tsinghua University, Beijing, China.
| | - Yuguang Wang
- Department of General Dentistry II/Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials & Central Laboratory Peking University School and Hospital, Beijing, China.
| |
Collapse
|
3
|
Fan K, Li Q, Qian Y, Zhang L, Lu D, Zhu L, Yan S, Xu B, Wang Y. Enhanced Photothermal Therapy under Low-Power Near-Infrared Irradiation Enabled by a Si-Cyclopentadithiophene-Based Organic Molecule. Adv Healthc Mater 2025; 14:e2403248. [PMID: 39555634 DOI: 10.1002/adhm.202403248] [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: 08/29/2024] [Revised: 10/30/2024] [Indexed: 11/19/2024]
Abstract
Due to the inadequate photothermal conversion efficiency (PCE), most photothermal agents (PTAs) have to be used under high-power near-infrared (NIR) irradiation, which significantly exceeds medical safety standards, for achieving effective photothermal therapy (PTT) in antitumor treatment. This significantly hinders practical PTT application. Herein, three acceptor-donor-acceptor(A-D-A)-type molecules are synthesized based on cyclopentadithiophene unit to develop effective PTAs. By incorporating the large-size Si atom in the A-D-A molecules, the photosensitizer displays an increased packing distance in the aggregate state, leading to a blue-shifted absorption spectrum that better matches the medial laser wavelength. Also, the Si incorporation strategy elevates the nonradiative decay rate constants (knr) of the A-D-A photosensitizer, and thereby a further enhancement in PCE is achieved for the PTA. Consequently, the SiO-4F-based nanoparticles exhibited 64.23% PCE, with excellent biosafety and photothermal stability. Under NIR irradiation with medical safety (808 nm, 0.33 W cm-2), SiO-4F nanoparticles with 100 µg mL-1 yield a death rate of over 91% for diverse tumor cells. Moreover, in vivo experiments, SiO-4F-based PTT effectively inhibited and eliminated tumors. These findings suggest that the Si-incorporated CDPT is promising for constructing effective A-D-A photosensitizers, enabling the PTT under NIR irradiation that meets medical safety standards.
Collapse
Affiliation(s)
- Kexin Fan
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Qingyang Li
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yuping Qian
- Department of General Dentistry II, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials & Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Ludan Zhang
- Department of General Dentistry II, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials & Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Di Lu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nano-science and Technology, Beijing, 100190, China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nano-science and Technology, Beijing, 100190, China
| | - Shouke Yan
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Bowei Xu
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yuguang Wang
- Department of General Dentistry II, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials & Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| |
Collapse
|
4
|
Li Z, Zhang Y, Lei J, Wu Y. Autophagy in oral cancer: Promises and challenges (Review). Int J Mol Med 2024; 54:116. [PMID: 39422076 PMCID: PMC11518578 DOI: 10.3892/ijmm.2024.5440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Autophagy captures damaged or dysfunctional proteins and organelles through the lysosomal pathway to achieve proper cellular homeostasis. Autophagy possesses distinct characteristics and is given recognized functions in numerous physiological and pathological conditions, such as cancer. Early stage cancer development can be stopped by autophagy. After tumor cells have successfully undergone transformation and progressed to a late stage, the autophagy-mediated system of dynamic degradation and recycling will support cancer cell growth and adaptation to various cellular stress responses while preserving energy homeostasis. In the present study, the dual function that autophagy plays in various oral cancer development contexts and stages, the existing arguments for and against autophagy, and the ways in which autophagy contributes to oral cancer modifications, such as carcinogenesis, drug resistance, invasion, metastasis and self-proliferation, are reviewed. Special attention is paid to the mechanisms and functions of autophagy in oral cancer processes, and the most recent findings on the application of certain conventional drugs or natural compounds as novel agents that modulate autophagy in oral cancer are discussed. Overall, further research is needed to determine the validity and reliability of autophagy promotion and inhibition while maximizing the difficult challenge of increasing cancer suppression to improve clinical outcomes.
Collapse
Affiliation(s)
- Zhou Li
- Department of Stomatology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030000, P.R. China
- Shanxi Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030000, P.R. China
| | - Yao Zhang
- Shanxi Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030000, P.R. China
| | - Jianhua Lei
- Department of Stomatology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030000, P.R. China
| | - Yunxia Wu
- Department of Stomatology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030000, P.R. China
| |
Collapse
|
5
|
Zhao J, Lei L, Dai W, Jiang A, Jin Q, Tang Z. Simultaneous inhibition of heat shock proteins and autophagy enhances radiofrequency ablation of hepatocellular carcinoma. Biomater Sci 2024; 12:6082-6098. [PMID: 39429155 DOI: 10.1039/d4bm01190b] [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: 10/22/2024]
Abstract
Radiofrequency ablation (RFA) is a commonly used minimally invasive treatment for hepatocellular carcinoma (HCC). However, incomplete radiofrequency ablation (iRFA) promotes tumor progression and metastasis. There is an urgent need to develop innovative strategies to enhance the efficacy of iRFA. The upregulation of heat shock proteins (HSPs) and activation of protective autophagy in tumor cells upon exposure to sublethal heat enhance the thermotolerance, thereby promoting tumor cell survival. Here, 3-methyladenine (3-MA) and lonidamine (LND) co-encapsulated liposomes (Lip@LND/3-MA) are designed to enhance the efficacy of iRFA by simultaneous inhibition of glycolysis and autophagy. On one hand, LND inhibits hexokinase, a key enzyme in glycolysis, and thus reduces ATP production and consequently suppresses the expression of HSPs. On the other hand, 3-MA, as an autophagy inhibitor, can inhibit protective autophagy after iRFA. Lip@LND/3-MA is confirmed to suppress the expression of HSPs and reduce the autophagy level during RFA. Therefore, the thermotolerance of tumor cells is significantly weakened, leading to remarkably enhanced therapeutic efficacy of iRFA. It is believed that simultaneous inhibition of HSPs and autophagy is a promising therapeutic strategy in clinical practice of RFA.
Collapse
Affiliation(s)
- Jinchao Zhao
- Department of General Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China.
| | - Lei Lei
- Department of General Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China.
| | - Wenbin Dai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China.
| | - Angfeng Jiang
- Department of General Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China.
| | - Qiao Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China.
| | - Zhe Tang
- Department of General Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China.
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| |
Collapse
|
6
|
Zhang W, Chen S, Bai Z, Gan M, Chen M, Zhang Y, Liu S, Liu D. Photodynamic Therapy for Oral Squamous Cell Carcinoma: Current Status, Challenges, and Prospects. Int J Nanomedicine 2024; 19:10699-10710. [PMID: 39464676 PMCID: PMC11512526 DOI: 10.2147/ijn.s481901] [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: 06/07/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most prevalent and deadly malignancy of the head and neck. The standard treatments for OSCC are surgery, radiotherapy, and chemoradiotherapy, which can cause severe cosmetic and functional damage to the oral cavity and impair the patients' quality of life. Photodynamic therapy (PDT) is a promising alternative that uses light-activated photosensitizers to induce selective phototoxicity and necrosis in the target tissues. PDT has several advantages over conventional treatments, such as minimal invasion, low side effects, high selectivity and preservation of the oral function and appearance. This review explores the principles, mechanisms, and current applications of PDT for OSCC. We address the challenges, such as the depth of light penetration and tissue hypoxia, and underscore the progressive innovations in photosensitizer enhancement, nanotechnological integration, and precision therapy. The exploration of biomarkers for refining patient selection and tailoring individualized treatment regimens is also undertaken. PDT holds promise as a secure and efficacious modality for OSCC management. Nonetheless, additional investigation is imperative to refine treatment protocols and validate sustained therapeutic success.
Collapse
Affiliation(s)
- Weiqian Zhang
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, People’s Republic of China
| | - Shuiying Chen
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, People’s Republic of China
| | - Zhongyu Bai
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, People’s Republic of China
| | - Menglai Gan
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, People’s Republic of China
| | - Mingwei Chen
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, People’s Republic of China
| | - Ying Zhang
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, People’s Republic of China
| | - Sai Liu
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, People’s Republic of China
| | - Dongjuan Liu
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, People’s Republic of China
| |
Collapse
|
7
|
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
|
8
|
Wang Y, Chang L, Gao H, Yu C, Gao Y, Peng Q. Nanomaterials-based advanced systems for photothermal / photodynamic therapy of oral cancer. Eur J Med Chem 2024; 272:116508. [PMID: 38761583 DOI: 10.1016/j.ejmech.2024.116508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024]
Abstract
The traditional clinical approaches for oral cancer consist of surgery, chemotherapy, radiotherapy, immunotherapy, and so on. However, these treatments often induce side effects and exhibit limited efficacy. Photothermal therapy (PTT) emerges as a promising adjuvant treatment, utilizing photothermal agents (PTAs) to convert light energy into heat for tumor ablation. Another innovative approach, photodynamic therapy (PDT), leverages photosensitizers (PSs) and specific wavelength laser irradiation to generate reactive oxygen species (ROS), offering an effective and non-toxic alternative. The relevant combination therapies have been reported in the field of oral cancer. Simultaneously, the advancement of nanomaterials has propelled the clinical application of PTT and PDT. Therefore, a comprehensive understanding of PTT and PDT is required for better application in oral cancer treatment. Here, we review the use of PTT and PDT in oral cancer, including noble metal materials (e.g., Au nanoparticles), carbon materials (e.g., graphene oxide), organic dye molecules (e.g., indocyanine green), organic molecule-based agents (e.g., porphyrin-analog phthalocyanine) and other inorganic materials (e.g., MXenes), exemplify the advantages and disadvantages of common PTAs and PSs, and summarize the combination therapies of PTT with PDT, PTT/PDT with chemotherapy, PTT with radiotherapy, PTT/PDT with immunotherapy, and PTT/PDT with gene therapy in the treatment of oral cancer. The challenges related to the PTT/PDT combination therapy and potential solutions are also discussed.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Lili Chang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hongyu Gao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chenhao Yu
- Department of Periodontology, National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, China
| | - Yujie Gao
- Department of Stomatology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, 610500, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
9
|
Fan K, Zhang L, Zhong Q, Xiang Y, Xu B, Wang Y. Acceptor-donor-acceptor-type molecules with large electrostatic potential difference for effective NIR photothermal therapy. J Mater Chem B 2024; 12:5140-5149. [PMID: 38712564 DOI: 10.1039/d4tb00187g] [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: 05/08/2024]
Abstract
Although acceptor-donor-acceptor (A-D-A)-type molecules offer advantages in constructing NIR absorbing photothermal agents (PTAs) due to their strong intramolecular charge transfer and molecular planarity, their applications in photothermal therapy (PTT) of tumors remain insufficiently explored. In particular, the influence of ESP distribution on the optical properties of A-D-A photosensitizers has not been investigated. Herein, we analyze and compare the difference in ESP distribution between A-D-A-type small molecules and polymers to construct NIR absorbing PTAs with a high extinction coefficient (ε) and high photothermal conversion efficiency (PCE). The calculation results of density functional theory (DFT) indicate that the large ESP difference makes A-D-A-type small molecules superior to their polymer counterparts in realizing tight molecular packing and strong NIR absorbance. Among the as-prepared nanoparticles (NPs), Y6 NPs exhibited an obvious bathochromic shift of absorption peak from 711 nm to 822 nm, with the NIR-II emission extended to 1400 nm. Moreover, a high ε value of 5.69 L g-1 cm-1 and a PCE of 66.3% were attained, making Y6 NPs suitable for PTT. With a concentration of 100 μg mL-1, Y6 NPs in aqueous dispersion yielded a death rate of 93.4% for 4T1 cells upon 808 nm laser irradiation (1 W cm-2) for 10 min, which is comparable with the best results of recently reported PTT agents.
Collapse
Affiliation(s)
- Kexin Fan
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering Beijing University of Chemical Technology, Beijing 100029, China.
| | - Ludan Zhang
- National Engineering Laboratory for Digital and Material Technology of Stomatology Center of Digital Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Qinqiu Zhong
- National Engineering Laboratory for Digital and Material Technology of Stomatology Center of Digital Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Yanhe Xiang
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering Beijing University of Chemical Technology, Beijing 100029, China.
| | - Bowei Xu
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering Beijing University of Chemical Technology, Beijing 100029, China.
| | - Yuguang Wang
- National Engineering Laboratory for Digital and Material Technology of Stomatology Center of Digital Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| |
Collapse
|
10
|
Xie W, Lu J, Chen Y, Wang X, Lu H, Li Q, Jin N, He J, Ou L, Ni J, Shen Y, Shao L. TCL1A-expressing B cells are critical for tertiary lymphoid structure formation and the prognosis of oral squamous cell carcinoma. J Transl Med 2024; 22:477. [PMID: 38764038 PMCID: PMC11103841 DOI: 10.1186/s12967-024-05292-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/11/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is a malignant tumor with a poor prognosis. Traditional treatments have limited effectiveness. Regulation of the immune response represents a promising new approach for OSCC treatment. B cells are among the most abundant immune cells in OSCC. However, the role of B cells in OSCC treatment has not been fully elucidated. METHODS Single-cell RNA sequencing analysis of 13 tissues and 8 adjacent normal tissues from OSCC patients was performed to explore differences in B-cell gene expression between OSCC tissues and normal tissues. We further investigated the relationship between differentially expressed genes and the immune response to OSCC. We utilized tissue microarray data for 146 OSCC clinical samples and RNA sequencing data of 359 OSCC samples from The Cancer Genome Atlas (TCGA) to investigate the role of T-cell leukemia 1 A (TCL1A) in OSCC prognosis. Multiplex immunohistochemistry (mIHC) was employed to investigate the spatial distribution of TCL1A in OSCC tissues. We then investigated the effect of TCL1A on B-cell proliferation and trogocytosis. Finally, lentiviral transduction was performed to induce TCL1A overexpression in B lymphoblastoid cell lines (BLCLs) to verify the function of TCL1A. RESULTS Our findings revealed that TCL1A was predominantly expressed in B cells and was associated with a better prognosis in OSCC patients. Additionally, we found that TCL1A-expressing B cells are located at the periphery of lymphatic follicles and are associated with tertiary lymphoid structures (TLS) formation in OSCC. Mechanistically, upregulation of TCL1A promoted the trogocytosis of B cells on dendritic cells by mediating the upregulation of CR2, thereby improving antigen-presenting ability. Moreover, the upregulation of TCL1A expression promoted the proliferation of B cells. CONCLUSION This study revealed the role of B-cell TCL1A expression in TLS formation and its effect on OSCC prognosis. These findings highlight TCL1A as a novel target for OSCC immunotherapy.
Collapse
Affiliation(s)
- Wenqiang Xie
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Jinjin Lu
- Department of Periodontics, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, PR China
| | - Yichen Chen
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Xi Wang
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, 510055, PR China
| | - Huanzi Lu
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, 510055, PR China
| | - Qunxing Li
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Nianqiang Jin
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Jiankang He
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Lingling Ou
- Department of Periodontics, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, PR China
| | - Jia Ni
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Yuqin Shen
- Department of Periodontics, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, PR China
| | - Longquan Shao
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China.
| |
Collapse
|
11
|
Zou J, Zhang Y, Pan Y, Mao Z, Chen X. Advancing nanotechnology for neoantigen-based cancer theranostics. Chem Soc Rev 2024; 53:3224-3252. [PMID: 38379286 DOI: 10.1039/d3cs00162h] [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: 02/22/2024]
Abstract
Neoantigens play a pivotal role in the field of tumour therapy, encompassing the stimulation of anti-tumour immune response and the enhancement of tumour targeting capability. Nonetheless, numerous factors directly influence the effectiveness of neoantigens in bolstering anti-tumour immune responses, including neoantigen quantity and specificity, uptake rates by antigen-presenting cells (APCs), residence duration within the tumour microenvironment (TME), and their ability to facilitate the maturation of APCs for immune response activation. Nanotechnology assumes a significant role in several aspects, including facilitating neoantigen release, promoting neoantigen delivery to antigen-presenting cells, augmenting neoantigen uptake by dendritic cells, shielding neoantigens from protease degradation, and optimizing interactions between neoantigens and the immune system. Consequently, the development of nanotechnology synergistically enhances the efficacy of neoantigens in cancer theranostics. In this review, we provide an overview of neoantigen sources, the mechanisms of neoantigen-induced immune responses, and the evolution of precision neoantigen-based nanomedicine. This encompasses various therapeutic modalities, such as neoantigen-based immunotherapy, phototherapy, radiotherapy, chemotherapy, chemodynamic therapy, and other strategies tailored to augment precision in cancer therapeutics. We also discuss the current challenges and prospects in the application of neoantigen-based precision nanomedicine, aiming to expedite its clinical translation.
Collapse
Affiliation(s)
- Jianhua Zou
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yu Zhang
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yuanbo Pan
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, P. R. China.
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumour of Zhejiang Province, Hangzhou, Zhejiang 310009, P. R. China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| |
Collapse
|
12
|
Lu Y, Fan L, Wang J, Hu M, Wei B, Shi P, Li J, Feng J, Zheng Y. Cancer Cell Membrane-Based Materials for Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306540. [PMID: 37814370 DOI: 10.1002/smll.202306540] [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: 08/01/2023] [Revised: 09/18/2023] [Indexed: 10/11/2023]
Abstract
The nanodelivery system provides a novel direction for disease diagnosis and treatment; however, its delivery effectiveness is restricted by the short biological half-life and inadequate tumor targeting. The immune evasion properties and homologous targeting capabilities of natural cell membranes, particularly those of cancer cell membranes (CCM), have gained significant interest. The integration of CCM and nanoparticles has resulted in the emergence of CCM-based nanoplatforms (CCM-NPs), which have gained significant attention due to their unique properties. CCM-NPs not only prolong the blood circulation time of core nanoparticles, but also direct them for homologous tumor targeting. Herein, the history and development of CCM-NPs as well as how these platforms have been used for biomedical applications are discussed. The application of CCM-NPs for cancer therapy will be described in detail. Translational efforts are currently under way and further research to address key areas of need will ultimately be required to facilitate the successful clinical adoption of CCM-NPs.
Collapse
Affiliation(s)
- Yongping Lu
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
- Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Linming Fan
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Jun Wang
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Mingxiang Hu
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Baogang Wei
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Ping Shi
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| | - Jinyan Feng
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Yu Zheng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
13
|
Xu Y, Yang L, Wang C, Sun W, Zheng Y, Ou B, Wu L, Shi L, Lin X, Chen W. Ferroptosis boosted oral cancer photodynamic therapy by carrier-free Sorafenib-Ce6 self-assembly nanoparticles. J Control Release 2024; 366:798-811. [PMID: 38184236 DOI: 10.1016/j.jconrel.2023.12.056] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/19/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
Oral cancer is a disease with high morbidity and mortality worldwide and greatly impacts the quality of life, especially in patients with advanced stages. Photodynamic therapy (PDT) is one of the most effective clinical treatments for oral cancers. However, most clinically applied photosensitizers have several deficiencies, including oxygen dependence, poor aqueous solubility, and a lack of tumor-targeting ability. Herein, the carrier-free multifunctional Sorafenib (Sor), chlorin e6 (Ce6), and Fe3+ self-assembly co-delivery nanoparticles (Sor-Ce6 NPs) were constructed via combining a ferroptosis inducer Sor and a photosensitizer Ce6 for synergetic therapy. The as-synthesized Sor-Ce6 NPs presented excellent colloidal stability and water dispersity with good in vivo tumor-targeting ability. More significantly, the low dose of Sor-Ce6 NPs had little dark toxicity but produced significantly enhanced ROS and supplied O2 sustainably to increase phototoxicity through ferroptosis pathway. Notably, the Sor-Ce6 NPs showed significantly higher in vitro and in vivo anti-tumor efficacy than the Sor/Ce6 mixture due to the improvement of cellular uptake and the incorporation of foreign Fe ions in the system, which also confer the T1 magnetic resonance-guided imaging ability to the formed Sor-Ce6 NPs. Our study demonstrates a promising self-assembled strategy for overcoming hypoxia-related PDT resistance for oral cancer treatment.
Collapse
Affiliation(s)
- Yingying Xu
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Liu Yang
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Chengyan Wang
- Laboratory Animal Center, Fujian Medical University, Fuzhou 350122, China
| | - Weiming Sun
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Yijing Zheng
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Beiwei Ou
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Lixian Wu
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Leilei Shi
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Xi Lin
- Public Technology Service Center, Fujian Medical University, Fuzhou 350122, China.
| | - Wei Chen
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
14
|
Farzipour S, Zefrei FJ, Bahadorikhalili S, Alvandi M, Salari A, Shaghaghi Z. Nanotechnology Utilizing Ferroptosis Inducers in Cancer Treatment. Anticancer Agents Med Chem 2024; 24:571-589. [PMID: 38275050 DOI: 10.2174/0118715206278427231215111526] [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: 10/06/2023] [Revised: 11/11/2023] [Accepted: 11/20/2023] [Indexed: 01/27/2024]
Abstract
Current cancer treatment options have presented numerous challenges in terms of reaching high efficacy. As a result, an immediate step must be taken to create novel therapies that can achieve more than satisfying outcomes in the fight against tumors. Ferroptosis, an emerging form of regulated cell death (RCD) that is reliant on iron and reactive oxygen species, has garnered significant attention in the field of cancer therapy. Ferroptosis has been reported to be induced by a variety of small molecule compounds known as ferroptosis inducers (FINs), as well as several licensed chemotherapy medicines. These compounds' low solubility, systemic toxicity, and limited capacity to target tumors are some of the significant limitations that have hindered their clinical effectiveness. A novel cancer therapy paradigm has been created by the hypothesis that ferroptosis induced by nanoparticles has superior preclinical properties to that induced by small drugs and can overcome apoptosis resistance. Knowing the different ideas behind the preparation of nanomaterials that target ferroptosis can be very helpful in generating new ideas. Simultaneously, more improvement in nanomaterial design is needed to make them appropriate for therapeutic treatment. This paper first discusses the fundamentals of nanomedicine-based ferroptosis to highlight the potential and characteristics of ferroptosis in the context of cancer treatment. The latest study on nanomedicine applications for ferroptosis-based anticancer therapy is then highlighted.
Collapse
Affiliation(s)
- Soghra Farzipour
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Jalali Zefrei
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Saeed Bahadorikhalili
- Department of Electronic Engineering, Universitat Rovira i Virgili, 43007, Tarragona, Spain
| | - Maryam Alvandi
- Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Nuclear Medicine and Molecular Imaging, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Arsalan Salari
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Shaghaghi
- Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
15
|
Zhang Y, Wu Y, Du H, Li Z, Bai X, Wu Y, Li H, Zhou M, Cao Y, Chen X. Nano-Drug Delivery Systems in Oral Cancer Therapy: Recent Developments and Prospective. Pharmaceutics 2023; 16:7. [PMID: 38276483 PMCID: PMC10820767 DOI: 10.3390/pharmaceutics16010007] [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: 10/10/2023] [Revised: 11/16/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Oral cancer (OC), characterized by malignant tumors in the mouth, is one of the most prevalent malignancies worldwide. Chemotherapy is a commonly used treatment for OC; however, it often leads to severe side effects on human bodies. In recent years, nanotechnology has emerged as a promising solution for managing OC using nanomaterials and nanoparticles (NPs). Nano-drug delivery systems (nano-DDSs) that employ various NPs as nanocarriers have been extensively developed to enhance current OC therapies by achieving controlled drug release and targeted drug delivery. Through searching and analyzing relevant research literature, it was found that certain nano-DDSs can improve the therapeutic effect of drugs by enhancing drug accumulation in tumor tissues. Furthermore, they can achieve targeted delivery and controlled release of drugs through adjustments in particle size, surface functionalization, and drug encapsulation technology of nano-DDSs. The application of nano-DDSs provides a new tool and strategy for OC therapy, offering personalized treatment options for OC patients by enhancing drug delivery, reducing toxic side effects, and improving therapeutic outcomes. However, the use of nano-DDSs in OC therapy still faces challenges such as toxicity, precise targeting, biodegradability, and satisfying drug-release kinetics. Overall, this review evaluates the potential and limitations of different nano-DDSs in OC therapy, focusing on their components, mechanisms of action, and laboratory therapeutic effects, aiming to provide insights into understanding, designing, and developing more effective and safer nano-DDSs. Future studies should focus on addressing these issues to further advance the application and development of nano-DDSs in OC therapy.
Collapse
Affiliation(s)
- Yun Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China; (Y.Z.); (Y.W.); (Z.L.); (X.B.); (Y.W.); (H.L.); (M.Z.)
| | - Yongjia Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China; (Y.Z.); (Y.W.); (Z.L.); (X.B.); (Y.W.); (H.L.); (M.Z.)
| | - Hongjiang Du
- Department of Stomatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, China;
| | - Zhiyong Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China; (Y.Z.); (Y.W.); (Z.L.); (X.B.); (Y.W.); (H.L.); (M.Z.)
| | - Xiaofeng Bai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China; (Y.Z.); (Y.W.); (Z.L.); (X.B.); (Y.W.); (H.L.); (M.Z.)
| | - Yange Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China; (Y.Z.); (Y.W.); (Z.L.); (X.B.); (Y.W.); (H.L.); (M.Z.)
| | - Huimin Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China; (Y.Z.); (Y.W.); (Z.L.); (X.B.); (Y.W.); (H.L.); (M.Z.)
| | - Mengqi Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China; (Y.Z.); (Y.W.); (Z.L.); (X.B.); (Y.W.); (H.L.); (M.Z.)
| | - Yifeng Cao
- Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Xuepeng Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China; (Y.Z.); (Y.W.); (Z.L.); (X.B.); (Y.W.); (H.L.); (M.Z.)
| |
Collapse
|
16
|
Bai Y, Yu EY, Liu Y, Jin H, Liu X, Wu X, Zhang M, Feng N, Huang P, Zhang H, Kwok RTK, Xia X, Li Y, Tang BZ, Wang H. Molecular Engineering of AIE Photosensitizers for Inactivation of Rabies Virus. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303542. [PMID: 37431212 DOI: 10.1002/smll.202303542] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/30/2023] [Indexed: 07/12/2023]
Abstract
Rabies is a zoonotic neurological disease caused by the rabies virus (RABV) that is fatal to humans and animals. While several post-infection treatment have been suggested, developing more efficient and innovative antiviral methods are necessary due to the limitations of current therapeutic approaches. To address this challenge, a strategy combining photodynamic therapy and immunotherapy, using a photosensitizer (TPA-Py-PhMe) with high type I and type II reactive oxygen species (ROS) generation ability is proposed. This approach can inactivate the RABV by killing the virus directly and activating the immune response. At the cellular level, TPA-Py-PhMe can reduce the virus titer under preinfection prophylaxis and postinfection treatment, with its antiviral effect mainly dependent on ROS and pro-inflammatory factors. Intriguingly, when mice are injected with TPA-Py-PhMe and exposed to white light irradiation at three days post-infection, the onset of disease is delayed, and survival rates improved to some extent. Overall, this study shows that photodynamic therapy and immunotherapy open new avenues for future antiviral research.
Collapse
Affiliation(s)
- Yujie Bai
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Eric Y Yu
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, 999077, China
| | - Yongsai Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Hongli Jin
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xingqi Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xiaoyu Wu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Mengyao Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Na Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Pei Huang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Haili Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Ryan T K Kwok
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, 999077, China
| | - Xianzhu Xia
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Yuanyuan Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Ben Zhong Tang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Shenzhen, Guangdong, 518172, China
| | - Hualei Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| |
Collapse
|
17
|
Cui S, Liu H, Cui G. Nanoparticles as drug delivery systems in the treatment of oral squamous cell carcinoma: current status and recent progression. Front Pharmacol 2023; 14:1176422. [PMID: 37292147 PMCID: PMC10244499 DOI: 10.3389/fphar.2023.1176422] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/15/2023] [Indexed: 06/10/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a common human malignancy with an estimated incidence of around 377,713 new cases worldwide in 2020. Despite the advance in clinical management, some of OSCC patients still miss the opportunity of completable resection of tumor, and have to accept medical therapies, e.g., chemotherapy, radiotherapy, or immunotherapy when the disease develops into the advanced stage. However, these therapies have been reported to be far from ideal due to the low efficiency of conventional delivery approaches. To obtain a better therapeutic effect, considerable attempts have been made toward to develop an effective drug delivery system (DDS). Nanoparticles (NPs) including inorganic NPs, polymer NPs, lipid NP, extracellular vesicles and cell membrane-based NPs have been evaluated as the better DDS candidates that can specifically accumulate in the tumor microenvironment along with a large amount of blood vessels. Emerging evidence suggested that NPs formulated with anticancer drugs including chemotherapeutic drugs, radiotherapy and immunotarget antibodies could remarkably improve the release and increase concentration of these drugs at the tumor site and show a better therapeutic efficacy, suggesting that NPs might serve as promising DDSs in the treatment of OSCC. Therefore, we have conducted this review to summarize recent progression and current status of diverse NPs as DDSs in this research field.
Collapse
Affiliation(s)
- Shuxia Cui
- Henan Stomatological Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hanzhe Liu
- School of Stomatology, Wuhan University, Wuhan, China
| | - Guanglin Cui
- Faculty of Health Science, Campus Levanger, Nord University, Levanger, Norway
| |
Collapse
|
18
|
Elmehrath S, Nguyen HL, Karam SM, Amin A, Greish YE. BioMOF-Based Anti-Cancer Drug Delivery Systems. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:953. [PMID: 36903831 PMCID: PMC10005089 DOI: 10.3390/nano13050953] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/19/2023] [Accepted: 03/03/2023] [Indexed: 06/18/2023]
Abstract
A variety of nanomaterials have been developed specifically for biomedical applications, such as drug delivery in cancer treatment. These materials involve both synthetic and natural nanoparticles and nanofibers of varying dimensions. The efficacy of a drug delivery system (DDS) depends on its biocompatibility, intrinsic high surface area, high interconnected porosity, and chemical functionality. Recent advances in metal-organic framework (MOF) nanostructures have led to the achievement of these desirable features. MOFs consist of metal ions and organic linkers that are assembled in different geometries and can be produced in 0, 1, 2, or 3 dimensions. The defining features of MOFs are their outstanding surface area, interconnected porosity, and variable chemical functionality, which enable an endless range of modalities for loading drugs into their hierarchical structures. MOFs, coupled with biocompatibility requisites, are now regarded as highly successful DDSs for the treatment of diverse diseases. This review aims to present the development and applications of DDSs based on chemically-functionalized MOF nanostructures in the context of cancer treatment. A concise overview of the structure, synthesis, and mode of action of MOF-DDS is provided.
Collapse
Affiliation(s)
- Sandy Elmehrath
- Department of Chemistry, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Ha L. Nguyen
- Department of Chemistry University of California—Berkeley, Kavli Energy Nanoscience Institute at UC Berkeley, and Berkeley Global Science Institute, Berkeley, CA 94720, USA
- Joint UAEU−UC Berkeley Laboratories for Materials Innovations, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Sherif M. Karam
- Department of Anatomy, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
- Zayed Centre for Health Sciences, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Amr Amin
- Zayed Centre for Health Sciences, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
- Department of Biology, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Yaser E. Greish
- Department of Chemistry, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
- Joint UAEU−UC Berkeley Laboratories for Materials Innovations, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
- Zayed Centre for Health Sciences, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| |
Collapse
|
19
|
Lin Q, Peng Y, Wen Y, Li X, Du D, Dai W, Tian W, Meng Y. Recent progress in cancer cell membrane-based nanoparticles for biomedical applications. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2023; 14:262-279. [PMID: 36895440 PMCID: PMC9989677 DOI: 10.3762/bjnano.14.24] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
Immune clearance and insufficient targeting have limited the efficacy of existing therapeutic strategies for cancer. Toxic side effects and individual differences in response to treatment have further limited the benefits of clinical treatment for patients. Biomimetic cancer cell membrane-based nanotechnology has provided a new approach for biomedicine to overcome these obstacles. Biomimetic nanoparticles exhibit various effects (e.g., homotypic targeting, prolonging drug circulation, regulating the immune system, and penetrating biological barriers) after encapsulation by cancer cell membranes. The sensitivity and specificity of diagnostic methods will also be improved by utilizing the properties of cancer cell membranes. In this review, different properties and functions of cancer cell membranes are presented. Utilizing these advantages, nanoparticles can exhibit unique therapeutic capabilities in various types of diseases, such as solid tumors, hematological malignancies, immune system diseases, and cardiovascular diseases. Furthermore, cancer cell membrane-encapsulated nanoparticles show improved effectiveness and efficiency in combination with current diagnostic and therapeutic methods, which will contribute to the development of individualized treatments. This strategy has promising clinical translation prospects, and the associated challenges are discussed.
Collapse
Affiliation(s)
- Qixiong Lin
- The Ninth Clinical Medical School of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Yueyou Peng
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Yanyan Wen
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiaoqiong Li
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Donglian Du
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Weibin Dai
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Wei Tian
- Department of General Surgery, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi 030024, China
| | - Yanfeng Meng
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| |
Collapse
|
20
|
Tao Y, Liu Y, Dong Z, Chen X, Wang Y, Li T, Li J, Zang S, He X, Chen D, Zhao Z, Li M. Cellular Hypoxia Mitigation by Dandelion-like Nanoparticles for Synergistic Photodynamic Therapy of Oral Squamous Cell Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2022; 14:44039-44053. [PMID: 36153957 DOI: 10.1021/acsami.2c10021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Hypoxia at the tumor site limits the therapeutic effects of photodynamic therapy (PDT) in oral squamous cell carcinoma (OSCC), which is an oxygen-consumption process. Inhibiting cellular oxygen consumption and reducing cellular ATP production are expected to enhance PDT. In this study, we designed and constructed dandelion-like size-shrinkable nanoparticles for tumor-targeted delivery of hypoxia regulator resveratrol (RES) and photodynamic agent chlorine e6 (CE6). Both drugs were co-encapsulated in small-sized micelles modified with EGFR targeting ligand GE11, which was further conjugated on hyaluronic nanogel (NG) to afford RC-GMN. After targeted accumulation in tumors mediated by GE11 and enhanced penetration and retention (EPR) effects, RC-GMN was degraded by hyaluronidase (HAase) and resulted in small-sized micelles, allowing for deep penetration and dual-receptor-mediated cellular internalization. Resveratrol inhibited cellular oxygen consumption and provided sufficient oxygen for PDT, which consequently activated PDT to produce reactive oxygen species (ROS). Notably, we found that autophagy was overactivated in PDT, which was further strengthened by the hypoxia regulator resveratrol, elevating autophagic cell death. The synergistic effects of resveratrol and CE6 promoted autophagic cell death and apoptosis in the enhanced PDT, resulting in stronger antitumor effects in the orthotopic OSCC model. Therefore, the facilitated delivery of hypoxia regulator enhanced PDT efficacy by elevating oxygen content in tumor cells and inducing autophagic cell death and apoptosis, which offers an alternative strategy for enhancing the PDT effects against OSCC.
Collapse
Affiliation(s)
- Yuan Tao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yingke Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, People's Republic of China
| | - Ziyan Dong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xiaoxiao Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yashi Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Ting Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jiaxin Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Shuya Zang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xuan He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Dong Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, People's Republic of China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|