1
|
Cai Y, Chai T, Nguyen W, Liu J, Xiao E, Ran X, Ran Y, Du D, Chen W, Chen X. Phototherapy in cancer treatment: strategies and challenges. Signal Transduct Target Ther 2025; 10:115. [PMID: 40169560 PMCID: PMC11961771 DOI: 10.1038/s41392-025-02140-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/11/2024] [Accepted: 01/13/2025] [Indexed: 04/03/2025] Open
Abstract
Phototherapy has emerged as a promising modality in cancer treatment, garnering considerable attention for its minimal side effects, exceptional spatial selectivity, and optimal preservation of normal tissue function. This innovative approach primarily encompasses three distinct paradigms: Photodynamic Therapy (PDT), Photothermal Therapy (PTT), and Photoimmunotherapy (PIT). Each of these modalities exerts its antitumor effects through unique mechanisms-specifically, the generation of reactive oxygen species (ROS), heat, and immune responses, respectively. However, significant challenges impede the advancement and clinical application of phototherapy. These include inadequate ROS production rates, subpar photothermal conversion efficiency, difficulties in tumor targeting, and unfavorable physicochemical properties inherent to traditional phototherapeutic agents (PTs). Additionally, the hypoxic microenvironment typical of tumors complicates therapeutic efficacy due to limited agent penetration in deep-seated lesions. To address these limitations, ongoing research is fervently exploring innovative solutions. The unique advantages offered by nano-PTs and nanocarrier systems aim to enhance traditional approaches' effectiveness. Strategies such as generating oxygen in situ within tumors or inhibiting mitochondrial respiration while targeting the HIF-1α pathway may alleviate tumor hypoxia. Moreover, utilizing self-luminescent materials, near-infrared excitation sources, non-photoactivated sensitizers, and wireless light delivery systems can improve light penetration. Furthermore, integrating immunoadjuvants and modulating immunosuppressive cell populations while deploying immune checkpoint inhibitors holds promise for enhancing immunogenic cell death through PIT. This review seeks to elucidate the fundamental principles and biological implications of phototherapy while discussing dominant mechanisms and advanced strategies designed to overcome existing challenges-ultimately illuminating pathways for future research aimed at amplifying this intervention's therapeutic efficacy.
Collapse
Affiliation(s)
- Yeyu Cai
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Tian Chai
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, Shanxi Province, China
| | - William Nguyen
- School of Chips, XJTLU Entrepreneur College (Taicang), Xi'an Jiaotong-Liverpool University, Taicang, Suzhou, China
| | - Jiayi Liu
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Enhua Xiao
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Xin Ran
- Department of Dermatovenereology, The West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuping Ran
- Department of Dermatovenereology, The West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Dan Du
- Department of Dermatovenereology, The West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Chen
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, Shanxi Province, China.
| | - Xiangyu Chen
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China.
| |
Collapse
|
2
|
Han J, Xu X, Jin F, Xu X, Fang T, Du Y. Tumor oxygenation nanoliposomes promote deep photodynamic therapy for triple-negative breast cancer. Biomater Sci 2024; 12:4967-4979. [PMID: 39158634 DOI: 10.1039/d4bm00847b] [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: 08/20/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive type of breast cancer and has many characteristics including high metastatic rates, poor overall survival, and low response to traditional chemotherapy. Photodynamic therapy (PDT), emerging as a precise treatment modality, has shown promise in improving the antitumor response. However, it still faces challenges such as limited light penetration depth, rapid oxygen consumption, and inadequate targeting ability. In this study, we developed Rose Bengal (RB, photosensitizer) and oxygen co-loaded CREKA-modified UCNP-based nanoliposomes (CLIP-RB-PFOB@UCNP) for tumor targeting and near-infrared (NIR)-triggered deep and long-lasting PDT. Our results demonstrated that CLIP-RB-PFOB@UCNP effectively targeted and accumulated in tumor tissue through the interaction between CREKA and fibronectin, which is overexpressed in tumor cells. Under NIR irradiation, CLIP-RB-PFOB@UCNP exhibited significant destruction of orthotopic tumors, reduced the level of HIF-1α, and efficiently suppressed lung metastasis in a metastatic TNBC model. In conclusion, this study offers new avenues for improving the therapeutic outcomes of PDT for clinical TNBC treatment.
Collapse
Affiliation(s)
- Jianhua Han
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China.
| | - Xinyi Xu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China.
| | - Feiyang Jin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China.
| | - Xiaoling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang 310015, P. R. China.
| | - Tao Fang
- Department of Anesthesiology, The Affiliated Jinhua Hospital of Wenzhou Medical University, Jinhua, Zhejiang 321000, P. R. China.
| | - Yongzhong Du
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China.
- Innovation Center of Translational Pharmacy, Jinhua Institute of Zhejiang University, Jinhua 321299, P. R. China
| |
Collapse
|
3
|
Kah G, Chandran R, Abrahamse H. Biogenic Silver Nanoparticles for Targeted Cancer Therapy and Enhancing Photodynamic Therapy. Cells 2023; 12:2012. [PMID: 37566091 PMCID: PMC10417642 DOI: 10.3390/cells12152012] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/01/2023] [Accepted: 08/06/2023] [Indexed: 08/12/2023] Open
Abstract
Different conventional therapeutic procedures are utilized globally to manage cancer cases, yet the mortality rate in patients with cancer remains considerably high. Developments in the field of nanotechnology have included novel therapeutic strategies to deal with cancer. Biogenic (green) metallic silver nanoparticles (AgNPs) obtained using plant-mediated protocols are attractive to researchers exploring cancer treatment. Biogenic AgNPs present advantages, since they are cost-effective, easy to obtain, energy efficient, and less toxic compared to chemically and physically obtained AgNPs. Also, they present excellent anticancer abilities thanks to their unique sizes, shapes, and optical properties. This review provides recent advancements in exploring biogenic AgNPs as a drug or agent for cancer treatment. Thus, great attention was paid to the anticancer efficacy of biogenic AgNPs, their anticancer mechanisms, their efficacy in cancer photodynamic therapy (PDT), their efficacy in targeted cancer therapy, and their toxicity.
Collapse
Affiliation(s)
| | - Rahul Chandran
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg 2028, South Africa; (G.K.); (H.A.)
| | | |
Collapse
|
4
|
Jia D, Liu H, Zheng S, Yuan D, Sun R, Wang F, Li Y, Li H, Yuan F, Fan Q, Zhao Z. ICG-Dimeric Her2-Specific Affibody Conjugates for Tumor Imaging and Photothermal Therapy for Her2-Positive Tumors. Mol Pharm 2023; 20:427-437. [PMID: 36315025 DOI: 10.1021/acs.molpharmaceut.2c00708] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Human epidermal growth factor receptor 2 (Her2) is abundantly expressed in various solid tumors. The Her2-specific Affibody (ZHer2:2891) has been clinically tested in patients with Her2-positive breast cancer and is regarded as an ideal drug carrier for tumor diagnosis and targeted treatment. Indocyanine green (ICG) can be used as a photosensitizer for photothermal therapy (PTT), in addition to fluorescent dyes for tumor imaging. In this study, a dimeric Her2-specific Affibody (ZHer2) based on ZHer2:2891 was prepared using the E. coli expression system and then coupled to ICG through an N-hydroxysuccinimide (NHS) ester reactive group to construct a novel bifunctional protein drug (named ICG-ZHer2) for tumor diagnosis and PTT. In vitro, ICG-ZHer2-mediated PTT selectively and efficiently killed Her2-positive BT-474 and SKOV-3 tumor cells rather than Her2-negative HeLa tumor cells. In vivo, ICG-ZHer2 specifically accumulated in Her2-positive SKOV-3 tumor grafts rather than Her2-negative HeLa tumor grafts; high-contrast tumor optical images were obtained. However, Her2-negative HeLa tumor grafts were not detected. More importantly, ICG-ZHer2-mediated PTT exhibited a significantly enhanced antitumor effect in mice bearing SKOV-3 tumor grafts owing to the good photothermal properties of ICG-ZHer2. Of note, ICG-ZHer2 did not exhibit acute toxicity in mice during short-term treatment. Overall, our findings indicate that ICG-ZHer2 is a promising bifunctional drug for Her2-positive tumor diagnosis and PTT.
Collapse
Affiliation(s)
- Dianlong Jia
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Huimin Liu
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Shuhui Zheng
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Dandan Yuan
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Ruohan Sun
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Fei Wang
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Yang Li
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Hui Li
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Fengjiao Yuan
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Qing Fan
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Zhenxiong Zhao
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang 317700, China
| |
Collapse
|
5
|
Alavi N, Maghami P, Pakdel AF, Rezaei M, Avan A. Antibody-modified Gold Nanobiostructures: Advancing Targeted Photodynamic Therapy for Improved Cancer Treatment. Curr Pharm Des 2023; 29:3103-3122. [PMID: 37990429 DOI: 10.2174/0113816128265544231102065515] [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: 06/10/2023] [Accepted: 10/03/2023] [Indexed: 11/23/2023]
Abstract
Photodynamic therapy (PDT) is an innovative, non-invasive method of treating cancer that uses light-activated photosensitizers to create reactive oxygen species (ROS). However, challenges associated with the limited penetration depth of light and the need for precise control over photosensitizer activation have hindered its clinical translation. Nanomedicine, particularly gold nanobiostructures, offers promising solutions to overcome these limitations. This paper reviews the advancements in PDT and nanomedicine, focusing on applying antibody-modified gold nanobiostructures as multifunctional platforms for enhanced PDT efficacy and improved cancer treatment outcomes. The size, shape, and composition of gold nanobiostructures can significantly influence their PDT efficacy, making synthetic procedures crucial. Functionalizing the surface of gold nanobiostructures with various molecules, such as antibodies or targeting agents, bonding agents, PDT agents, photothermal therapy (PTT) agents, chemo-agents, immunotherapy agents, and imaging agents, allows composition modification. Integrating gold nanobiostructures with PDT holds immense potential for targeted cancer therapy. Antibody-modified gold nanobiostructures, in particular, have gained significant attention due to their tunable plasmonic characteristics, biocompatibility, and surface functionalization capabilities. These multifunctional nanosystems possess unique properties that enhance the efficacy of PDT, including improved light absorption, targeted delivery, and enhanced ROS generation. Passive and active targeting of gold nanobiostructures can enhance their localization near cancer cells, leading to efficient eradication of tumor tissues upon light irradiation. Future research and clinical studies will continue to explore the potential of gold nanobiostructures in PDT for personalized and effective cancer therapy. The synthesis, functionalization, and characterization of gold nanobiostructures, their interaction with light, and their impact on photosensitizers' photophysical and photochemical properties, are important areas of investigation. Strategies to enhance targeting efficiency and the evaluation of gold nanobiostructures in vitro and in vivo studies will further advance their application in PDT. The integrating antibody-modified gold nanobiostructures in PDT represents a promising strategy for targeted cancer therapy. These multifunctional nanosystems possess unique properties that enhance PDT efficacy, including improved light absorption, targeted delivery, and enhanced ROS generation. Continued research and development in this field will contribute to the advancement of personalized and effective cancer treatment approaches.
Collapse
Affiliation(s)
- Negin Alavi
- Department of Biology, Islamic Azad University Science and Research Branch, Tehran, Iran
| | - Parvaneh Maghami
- Department of Biology, Islamic Azad University Science and Research Branch, Tehran, Iran
| | - Azar Fani Pakdel
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Rezaei
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane 4059, Australia
| |
Collapse
|
6
|
Multifunctional Photoactive Nanomaterials for Photodynamic Therapy against Tumor: Recent Advancements and Perspectives. Pharmaceutics 2022; 15:pharmaceutics15010109. [PMID: 36678738 PMCID: PMC9866498 DOI: 10.3390/pharmaceutics15010109] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
Numerous treatments are available for cancer, including chemotherapy, immunotherapy, radiation therapy, hormone therapy, biomarker testing, surgery, photodynamic therapy, etc. Photodynamic therapy (PDT) is an effective, non-invasive, novel, and clinically approved strategy to treat cancer. In PDT, three main agents are utilized, i.e., photosensitizer (PS) drug, oxygen, and light. At first, the photosensitizer is injected into blood circulation or applied topically, where it quickly becomes absorbed or accumulated at the tumor site passively or actively. Afterward, the tumor is irradiated with light which leads to the activation of the photosensitizing molecule. PS produces the reactive oxygen species (ROS), resulting in the death of the tumor cell. However, the effectiveness of PDT for tumor destruction is mainly dependent on the cellular uptake and water solubility of photosensitizer molecules. Therefore, the delivery of photosensitizer molecules to the tumor cell is essential in PDT against cancer. The non-specific distribution of photosensitizer results in unwanted side effects and unsuccessful therapeutic outcomes. Therefore, to improve PDT clinical outcomes, the current research is mostly focused on developing actively targeted photosensitizer molecules, which provide a high cellular uptake and high absorption capacity to the tumor site by overcoming the problem associated with conventional PDT. Therefore, this review aims to provide current knowledge on various types of actively and passively targeted organic and inorganic nanocarriers for different cancers.
Collapse
|
7
|
Mokoena D, George BP, Abrahamse H. Conjugation of Hypericin to Gold Nanoparticles for Enhancement of Photodynamic Therapy in MCF-7 Breast Cancer Cells. Pharmaceutics 2022; 14:2212. [PMID: 36297648 PMCID: PMC9611363 DOI: 10.3390/pharmaceutics14102212] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 09/28/2023] Open
Abstract
Breast cancer, among the different types of cancer, is one of the most diagnosed cancers and the leading cause of mortalities amongst women. Factors, including genetic and epigenetic alterations in tumors, make it resistant to therapies, which results in treatment failures and/or recurrence. Furthermore, the existing therapies have many unfavorable side effects leading to poor prognosis and reduced therapeutic outcomes. Photodynamic therapy (PDT) is one of the most effective cancer therapies with increased selectivity and specificity toward cancer cells. As a result, the use of gold nanoparticles (AuNP) further improves the effectiveness of PDT by increasing the drug loading capacity into the cells. In this study, hypericin (Hyp) photosensitizer (PS) was adsorbed on gold nanoparticles (AuNPs) by sonication to achieve physical adsorption of the PS to AuNP. The resulting compound was characterized by FTIR, Zeta potential, UV-Vis spectroscopy, and TEM. The compound was used for the PDT treatment of MCF-7 human breast cancer in vitro. Cellular responses at 12 h post-PDT at 10 J/cm2 were observed. Cellular morphology, LDH membrane integrity, ATP luminescence assay, and Annexin V/PI staining were performed. The results demonstrated typical cell death morphological features while the biochemical responses indicated increased LDH and decreased ATP levels. In conclusion, this study presents an insight into the application of advanced PDT in breast cancer cells by inducing cancer cell death in vitro via apoptosis.
Collapse
Affiliation(s)
| | | | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Johannesburg 2028, South Africa
| |
Collapse
|
8
|
Wang J, Yang B, Lv C, Chen T, Sun L, Sun L, Hao J, Ding F, Wang T, Jiang J, Qin Y. Amino porphyrin-peptide assemblies induce ribosome damage and cancer stem cell inhibition for an enhanced photodynamic therapy. Biomaterials 2022; 289:121812. [PMID: 36152516 DOI: 10.1016/j.biomaterials.2022.121812] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022]
Abstract
Cancer stem cells (CSCs) are the subpopulation of tumor cells with the properties of tumorigenesis, multilineage differentiation potential and self-renewal, which is the driving force of tumor recurrence and metastasis. However, targeting CSCs is still the main challenge in cancer therapy due to their rapid growth and fast mutation rate. Herein, we developed a simple strategy of photodynamic therapy (PDT) targeting CSCs, dependent on much more abundant ribosomes in CSCs. The interactions between positively charged nanoparticles with negatively charged nucleic acids architectures in cancer cells could lead ribosomes targeting as well as CSCs targeting. The co-assembly of simple amino porphyrin (m-TAPP) with short peptide (Fmoc-L3-OMe) formed nanoparticles (NPs) with good biocompatibility and photoactivity, became positively charged due to low pH value of tumour microenvironment, and efficiently accessed cancer cell ribosome, approached cancer cell nuclei, therefore enriched in the fast-amplifying CSCs. The inhibitive effect on CSCs by m-TAPP assemblies was verified by the significant reduction of CSCs markers CD44, CD133 and ribosome amount in cancer cells and tissues. Upon light irradiation, the NPs induced ROS generation to provoke destructive cancer cell ribosome damage and subsequent apoptosis to prevent tumor growth markedly. Based on the assemblies of small organic molecules, our study not only achieves ribosome degradation induced cancer cells apoptosis, but also indicates new possibility of performing CSCs targeting PDT.
Collapse
Affiliation(s)
- Jian Wang
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China; Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Baochan Yang
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China; Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Chaofan Lv
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Tiancheng Chen
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Lixin Sun
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China; Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Lei Sun
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Junfeng Hao
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Fang Ding
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China.
| | - Tianyu Wang
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China.
| | - Jianzhuang Jiang
- Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry and Chemical Engineering, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China.
| | - Yan Qin
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China.
| |
Collapse
|
9
|
Ye J, Yi Y, Wang H, Wang G, Sun Y, Liu E, Tao X, He C. A Study of Glutathione-Responsive Dual-Drug-Loaded Nanoparticles in Anti-Osteosarcoma Treatment. J Biomed Nanotechnol 2022. [DOI: 10.1166/jbn.2022.3411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We connected polyglutamic acid and methotrexate (MTX) through disulfide bonds to prepare glutathione-responsive nanoparticles (MTX NPs) and encapsulated doxorubicin (DOX) to obtain dual drug-loaded NPs (DOX/MTX NPs) (Fig. 1). The appearance of the carbonyl stretching vibration peak
at approximately 1640 cm−1 in the results of the infrared spectrum proved the successful synthesis of three kinds of nanoparticles (NPs) with different feeding ratios. The particle sizes of NPs with different feeding ratios were 100–200 nm, and the encapsulation of DOX
slightly increased the size, while the surface charge was always negative. The release of MTX at 10 mM glutathione (GSH) was as high as 91.45%, and that of DOX was 89.44%, suggesting that the breakage of disulfide bonds leads to the disintegration of NPs. The results of the cell experiment
showed that the encapsulation of DOX effectively increased toxicity and side effects in 143B cells and significantly induced cell apoptosis, and the inhibition of the migration rate increased as the feeding ratio increased. In animal experiments, DOX/MTX NPs significantly induced tumor cell
apoptosis and inhibited cell proliferation and tumor growth. The nanoparticles had excellent tumor-targeting properties. Tumor-targeted NPs with the combined action of the two drugs provided a good strategy for the efficient and precise treatment of osteosarcoma.
Collapse
Affiliation(s)
- Jia Ye
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Yangfei Yi
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Hongyi Wang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Guowei Wang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Yuting Sun
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Enze Liu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Xiaojun Tao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Chunlian He
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| |
Collapse
|
10
|
Lebedeva NS, Koifman OI. Supramolecular Systems Based on Macrocyclic Compounds with Proteins: Application Prospects. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1068162022010071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
11
|
Jin ZY, Fatima H, Zhang Y, Shao Z, Chen XJ. Recent Advances in Bio‐Compatible Oxygen Singlet Generation and Its Tumor Treatment. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Zheng Yang Jin
- The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| | - Hira Fatima
- Western Australia School of Mines: Minerals Energy and Chemical Engineering (WASM‐MECE) Curtin University Perth Western Australia 6102 Australia
| | - Yue Zhang
- The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| | - Zongping Shao
- Western Australia School of Mines: Minerals Energy and Chemical Engineering (WASM‐MECE) Curtin University Perth Western Australia 6102 Australia
- State Key Laboratory of Materials‐Oriented Chemical Engineering College of Chemical Engineering Nanjing Tech University Nanjing Jiangsu 211816 P. R. China
| | - Xiang Jian Chen
- The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| |
Collapse
|
12
|
Shang L, Zhou X, Zhang J, Shi Y, Zhong L. Metal Nanoparticles for Photodynamic Therapy: A Potential Treatment for Breast Cancer. Molecules 2021; 26:molecules26216532. [PMID: 34770941 PMCID: PMC8588551 DOI: 10.3390/molecules26216532] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/13/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer (BC) is the most common malignant tumor in women worldwide, which seriously threatens women’s physical and mental health. In recent years, photodynamic therapy (PDT) has shown significant advantages in cancer treatment. PDT involves activating photosensitizers with appropriate wavelengths of light, producing transient levels of reactive oxygen species (ROS). Compared with free photosensitizers, the use of nanoparticles in PDT shows great advantages in terms of solubility, early degradation, and biodistribution, as well as more effective intercellular penetration and targeted cancer cell uptake. Under the current circumstances, researchers have made promising efforts to develop nanocarrier photosensitizers. Reasonably designed photosensitizer (PS) nanoparticles can be achieved through non-covalent (self-aggregation, interfacial deposition, interfacial polymerization or core-shell embedding and physical adsorption) or covalent (chemical immobilization or coupling) processes and accumulate in certain tumors through passive and/or active targeting. These PS loading methods provide chemical and physical stability to the PS payload. Among nanoparticles, metal nanoparticles have the advantages of high stability, adjustable size, optical properties, and easy surface functionalization, making them more biocompatible in biological applications. In this review, we summarize the current development and application status of photodynamic therapy for breast cancer, especially the latest developments in the application of metal nanocarriers in breast cancer PDT, and highlight some of the recent synergistic therapies, hopefully providing an accessible overview of the current knowledge that may act as a basis for new ideas or systematic evaluations of already promising results.
Collapse
Affiliation(s)
- Liang Shang
- Department of Breast Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (L.S.); (J.Z.); or (Y.S.)
| | - Xinglu Zhou
- Department of PET/CT Center, Harbin Medical University Cancer Hospital, Harbin 150081, China;
| | - Jiarui Zhang
- Department of Breast Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (L.S.); (J.Z.); or (Y.S.)
| | - Yujie Shi
- Department of Breast Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (L.S.); (J.Z.); or (Y.S.)
| | - Lei Zhong
- Department of Breast Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (L.S.); (J.Z.); or (Y.S.)
- Department of Breast Surgery, Sixth Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Correspondence:
| |
Collapse
|
13
|
Algorri JF, Ochoa M, Roldán-Varona P, Rodríguez-Cobo L, López-Higuera JM. Photodynamic Therapy: A Compendium of Latest Reviews. Cancers (Basel) 2021; 13:4447. [PMID: 34503255 PMCID: PMC8430498 DOI: 10.3390/cancers13174447] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising therapy against cancer. Even though it has been investigated for more than 100 years, scientific publications have grown exponentially in the last two decades. For this reason, we present a brief compendium of reviews of the last two decades classified under different topics, namely, overviews, reviews about specific cancers, and meta-analyses of photosensitisers, PDT mechanisms, dosimetry, and light sources. The key issues and main conclusions are summarized, including ways and means to improve therapy and outcomes. Due to the broad scope of this work and it being the first time that a compendium of the latest reviews has been performed for PDT, it may be of interest to a wide audience.
Collapse
Affiliation(s)
- José Francisco Algorri
- Photonics Engineering Group, University of Cantabria, 39005 Santander, Spain; (M.O.); (P.R.-V.); (J.M.L.-H.)
- CIBER-bbn, Institute of Health Carlos III, 28029 Madrid, Spain;
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Mario Ochoa
- Photonics Engineering Group, University of Cantabria, 39005 Santander, Spain; (M.O.); (P.R.-V.); (J.M.L.-H.)
- CIBER-bbn, Institute of Health Carlos III, 28029 Madrid, Spain;
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Pablo Roldán-Varona
- Photonics Engineering Group, University of Cantabria, 39005 Santander, Spain; (M.O.); (P.R.-V.); (J.M.L.-H.)
- CIBER-bbn, Institute of Health Carlos III, 28029 Madrid, Spain;
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain
| | | | - José Miguel López-Higuera
- Photonics Engineering Group, University of Cantabria, 39005 Santander, Spain; (M.O.); (P.R.-V.); (J.M.L.-H.)
- CIBER-bbn, Institute of Health Carlos III, 28029 Madrid, Spain;
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain
| |
Collapse
|
14
|
Keratin nanoparticles and photodynamic therapy enhance the anticancer stem cells activity of salinomycin. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111899. [DOI: 10.1016/j.msec.2021.111899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/23/2020] [Accepted: 01/17/2021] [Indexed: 12/20/2022]
|
15
|
Li WP, Yen CJ, Wu BS, Wong TW. Recent Advances in Photodynamic Therapy for Deep-Seated Tumors with the Aid of Nanomedicine. Biomedicines 2021; 9:69. [PMID: 33445690 PMCID: PMC7828119 DOI: 10.3390/biomedicines9010069] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
Photodynamic therapy (PDT) works through photoactivation of a specific photosensitizer (PS) in a tumor in the presence of oxygen. PDT is widely applied in oncology to treat various cancers as it has a minimally invasive procedure and high selectivity, does not interfere with other treatments, and can be repeated as needed. A large amount of reactive oxygen species (ROS) and singlet oxygen is generated in a cancer cell during PDT, which destroys the tumor effectively. However, the efficacy of PDT in treating a deep-seated tumor is limited due to three main reasons: Limited light penetration depth, low oxygen concentration in the hypoxic core, and poor PS accumulation inside a tumor. Thus, PDT treatments are only approved for superficial and thin tumors. With the advancement of nanotechnology, PDT to treat deep-seated or thick tumors is becoming a reachable goal. In this review, we provide an update on the strategies for improving PDT with nanomedicine using different sophisticated-design nanoparticles, including two-photon excitation, X-ray activation, targeting tumor cells with surface modification, alteration of tumor cell metabolism pathways, release of therapeutic gases, improvement of tumor hypoxia, and stimulation of host immunity. We focus on the difficult-to-treat pancreatic cancer as a model to demonstrate the influence of advanced nanomedicine in PDT. A bright future of PDT application in the treatment of deep-seated tumors is expected.
Collapse
Affiliation(s)
- Wei-Peng Li
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chia-Jui Yen
- Division of Hematology and Oncology, Department of Internal Medicine, Graduate Institute of Clinical Medicine, National Cheng Kung University Hospital, Tainan 704, Taiwan;
| | - Bo-Sheng Wu
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Tak-Wah Wong
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
16
|
Wang C, Ding S, Wang S, Shi Z, Pandey NK, Chudal L, Wang L, Zhang Z, Wen Y, Yao H, Lin L, Chen W, Xiong L. Endogenous tumor microenvironment-responsive multifunctional nanoplatforms for precision cancer theranostics. Coord Chem Rev 2021; 426:213529. [DOI: 10.1016/j.ccr.2020.213529] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
17
|
Prabhu P. Tumoral delivery of nanotherapeutics. HANDBOOK ON NANOBIOMATERIALS FOR THERAPEUTICS AND DIAGNOSTIC APPLICATIONS 2021:53-101. [DOI: 10.1016/b978-0-12-821013-0.00024-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
18
|
Zhang Q, Wu L, Liu S, Chen Q, Zeng L, Chen X, Zhang Q. Targeted nanobody complex enhanced photodynamic therapy for lung cancer by overcoming tumor microenvironment. Cancer Cell Int 2020; 20:570. [PMID: 33292202 PMCID: PMC7694906 DOI: 10.1186/s12935-020-01613-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023] Open
Abstract
Background To investigate the efficacy of a PLGA-based nanobody complex in photodynamic therapy (PDT) and NIR-II imaging in A549 tumor hypoxic model. Method IR1048-MZ was firstly synthesized by conjugating a nitro imidazole group to IR1048. IR1048-MZ and Cat were then encapsulated in PLGA-SH solution. Anti-EGFR-Nanobody was also expressed and purified, and finally Anti-EGFR-Nanobody@PLGA-IR1048MZ-Cat (Nb@IC-NPs) nanobody complex was obtained based on the formation of desulfide bond between PLGA-SH and Anti-EGFR-Nanobody. Size distribution and morphology were characterized by TEM and DLS. Spectrum of Nb@IC-NPs towards NTR was measured by UV and fluorescence, while the particle’s selective response was studied using fluorescence. The uptake of Nb@IC-NPs in A549 cells was observed by flow cytometry and CLSM. In the meantime, its’ catalytic ability that decomposes H2O2 both extra-and intra-cellular was observed by fluorescence and CLSM. In vitro photodynamic toxicity of Nb@IC-NPs was examined by MTT, Live/Dead Cell Staining, Flow Cytometry and Apoptosis Assay. Tumor-bearing model was constructed to observe a semi-quantitative fluorescent distribution and the possibility of NIR-II fluorescence/photoacoustic (PA) imaging. Effect of Nb@IC-NPs on enhancing A549 tumor hypoxia and expression profile of HIF-1α was investigated in the presence of NIR. An A549 tumor metastasis model was also constructed to confirm the complex’ potential to destroy primary tumor, inhibit lung metastasis, and prolong mice’ survival. Lastly, impact of Nb@IC-NPs on mice’ main organs and blood indices was observed. Results Nb@IC-NPs was successfully fabricated with good homogeneity. The fluorescent absorbance of Nb@IC-NPs showed a linear relationship with the concentration of NTR, and a higher concentration of NTR corresponded to a stronger photoacoustic signal. In addition, Nb@IC-NPs showed a stable selectivity toward NTR. Our results also suggested a high efficient uptake of Nb@IC-NPs in A549 cells, which was more efficient than IC-NPs and IR1048-MZ alone. In vitro assays confirmed the effects of Nb@IC-NPs on catalytic O2 generation even in hypoxic cells. The cell viability was upregulated with the nanocomplex at the absence of the laser, whereas it was dramatically declined with laser treatment that excited at 980 nm. Nb@IC-NPs achieved tumor hypoxia NIR-II/PA imaging through assisting A549 gathering. When NIR was applied, Nb@IC-NPs can significantly relieve A549 cellular/tumor hypoxia by generating more reactive oxygen species (ROS), which in turn helps lower the expression level of HIF-1α. In summary, Nb@IC-NPs based PDT can efficiently decimate A549 primary tumor, inhibit metastatic lung cancer, and prolong the lifespan of the mice under tolerable dosage. At last, in vivo toxicity tests of the nanocomplex showed its biosafety to the main organs and normal blood indices values. Conclusion Nb@IC-NPs improves tumor hypoxia through catalytic reaction and lowers the expression level of HIF-1α. It achieves tumor PA imaging through intensified NIR-II fluorescence signal that caused by response of the complex to the lesion’s nitroreductase (NTR). Nb@IC-NPs based PDT can efficiently kill A549 primary tumor, inhibit a lung metastasis, as well as prolong mice’ survival cycle.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Donghu District, Nanchang, 330006, People's Republic of China
| | - Lian Wu
- Department of Nephrology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Shaozheng Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Donghu District, Nanchang, 330006, People's Republic of China
| | - Qingjie Chen
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Donghu District, Nanchang, 330006, People's Republic of China
| | - Lingpeng Zeng
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Donghu District, Nanchang, 330006, People's Republic of China
| | - Xuezhong Chen
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Donghu District, Nanchang, 330006, People's Republic of China
| | - Qing Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Donghu District, Nanchang, 330006, People's Republic of China.
| |
Collapse
|
19
|
Gierlich P, Mata AI, Donohoe C, Brito RMM, Senge MO, Gomes-da-Silva LC. Ligand-Targeted Delivery of Photosensitizers for Cancer Treatment. Molecules 2020; 25:E5317. [PMID: 33202648 PMCID: PMC7698280 DOI: 10.3390/molecules25225317] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/26/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising cancer treatment which involves a photosensitizer (PS), light at a specific wavelength for PS activation and oxygen, which combine to elicit cell death. While the illumination required to activate a PS imparts a certain amount of selectivity to PDT treatments, poor tumor accumulation and cell internalization are still inherent properties of most intravenously administered PSs. As a result, common consequences of PDT include skin photosensitivity. To overcome the mentioned issues, PSs may be tailored to specifically target overexpressed biomarkers of tumors. This active targeting can be achieved by direct conjugation of the PS to a ligand with enhanced affinity for a target overexpressed on cancer cells and/or other cells of the tumor microenvironment. Alternatively, PSs may be incorporated into ligand-targeted nanocarriers, which may also encompass multi-functionalities, including diagnosis and therapy. In this review, we highlight the major advances in active targeting of PSs, either by means of ligand-derived bioconjugates or by exploiting ligand-targeting nanocarriers.
Collapse
Affiliation(s)
- Piotr Gierlich
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James’s Hospital, D08W9RT Dublin, Ireland;
| | - Ana I. Mata
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
| | - Claire Donohoe
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James’s Hospital, D08W9RT Dublin, Ireland;
| | - Rui M. M. Brito
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
- BSIM Therapeutics, Instituto Pedro Nunes, 3030-199 Coimbra, Portugal
| | - Mathias O. Senge
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James’s Hospital, D08W9RT Dublin, Ireland;
| | - Lígia C. Gomes-da-Silva
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
| |
Collapse
|
20
|
Simões JCS, Sarpaki S, Papadimitroulas P, Therrien B, Loudos G. Conjugated Photosensitizers for Imaging and PDT in Cancer Research. J Med Chem 2020; 63:14119-14150. [PMID: 32990442 DOI: 10.1021/acs.jmedchem.0c00047] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Early cancer detection and perfect understanding of the disease are imperative toward efficient treatments. It is straightforward that, for choosing a specific cancer treatment methodology, diagnostic agents undertake a critical role. Imaging is an extremely intriguing tool since it assumes a follow up to treatments to survey the accomplishment of the treatment and to recognize any conceivable repeating injuries. It also permits analysis of the disease, as well as to pursue treatment and monitor the possible changes that happen on the tumor. Likewise, it allows screening the adequacy of treatment and visualizing the state of the tumor. Additionally, when the treatment is finished, observing the patient is imperative to evaluate the treatment methodology and adjust the treatment if necessary. The goal of this review is to present an overview of conjugated photosensitizers for imaging and therapy.
Collapse
Affiliation(s)
- João C S Simões
- Institute of Chemistry, University of Neuchatel, Avenue de Bellevaux 51, CH-2000 Neuchatel, Switzerland.,BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| | - Sophia Sarpaki
- BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| | | | - Bruno Therrien
- Institute of Chemistry, University of Neuchatel, Avenue de Bellevaux 51, CH-2000 Neuchatel, Switzerland
| | - George Loudos
- BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| |
Collapse
|
21
|
Keerthiga R, Zhao Z, Pei D, Fu A. Photodynamic Nanophotosensitizers: Promising Materials for Tumor Theranostics. ACS Biomater Sci Eng 2020; 6:5474-5485. [PMID: 33320544 DOI: 10.1021/acsbiomaterials.0c01058] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Photodynamic theranostics/therapy (PDT) is a potential strategy for selectively imaging malignant sites and treating cancer via a non-invasive therapeutic method. Photosensitizers, the crucial components of PDT, enable colocalization of photons and light, and photon/light therapy in the therapeutic window of 400-900 nm exhibits photocytotoxicity to tumor cells. Due to their high biostability and photocytotoxicity, nanophotosensitizers (NPSs) are of much interest for malignant tumor theranostics at present. NPS-activated photons transfer energy through the absorption of a photon and convert molecular oxygen to the singlet reactive oxygen species, which leads to apoptosis and necrosis. Moreover, NPSs modified by polymers, including PLGA, PEG-PLA, PDLLA, PVCL-g-PLA, and P(VCL-co-VIM)-g-PLA, exhibit excellent biocompatibility, and a tumor-targeting molecule linked on the nanoparticle surface can precisely deliver NPSs into the tumor region. The development of NPSs will accelerate the progress in tumor theranostics through the photon/light pathway.
Collapse
Affiliation(s)
- Rajendiran Keerthiga
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zizhen Zhao
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Desheng Pei
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Ailing Fu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
22
|
Jiang L, Liu L, Lv F, Wang S, Ren X. Integration of Self‐Luminescence and Oxygen Self‐Supply: A Potential Photodynamic Therapy Strategy for Deep Tumor Treatment. Chempluschem 2020; 85:510-518. [DOI: 10.1002/cplu.202000083] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/02/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Linye Jiang
- Department of Environmental Science and EngineeringCollege of Resources and Environmental SciencesChina Agricultural University Beijing 100193 P. R. China
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Organic SolidsInstitute of ChemistryChinese Academy of Sciences Beijing 100190 P. R. China
| | - Libing Liu
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Organic SolidsInstitute of ChemistryChinese Academy of Sciences Beijing 100190 P. R. China
| | - Fengting Lv
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Organic SolidsInstitute of ChemistryChinese Academy of Sciences Beijing 100190 P. R. China
| | - Shu Wang
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Organic SolidsInstitute of ChemistryChinese Academy of Sciences Beijing 100190 P. R. China
| | - Xueqin Ren
- Department of Environmental Science and EngineeringCollege of Resources and Environmental SciencesChina Agricultural University Beijing 100193 P. R. China
- Beijing Key Laboratory of Farmland SoilPollution Prevention and RemediationChina Agricultural University Beijing 100193 P. R. China
| |
Collapse
|
23
|
Zhang SQ, Liu X, Sun QX, Johnson O, Yang T, Chen ML, Wang JH, Chen W. CuS@PDA-FA nanocomposites: a dual stimuli-responsive DOX delivery vehicle with ultrahigh loading level for synergistic photothermal-chemotherapies on breast cancer. J Mater Chem B 2020; 8:1396-1404. [PMID: 31971208 PMCID: PMC7390509 DOI: 10.1039/c9tb02440a] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In this study, CuS@PDA nanoparticles were synthesized and used to create a novel tumor-targeting nanocomposite platform composed of copper sulfide@polydopamine-folic acid/doxorubicin (CuS@PDA-FA/DOX) for performing both photothermal and chemotherapeutic cancer treatment. The nanocomposite platform has ultrahigh loading levels (4.2 ± 0.2 mg mg-1) and a greater photothermal conversion efficiency (η = 42.7%) than CuS/PDA alone. The uptake of CuS@PDA-FA/DOX nanocomposites is much higher in MCF-7 cells than in A549 cells because MCF-7 cells have much higher folic acid receptors than A549. Under near infrared (NIR) irradiation, the CuS@PDA-FA/DOX system using a synergistic combination of photothermal therapy and chemotherapy yields a better therapeutic effect than either photothermal therapy or chemotherapy alone. The treatment is very effective with the cell viability is only 5.6 ± 1.4%.
Collapse
Affiliation(s)
- Shang-Qing Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Pourhajibagher M, Plotino G, Chiniforush N, Bahador A. Dual wavelength irradiation antimicrobial photodynamic therapy using indocyanine green and metformin doped with nano-curcumin as an efficient adjunctive endodontic treatment modality. Photodiagnosis Photodyn Ther 2019; 29:101628. [PMID: 31870895 DOI: 10.1016/j.pdpdt.2019.101628] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 12/04/2019] [Accepted: 12/18/2019] [Indexed: 11/24/2022]
Abstract
BACKGROUND Indocyanine green (ICG) doped with nano-curcumin (N-CUR) can increase photosensitivity in antimicrobial photodynamic therapy (aPDT). Since metformin (Met) provides a synergistic advantage with photosensitivity, it was conjugated with N-CUR@ICG. Aim of the study was to evaluate the photosensitizing effect of N-CUR@ICG-Met used as a new photosensitizer in dual wavelengths irradiation (diode laser and light-emitting diode, LED) aPDT in root canals infected with Enterococcus faecalis biofilm. MATERIALS AND METHODS Following synthesis and confirmation of N-CUR@ICG-Met by Scanning electrone microscope (SEM), Fourier transform infrared (FT-IR) spectroscopy, X-ray diffraction (XRD) and Zetasizer analysis, the mature microbial biofilm was formed. The quantitative and qualitative evaluations of E. faecalis biofilm were made using microbial viability and SEM analysis of the following groups of treatment modalities (n = 5): 1- N-CUR, 2- ICG, 3- Met, 4- N-CUR@ICG, 5- N-CUR@ICG-Met, 6- Diode laser, 7- LED, 8- aPDT/diode, 9- aPDT/LED, 10- aPDT/diode laser + LED, 11- aPDT/LED + diode laser, 12- 2.5 % sodium hypochlorite (NaOCl). In addition, E. faecalis not treated served as negative control. Data were statistically analyzed using One-way Analysis of Variance (ANOVA) and the post-Hoc Bonferroni tests with a level of significance set at P < 0.05. RESULTS SEM, FT-IR, XRD, Zetasizer analysis confirmed successful doping of ICG-Met onto/into N-CUR. The treatment modality of N-CUR@ICG-Met mediated aPDT/diode laser, aPDT/LED, aPDT/diode laser + LED, and aPDT/LED + diode laser statistically decreased the cell viability of E. faecalis for 69.40 %, 75.52 %, 82.74 %, and 83.84 %, respectively compared with the negative control group (P < 0.05). The double wavelengths irradiation technique, which exposed the N-CUR@ICG-Met with diode laser irradiation and immediately after with LED (i.e. group No. 10), as well as, N-CUR@ICG-Met with LED irradiation and immediately after with diode laser (i.e. group No. 11) reduced the viable E. faecalis count in biofilm structures statistically more than the other treatment modalities (P < 0.05). CONCLUSIONS N-CUR@ICG-Met as a new photosensitizer in dual wavelengths irradiation method may improve anti-biofilm activity of aPDT against E. faecalis.
Collapse
Affiliation(s)
- Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Nasim Chiniforush
- Laser Research Center of Dentistry, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Bahador
- Oral Microbiology Laboratory, Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Wang C, Fan W, Zhang Z, Wen Y, Xiong L, Chen X. Advanced Nanotechnology Leading the Way to Multimodal Imaging-Guided Precision Surgical Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1904329. [PMID: 31538379 DOI: 10.1002/adma.201904329] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/18/2019] [Indexed: 06/10/2023]
Abstract
Surgical resection is the primary and most effective treatment for most patients with solid tumors. However, patients suffer from postoperative recurrence and metastasis. In the past years, emerging nanotechnology has led the way to minimally invasive, precision and intelligent oncological surgery after the rapid development of minimally invasive surgical technology. Advanced nanotechnology in the construction of nanomaterials (NMs) for precision imaging-guided surgery (IGS) as well as surgery-assisted synergistic therapy is summarized, thereby unlocking the advantages of nanotechnology in multimodal IGS-assisted precision synergistic cancer therapy. First, mechanisms and principles of NMs to surgical targets are briefly introduced. Multimodal imaging based on molecular imaging technologies provides a practical method to achieve intraoperative visualization with high resolution and deep tissue penetration. Moreover, multifunctional NMs synergize surgery with adjuvant therapy (e.g., chemotherapy, immunotherapy, phototherapy) to eliminate residual lesions. Finally, key issues in the development of ideal theranostic NMs associated with surgical applications and challenges of clinical transformation are discussed to push forward further development of NMs for multimodal IGS-assisted precision synergistic cancer therapy.
Collapse
Affiliation(s)
- Cong Wang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Wenpei Fan
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Zijian Zhang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yu Wen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Li Xiong
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
26
|
R Mokoena D, P George B, Abrahamse H. Enhancing Breast Cancer Treatment Using a Combination of Cannabidiol and Gold Nanoparticles for Photodynamic Therapy. Int J Mol Sci 2019; 20:E4771. [PMID: 31561450 PMCID: PMC6801525 DOI: 10.3390/ijms20194771] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022] Open
Abstract
Indisputably, cancer is a global crisis that requires immediate intervention. Despite the use of conventional treatments over the past decades, it is acceptable to admit that these are expensive, invasive, associated with many side effects and, therefore, a reduced quality of life. One of the most possible solutions to this could be the use of gold nanoparticle (AuNP) conjugated photodynamic therapy (PDT) in combination with cannabidiol (CBD), a Cannabis derivative from the Cannabis sativa. Since the use of Cannabis has always been associated with recreation and psychoactive qualities, the positive effects of Cannabis or its derivatives on cancer treatment have been misunderstood and hence misinterpreted. On the other hand, AuNP-PDT is the most favoured form of treatment for cancer, due to its augmented specificity and minimal risk of side effects compared to conventional treatments. However, its use requires the consideration of several physical, biologic, pharmacologic and immunological factors, which may hinder its effectiveness if not taken into consideration. In this review, the role of gold nanoparticle mediated PDT combined with CBD treatment on breast cancer cells will be deliberated.
Collapse
Affiliation(s)
- Dimakatso R Mokoena
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box: 17011, Johannesburg 2028, South Africa.
| | - Blassan P George
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box: 17011, Johannesburg 2028, South Africa.
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box: 17011, Johannesburg 2028, South Africa.
| |
Collapse
|
27
|
Wang D, Li X, Li X, Kang A, Sun L, Sun M, Yang F, Xu C. Magnetic And pH Dual-Responsive Nanoparticles For Synergistic Drug-Resistant Breast Cancer Chemo/Photodynamic Therapy. Int J Nanomedicine 2019; 14:7665-7679. [PMID: 31571870 PMCID: PMC6756767 DOI: 10.2147/ijn.s214377] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/02/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Drug resistance is one of the prime reasons of chemotherapy failure in breast cancer and is also an important factor affecting prognosis. PURPOSE In this study, we constructed a functional magnetic mesoporous silica-based nanocomposite (MMSN) for breast cancer chemotherapy/photodynamic therapy. METHODS MMSN was characterized by scanning electron microscopy and transmission electron microscopy to observe the morphology. The size distribution and zeta potential of the MSNs were determined using Malvern Particle Size Analyzer. Anti-tumor activity in vitro was investigated by CCK-8 assay, flow cytometry and transwell experiment, and the anti-tumor activity in vivo was probed into by magnetic targeting, toxicity, and antitumor effects in breast cancer-bearing BABL/c nude mice. RESULTS The results showed that the release of doxorubicin in the nanocomposites was pH sensitive, and the cumulative release rate reached 80.53% at 60 h under acidic conditions. The nanocomposites had a high cellular uptake ability in MCF-7/ADR cells, and the IC50 value of the nanocomposites on MCF-7/ADR cells was 4.23 μg/mL, much smaller than that of free DOX (363.2 μg/mL). The nanocomposites could effectively reverse resistance and induce apoptosis of MCF-7/ADR cells. The blood biochemistry parameters and H&E staining results showed no serious adverse effects after treatment with the nanocomposites. Prussian blue staining showed that the nanocomposites were able to target tumor tissues in tumor-bearing mice under a magnetic field. The combined chemical/photodynamic therapy significantly inhibited tumor growth in vivo. CONCLUSION Nanocomposites with magnetic and pH dual-responsive performance has shown a promising platform for enhanced drug-resistant breast cancer treatment.
Collapse
Affiliation(s)
- Dan Wang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, People’s Republic of China
- Department of Gynaecology and Obstetrics, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Xuefen Li
- Department of Nephrology, Jiulongpo People’s Hospital, Chongqing, People’s Republic of China
| | - Xinfang Li
- Inorganic Chemistry Department, School of Pharmacy, Second Military Medical University, Shanghai, People’s Republic of China
| | - Anfeng Kang
- Inorganic Chemistry Department, School of Pharmacy, Second Military Medical University, Shanghai, People’s Republic of China
| | - Linhong Sun
- Inorganic Chemistry Department, School of Pharmacy, Second Military Medical University, Shanghai, People’s Republic of China
| | - Miao Sun
- Inorganic Chemistry Department, School of Pharmacy, Second Military Medical University, Shanghai, People’s Republic of China
| | - Feng Yang
- Inorganic Chemistry Department, School of Pharmacy, Second Military Medical University, Shanghai, People’s Republic of China
| | - Congjian Xu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
28
|
Yu Y, Wang B, Guo C, Zhao F, Chen D. Protoporphyrin IX-loaded laminarin nanoparticles for anticancer treatment, their cellular behavior, ROS detection, and animal studies. NANOSCALE RESEARCH LETTERS 2019; 14:316. [PMID: 31535237 PMCID: PMC6751237 DOI: 10.1186/s11671-019-3138-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/26/2019] [Indexed: 05/06/2023]
Abstract
Laminarin conjugate-based nano-scaled particles were in this study proposed as a delivery system for protoporphyrin IX (Pp IX) in photodynamic therapy (PDT) of human breast cancer cells (MCF-7). Hematin-Laminarin-Dithiodipropionic Acid-MGK, named as HLDM, was an amphiphilic carrier material with dual pH/redox sensitive that could be used to load hydrophobic drug to improve their solubility and enhance biocompatibility. Therefore, we combined photosensitizer (Pp IX) with HLDM to fabricate a novel nano-micelles, herein called Pp IX-loaded HLDM micelles. The Pp IX-loaded HLDM micelles were 149.3 ± 35 nm sized in neutral water. Phototoxicity, in vitro PDT effect, and dual sensibility to pH and redox microenvironment of Pp IX-loaded HLDM micelles were examined at different concentrations by using MCF-7 human breast cancer cells. The experiments on phototoxicity and reactive oxygen species (ROS) production proved that the micelles could produce PDT to kill the cancer cells with a certain wavelength light. The apoptosis experiment indicated that the micelles could cause nuclear damage. In vivo PDT effect of the micelles was studied by constructing the tumor-bearing nude mouse model of MCF-7 cells. In vivo studies showed that the Pp IX-loaded HLDM micelles could induce remarkable anti-tumor effect. A promising laminarin-based nanomedicine platform acts as a new drug delivery system to enhance the uptake, accumulation, and PDT efficacy of Pp IX in vitro and in vivo.
Collapse
Affiliation(s)
- Yueming Yu
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Bingjie Wang
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Chunjing Guo
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Feng Zhao
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Daquan Chen
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, Yantai, 264005, China.
| |
Collapse
|
29
|
Namvar MA, Vahedi M, Abdolsamadi HR, Mirzaei A, Mohammadi Y, Azizi Jalilian F. Effect of photodynamic therapy by 810 and 940 nm diode laser on Herpes Simplex Virus 1: An in vitro study. Photodiagnosis Photodyn Ther 2018; 25:87-91. [PMID: 30447412 DOI: 10.1016/j.pdpdt.2018.11.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/07/2018] [Accepted: 11/13/2018] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Herpes simplex virus (HSV) is among the most common viruses in humans. HSV1 is often responsible for oral and perioral herpetic lesions. Photodynamic therapy (PDT) is a novel antimicrobial modality that involves the use of laser and a photosensitizer with a specific wavelength. This study aimed to assess and compare the effect of PDT with 810 and 940 nm diode laser and indocyanine green (ICG) photosensitizer on HSV1. METHODS In this in vitro study, HSV1 isolated from herpes labialis and there were 6 experimental groups.The irradiation parameters were the same for all groups. Number of remaining viruses per milliliter in each group was determined using real-time polymerase chain reaction (PCR) and statistically analyzed by ANOVA. RESULTS The virus count in all groups significantly decreased compared to the control group (P < 0.05) except in group ICG- without irradiation (P > 0.05). Comparison of groups 810- and 940- (use of each laser alone) with groups 810+ and 940+ (use of each laser plus ICG) revealed that reduction in virus count in groups 810+ and 940+ was significantly greater than that in groups 810- and 940-. CONCLUSION 810 nm diode laser irradiation and ICG causes the greatest reduction in number of HSV1 compared to all the other groups. ICG without laser irradiation has not significant efficacy on reduction of virus count.
Collapse
Affiliation(s)
- Mahsa Alavi Namvar
- Postgraduate Student, Department of Oral and maxillofacial Medicine, Faculty of Dentistry, Hamedan University of Medical Sciences, Hamedan, Iran.
| | - Mohammad Vahedi
- Associate Professor, Dental research center, Department of Oral and maxillofacial Medicine, Faculty of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Hamid-Reza Abdolsamadi
- Professor, Dental research center, Department of Oral and maxillofacial Medicine, Faculty of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Alireza Mirzaei
- DDS, MS, DMD, member of dental laser research of Hamadan university of medical science, Hamadan, Iran.
| | - Younes Mohammadi
- Ph.D. in Epidemiology, Modeling of Noncommunicable Diseases Research Center, Department of Epidemiology, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Farid Azizi Jalilian
- Associate Professor of Medical Virology, Department of Medical Microbiology, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
30
|
Lamch Ł, Pucek A, Kulbacka J, Chudy M, Jastrzębska E, Tokarska K, Bułka M, Brzózka Z, Wilk KA. Recent progress in the engineering of multifunctional colloidal nanoparticles for enhanced photodynamic therapy and bioimaging. Adv Colloid Interface Sci 2018; 261:62-81. [PMID: 30262128 DOI: 10.1016/j.cis.2018.09.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/13/2018] [Accepted: 09/15/2018] [Indexed: 12/12/2022]
Abstract
This up-to-date review summarizes the design and current fabrication strategies that have been employed in the area of mono- and multifunctional colloidal nanoparticles - nanocarriers well suited for photodynamic therapy (PDT) and diagnostic purposes. Rationally engineered photosensitizer (PS)-loaded nanoparticles may be achieved via either noncovalent (i.e., self-aggregation, interfacial deposition, interfacial polymerization, or core-shell entrapment along with physical adsorption) or covalent (chemical immobilization or conjugation) processes. These PS loading approaches should provide chemical and physical stability to PS payloads. Their hydrophilic surfaces, capable of appreciable surface interactions with biological systems, can be further modified using functional groups (stealth effect) to achieve prolonged circulation in the body after administration and/or grafted by targeting agents (such as ligands, which bind to specific receptors uniquely expressed on the cell surface) or stimuli (e.g., pH, temperature, and light)-responsive moieties to improve their action and targeting efficiency. These attempts may in principle permit efficacious PDT, combination therapies, molecular diagnosis, and - in the case of nanotheranostics - simultaneous monitoring and treatment. Nanophotosensitizers (nano-PSs) should possess appropriate morphologies, sizes, unimodal distributions and surface processes to be successfully delivered to the place of action after systemic administration and should be accumulated in certain tumors by passive and/or active targeting. Additionally, physically facilitating drug delivery systems emerge as a promising approach to enhancing drug delivery, especially for the non-invasive treatment of deep-seated malignant tissues. Recent advances in nano-PSs are scrutinized, with an emphasis on design principles, via the promising use of colloid chemistry and nanotechnology.
Collapse
Affiliation(s)
- Łukasz Lamch
- Department of Organic and Pharmaceutical Technology, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Agata Pucek
- Department of Organic and Pharmaceutical Technology, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy with Division of Laboratory Diagnostics, Medical University of Wrocław, Borowska 211A, 50-556 Wrocław, Poland
| | - Michał Chudy
- The Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Elżbieta Jastrzębska
- The Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Katarzyna Tokarska
- The Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Magdalena Bułka
- The Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Zbigniew Brzózka
- The Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Kazimiera A Wilk
- Department of Organic and Pharmaceutical Technology, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland.
| |
Collapse
|
31
|
Zhdanova KA, Ezhov AV, Bragina NA, Mironov AF. Synthesis of new binary porphyrin–cyanine conjugates and their self-aggregation in organic-aqueous media. MENDELEEV COMMUNICATIONS 2018. [DOI: 10.1016/j.mencom.2018.11.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
32
|
Fu Y, Li X, Ren Z, Mao C, Han G. Multifunctional Electrospun Nanofibers for Enhancing Localized Cancer Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1801183. [PMID: 29952070 PMCID: PMC6342678 DOI: 10.1002/smll.201801183] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/26/2018] [Indexed: 05/16/2023]
Abstract
Localized cancer treatment is one of the most effective strategies in clinical destruction of solid tumors at early stages as it can minimize the side effects of cancer therapeutics. Electrospun nanofibers have been demonstrated as a promising implantable platform in localized cancer treatment, enabling the on-site delivery of therapeutic components and minimizing side effects to normal tissues. This Review discusses the recent cutting-edge research with regard to electrospun nanofibers used for various therapeutic approaches, including gene therapy, chemotherapy, photodynamic therapy, thermal therapy, and combination therapy, in enhancing localized cancer treatment. Furthermore, it extensively analyzes the current challenges and potential breakthroughs in utilizing this novel platform for clinical transition in localized cancer treatment.
Collapse
Affiliation(s)
- Yike Fu
- State Key Laboratory of Silicon Materials, School of Materials
Science and Engineering, Zhejiang University, Hangzhou, 310027, P.R.
China
| | - Xiang Li
- State Key Laboratory of Silicon Materials, School of Materials
Science and Engineering, Zhejiang University, Hangzhou, 310027, P.R. China.,
| | - Zhaohui Ren
- State Key Laboratory of Silicon Materials, School of Materials
Science and Engineering, Zhejiang University, Hangzhou, 310027, P.R. China.,
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life
Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway,
Norman, Oklahoma, 73019-5300, USA.,
| | - Gaorong Han
- State Key Laboratory of Silicon Materials, School of Materials
Science and Engineering, Zhejiang University, Hangzhou, 310027, P.R.
China
| |
Collapse
|
33
|
Yi L, Zhang Y, Shi X, Du X, Wang X, Yu A, Zhai G. Recent progress of functionalised graphene oxide in cancer therapy. J Drug Target 2018; 27:125-144. [DOI: 10.1080/1061186x.2018.1474359] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Lingyun Yi
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Jinan, China
| | - Yanan Zhang
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Jinan, China
| | - Xiaoqun Shi
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Jinan, China
| | - Xiyou Du
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Jinan, China
| | - Xinyi Wang
- College of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Aihua Yu
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Jinan, China
| | - Guangxi Zhai
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Jinan, China
| |
Collapse
|
34
|
Hsu CC, Lin SL, Chang CA. Lanthanide-Doped Core-Shell-Shell Nanocomposite for Dual Photodynamic Therapy and Luminescence Imaging by a Single X-ray Excitation Source. ACS APPLIED MATERIALS & INTERFACES 2018; 10:7859-7870. [PMID: 29405703 DOI: 10.1021/acsami.8b00015] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Photodynamic therapy (PDT) could be highly selective and noninvasive, with low side effects as an adjuvant therapy for cancer treatment. Because excitation sources such as UV and visible lights for most of the photosensitizers do not penetrate deeply enough into biological tissues, PDT is useful only when the lesions are located within 10 mm below the skin. In addition, there is no prior example of theranostics capable of both PDT and imaging with a single deep-penetrating X-ray excitation source. Here we report a new theranostic scintillator nanoparticle (ScNP) composite in a core-shell-shell arrangement, that is, NaLuF4:Gd(35%),Eu(15%)@NaLuF4:Gd(40%)@NaLuF4:Gd(35%),Tb(15%), which is capable of being excited by a single X-ray radiation source to allow potentially deep tissue PDT and optical imaging with a low dark cytotoxicity and effective photocytotoxicity. With the X-ray excitation, the ScNPs can emit visible light at 543 nm (from Tb3+) to stimulate the loaded rose bengal (RB) photosensitizer and cause death of efficient MDA-MB-231 and MCF-7 cancer cells. The ScNPs can also emit light at 614 and 695 nm (from Eu3+) for luminescence imaging. The middle shell in the core-shell-shell ScNPs is unique to separate the Eu3+ in the core and the Tb3+ in the outer shell to prevent resonance quenching between them and to result in good PDT efficiency. Also, it was demonstrated that although the addition of a mesoporous SiO2 layer resulted in the transfer of 82.7% fluorescence resonance energy between Tb3+ and RB, the subsequent conversion of the energy from RB to generate 1O2 was hampered, although the loaded amount of the RB was almost twice that without the mSiO2 layer. A unique method to compare the wt % and mol % compositions calculated by using the morphological transmission electron microscope images and the inductively coupled plasma elemental analysis data of the core, core-shell, and core-shell-shell ScNPs is also introduced.
Collapse
|
35
|
de Paula LB, Primo FL, Tedesco AC. Nanomedicine associated with photodynamic therapy for glioblastoma treatment. Biophys Rev 2017; 9:761-773. [PMID: 28823025 DOI: 10.1007/s12551-017-0293-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 07/27/2017] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma, also known as glioblastoma multiforme (GBM), is the most recurrent and malignant astrocytic glioma found in adults. Biologically, GBMs are highly aggressive tumors that often show diffuse infiltration of the brain parenchyma, making complete surgical resection difficult. GBM is not curable with surgery alone because tumor cells typically invade the surrounding brain, rendering complete resection unsafe. Consequently, present-day therapy for malignant glioma remains a great challenge. The location of the invasive tumor cells presents several barriers to therapeutic delivery. The blood-brain barrier regulates the trafficking of molecules to and from the brain. While high-grade brain tumors contain some "leakiness" in their neovasculature, the mechanisms of GBM onset and progression remain largely unknown. Recent advances in the understanding of the signaling pathways that underlie GBM pathogenesis have led to the development of new therapeutic approaches targeting multiple oncogenic signaling aberrations associated with the GBM. Among these, drug delivery nanosystems have been produced to target therapeutic agents and improve their biodistribution and therapeutic index in the tumor. These systems mainly include polymer or lipid-based carriers such as liposomes, metal nanoparticles, polymeric nanospheres and nanocapsules, micelles, dendrimers, nanocrystals, and nanogold. Photodynamic therapy (PDT) is a promising treatment for a variety of oncological diseases. PDT is an efficient, simple, and versatile method that is based on a combination of a photosensitive drug and light (generally laser-diode or laser); these factors are separately relatively harmless but when used together in the presence of oxygen molecules, free radicals are produced that initiate a sequence of biological events, including phototoxicity, vascular damage, and immune responses. Photodynamic pathways activate a cascade of activities, including apoptotic and necrotic cell death in both the tumor and the neovasculature, leading to a permanent lesion and destruction of GBM cells that remain in the healthy tissue. Glioblastoma tumors differ at the molecular level. For example, gene amplification epidermal growth factor receptor and its receptor are more highly expressed in primary GBM than in secondary GBM. Despite these distinguishing features, both types of tumors (primary and secondary) arise as a result dysregulation of numerous intracellular signaling pathways and have standard features, such as increased cell proliferation, survival and resistance to apoptosis, and loss of adhesion and migration, and may show a high degree of invasiveness. PDT may promote significant tumor regression and extend the lifetime of patients who experience glioma progression.
Collapse
Affiliation(s)
- Leonardo B de Paula
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering - Photobiology and Photomedicine Research Group, Faculty of Philosophy, Science and Letters of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, 14040-901, São Paulo, Brazil
| | - Fernando L Primo
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, 14801-903, São Paulo, Brazil
| | - Antonio C Tedesco
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering - Photobiology and Photomedicine Research Group, Faculty of Philosophy, Science and Letters of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, 14040-901, São Paulo, Brazil.
| |
Collapse
|
36
|
Peng Y, He G, Tang D, Xiong L, Wen Y, Miao X, Hong Z, Yao H, Chen C, Yan S, Lu L, Yang Y, Li Q, Deng X. Lovastatin Inhibits Cancer Stem Cells and Sensitizes to Chemo- and Photodynamic Therapy in Nasopharyngeal Carcinoma. J Cancer 2017; 8:1655-1664. [PMID: 28775785 PMCID: PMC5535721 DOI: 10.7150/jca.19100] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 03/31/2017] [Indexed: 02/06/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus-associated malignancy occurring at high incidence in Southeast Asia and southern China. In spite of the good response to radio- and chemo-therapy at the early stage, resistance and recurrence develop in NPC patients in the advanced setting. Cancer stem cells (CSCs) play an important role in drug resistance and cancer recurrence. Here we report that lovastatin, a natural compound and a lipophilic statin that has already been used in the clinic to treat hypercholesterolemia, inhibited the CSC properties and induced apoptosis and cell cycle arrest in sphere-forming cells derived from the 5-8F and 6-10B NPC cell lines. Furthermore, lovastatin conferred enhanced sensitivity to the chemotherapeutic and photodynamic agents in NPC CSCs. Together our findings suggest that targeting CSCs by lovastatin in combination with routine chemotherapeutic drugs or photodynamic therapy might be a promising approach to the treatment of NPC.
Collapse
Affiliation(s)
- Yikun Peng
- Department of Otorhinolaryngology-Head and Neck Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Guangchun He
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Da Tang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Li Xiong
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yu Wen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiongying Miao
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Hui Yao
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Chao Chen
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Shichao Yan
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Lu Lu
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Yingke Yang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Qinglong Li
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiyun Deng
- Medical College, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
37
|
Morales P, Moreno L, Fernández-Ruiz J, Jagerovic N. Synthesis of a novel CB2 cannabinoid-porphyrin conjugate based on an antitumor chromenopyrazoledione. J PORPHYR PHTHALOCYA 2017. [DOI: 10.1142/s1088424617500092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
With the objective of developing an antitumor agent, the synthesis of a chromenopyrazoledione conjugated to a tetraphenylporphyrin is described. A complete conformational analysis of the novel porphyrin conjugate was performed using ab initio Hartree–Fock calculations at the 6-31G* level. The novel conjugate (14) shows stronger absorption intensity for both Soret and Q-bands than the free meso-tetraphenylporphyrin. It binds weakly but selectively to the cannabinoid receptor type-2. During the synthetic approach, a new tetraphenylporphyrin, 5-[4-(3,5-dioxomorpholino)phenyl]-10,15,20-triphenylporphyrin (10), has been characterized.
Collapse
Affiliation(s)
- Paula Morales
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIS), Unidad Asociada, l+D+i IQM/Universidad Rey Juan Carlos (URJC), Calle Juan de la Cierva 3, 28006 Madrid, Spain
| | - Laura Moreno
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIS), Unidad Asociada, l+D+i IQM/Universidad Rey Juan Carlos (URJC), Calle Juan de la Cierva 3, 28006 Madrid, Spain
| | - Javier Fernández-Ruiz
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Centro de Investigación Biomédica en Red de Enfermedades, Neurodegenerativas (CIBERNED), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Nadine Jagerovic
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIS), Unidad Asociada, l+D+i IQM/Universidad Rey Juan Carlos (URJC), Calle Juan de la Cierva 3, 28006 Madrid, Spain
| |
Collapse
|
38
|
Zhang N, Liu S, Wang N, Deng S, Song L, Wu Q, Liu L, Su W, Wei Y, Xie Y, Gong C. Biodegradable polymeric micelles encapsulated JK184 suppress tumor growth through inhibiting Hedgehog signaling pathway. NANOSCALE 2015; 7:2609-24. [PMID: 25581613 DOI: 10.1039/c4nr06300g] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
JK184 can specially inhibit Gli in the Hedgehog (Hh) pathway, which showed great promise for cancer therapeutics. For developing aqueous formulation and improving anti-tumor activity of JK184, we prepared JK184 encapsulated MPEG-PCL micelles by the solid dispersion method without using surfactants or toxic organic solvents. The cytotoxicity and cellular uptake of JK184 micelles were both increased compared with the free drug. JK184 micelles induced more apoptosis and blocked proliferation of Panc-1 and BxPC-3 tumor cells. In addition, JK184 micelles exerted a sustained in vitro release behavior and had a stronger inhibitory effect on proliferation, migration and invasion of HUVECs than free JK184. Furthermore, JK184 micelles had stronger tumor growth inhibiting effects in subcutaneous Panc-1 and BxPC-3 tumor models. Histological analysis showed that JK184 micelles improved anti-tumor activity by inducing more apoptosis, decreasing microvessel density and reducing expression of CD31, Ki67, and VEGF in tumor tissues. JK184 micelles showed a stronger inhibition of Gli expression in Hh signaling, which played an important role in pancreatic carcinoma. Furthermore, circulation time of JK184 in blood was prolonged after entrapment in polymeric micelles. Our results suggested that JK184 micelles are a promising drug candidate for treating pancreatic tumors with a highly inhibitory effect on Hh activity.
Collapse
Affiliation(s)
- Nannan Zhang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Hu J, Tang Y, Elmenoufy AH, Xu H, Cheng Z, Yang X. Nanocomposite-Based Photodynamic Therapy Strategies for Deep Tumor Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:5860-87. [PMID: 26398119 DOI: 10.1002/smll.201501923] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/03/2015] [Indexed: 05/22/2023]
Abstract
Photodynamic therapy (PDT), as an emerging clinically approved modality, has been used for treatment of various cancer diseases. Conventional PDT strategies are mainly focused on superficial lesions because the wavelength of illumination light of most clinically approved photosensitizers (PSs) is located in the UV/VIS range that possesses limited tissue penetration ability, leading to ineffective therapeutic response for deep-seated tumors. The combination of PDT and nanotechnology is becoming a promising approach to fight against deep tumors. Here, the rapid development of new PDT modalities based on various smartly designed nanocomposites integrating with conventionally used PSs for deep tumor treatments is introduced. Until now many types of multifunctional nanoparticles have been studied, and according to the source of excitation energy they can be classified into three major groups: near infrared (NIR) light excited nanomaterials, X-ray excited scintillating/afterglow nanoparticles, and internal light emission excited nanocarriers. The in vitro and in vivo applications of these newly developed PDT modalities are further summarized here, which highlights their potential use as promising nano-agents for deep tumor therapy.
Collapse
Affiliation(s)
- Jun Hu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Yong'an Tang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Ahmed H Elmenoufy
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Department of Pharmaceutical Chemistry, College of Pharmacy, Misr University for Science and Technology, Al-Motamayez District, 6th of October City, P.O. Box: 77, Egypt
| | - Huibi Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University Stanford, California, USA
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| |
Collapse
|
40
|
Tang Y, Hu J, Elmenoufy AH, Yang X. Highly Efficient FRET System Capable of Deep Photodynamic Therapy Established on X-ray Excited Mesoporous LaF3:Tb Scintillating Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2015; 7:12261-9. [PMID: 25974980 DOI: 10.1021/acsami.5b03067] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Photodynamic therapy (PDT) for deep-seated tumor is largely impeded by the limited penetration depth of excitation light in tissue. X-ray is considered as an ideal energy source to activate photosensitizers (PSs) located deep within the body with the assistance of scintillating nanoparticles (ScNPs). However, the efficacy under this concept is not satisfying due to the low scintillating luminescence and weak energy transfer from ScNPs to PSs. Here, mesoporous LaF3:Tb ScNPs were successfully synthesized by a facile hydrothermal process to act as PS carriers and X-ray energy transducers, owing to their good ionizing radiation stopping power and high luminescence efficiency. The formation mechanism of porous structure was elucidated in detail with classical crystallization theory. After a systematic investigation, LaF3:Tb ScNPs with optimized scintillating luminescence were obtained for loading Rose Bengal (RB) to establish an efficient FRET system, owing to their excellent spectral match. The FRET efficiency between ScNPs and RB was calculated to be as high as 85%. Under irradiation, enhanced (1)O2 generation induced by LaF3:Tb-RB nanocomposites via FRET process was detected. This LaF3:Tb-RB FRET system shows great potential to be applied in X-ray stimulated PDT for deep-seated tumors in the future.
Collapse
Affiliation(s)
- Yong'an Tang
- National Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Jun Hu
- National Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Ahmed H Elmenoufy
- National Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Xiangliang Yang
- National Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| |
Collapse
|